1
|
Nusrat F, Gorgov E, Khanna A, Isesele O, Bowne W, Lavu H, Yeo CJ, Jiang W, Jain A, Nevler A. Prognostic Properties of KRAS Gene Mutation Subtypes in Resected Pancreatic Ductal Adenocarcinoma. Pancreas 2025; 54:e449-e454. [PMID: 40314742 DOI: 10.1097/mpa.0000000000002458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 01/04/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) has a distinct genomic profile, with somatic KRAS mutations occurring in 85%-95% of all PDAC cases. This study aimed to measure the prognostic impact of specific KRAS mutations in resected PDAC patients from a large, high-volume center. METHODS This retrospective study included a cohort of PDAC patients who underwent curative-intent pancreatic resection at our institution between 2016 and 2021. Demographic, histologic, and oncologic outcome data were recorded. KRAS status was assessed via next-generation sequencing. Thirty-six (12.8%) wtKRAS, 109 (38.8%) G12D, 76 (27.0%) G12V, 36 (12.8%) G12R, 11 (3.9%) Q61H. RESULTS A total of 281 patients were included with wtKRAS (12.8%), G12D (38.8%), G12V (27.0%), G12R (12.8%), accounting for over 90% of the KRAS genotypes. Kaplan-Meier analysis revealed wild-type KRAS to be associated with improved overall survival (68.5±0 vs. 32.1±2.3 mo, P=0.005), and disease-free survival (35.4±0 vs. 20.3±3.9 mo, P=0.043). Cox regression analysis demonstrated worse overall survival with increased age (HR=1.04/y, P<0.01), neoadjuvant chemotherapy (HR=2.01, P<0.01), the presence of lymphovascular invasion (HR=2.47, P<0.01), G12D or G12V KRAS subtypes (P≤0.05), and lack of adjuvant chemotherapy (HR=0.6, P=0.02). CONCLUSIONS Next-generation sequencing of the KRAS subtype in resectable PDAC tumors shows that the KRAS G12D/G12V subtypes confer a worse prognosis compared with wild-type KRAS tumors.
Collapse
Affiliation(s)
- Faria Nusrat
- Sidney Kimmel Cancer Center, Department of Pathology, Thomas Jefferson University, Philadelphia, PA
| | - Eliyahu Gorgov
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Akshay Khanna
- Sidney Kimmel Cancer Center, Department of Pathology, Thomas Jefferson University, Philadelphia, PA
| | - Obehioye Isesele
- Sidney Kimmel Cancer Center, Department of Pathology, Thomas Jefferson University, Philadelphia, PA
| | - Wilbur Bowne
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Harish Lavu
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Charles J Yeo
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Wei Jiang
- Sidney Kimmel Cancer Center, Department of Pathology, Thomas Jefferson University, Philadelphia, PA
| | - Aditi Jain
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Avinoam Nevler
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
2
|
Keane F, O’Connor C, Moss D, Chou JF, Perry MA, Crowley F, Saxena P, Chan A, Schoenfeld JD, Singhal A, Park W, Cowzer D, Harrold E, Varghese AM, El Dika I, Crane C, Harding JJ, Abou-Alfa GK, Kingham TP, Wei AC, Yu KH, D’Angelica MI, Balachandran VP, Drebin J, Jarnagin WR, Bandlamudi C, Kelsen D, Capanu M, Soares KC, Balogun F, O’Reilly EM. Adjuvant modified FOLFIRINOX for resected pancreatic adenocarcinoma: clinical insights and genomic features from a large contemporary cohort. J Natl Cancer Inst 2025; 117:496-506. [PMID: 39460946 PMCID: PMC11884847 DOI: 10.1093/jnci/djae269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Adjuvant modified leucovorin calcium, fluorouracil, irinotecan hydrochloride, and oxaliplatin (FOLFIRINOX) is standard of care for fit individuals with resected pancreatic ductal adenocarcinoma (PDAC). Data are limited on adjuvant modified FOLFIRINOX outcomes outside clinical trials. METHODS We queried institutional databases to identify patients with resected PDAC who received 1 or more doses of adjuvant modified FOLFIRINOX. Primary endpoints were recurrence-free survival (RFS) and overall survival. Secondary endpoints were clinical factors and genomic features associated with outcomes. We estimated RFS and overall survival by using the Kaplan-Meier method. A Cox proportional hazards regression model was used to associate clinicogenomic features with survival outcomes. RESULTS A search revealed 147 individuals with PDAC between January 2015 and January 2023. Median patient age was 67 years, with 57 (39%) patients older than 70 years. Unfavorable prognostic features included 52 (36%) patients with N2 nodal status, 115 (78%) patients with lymphovascular invasion, and 133 (90%) patients with perineural invasion. Median time from surgery to initiation of modified FOLFIRINOX was 1.78 months (IQR = 1.45-2.12). In total, 124 (84%) patients completed 12 doses; 98 (67%) patients stopped oxaliplatin early because of neuropathy (median = 10 doses, range = 4-12 doses). Further dosing characteristics are summarized in Table S3, with a median follow-up of 35.1 months, a median RFS of 26 months (95% confidence interval [CI] = 19 to 39), and a median overall survival not reached. For the cohort older than 70 years of age, the median RFS was 23 months (95% CI = 14 to not reached) and the median overall survival was 51 months (95% CI = 37 to not reached). Modified FOLFIRINOX started sooner than 8 weeks from resection was associated with improved RFS (hazard ratio = 0.62, 95% CI = 0.41 to 0.96; P = .033) and overall survival (hazard ratio = 0.53, 95% CI = 0.3 to 0.94; P = .030). KRAS variation and whole-genome doubling trended to shorter RFS and overall survival. Homologous recombination deficiency status did not confer improved survival outcomes. CONCLUSIONS Adjuvant modified FOLFIRINOX was effective and tolerated in patients with resected PDAC in a nontrial setting, including for patients older than 70 years of age.
Collapse
Affiliation(s)
- Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Catherine O’Connor
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Harvard Medical School, Boston, MA, United States
| | - Drew Moss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Icahn School of Medicine at Mount Sinai, Mount Sinai Morningside-West, New York, NY, United States
| | - Joanne F Chou
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Maria A Perry
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Fionnuala Crowley
- Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Parima Saxena
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Amelia Chan
- Pharmacy Department, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Joshua D Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Anupriya Singhal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Darren Cowzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Emily Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Imane El Dika
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Christopher Crane
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- School of Medicine, Trinity College, Dublin, Ireland
| | - James J Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
- School of Medicine, Trinity College, Dublin, Ireland
| | - T Peter Kingham
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Alice C Wei
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Michael I D’Angelica
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Vinod P Balachandran
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jeffrey Drebin
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - William R Jarnagin
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Chaitanya Bandlamudi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - David Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | | | - Kevin C Soares
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- School of Medicine, Trinity College, Dublin, Ireland
| | - Fiyinfolu Balogun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Eileen M O’Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- David M. Rubenstein Center for Pancreas Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- School of Medicine, Trinity College, Dublin, Ireland
| |
Collapse
|
3
|
Raji S, Zaribafzadeh H, Jones T, Kanu E, Tong K, Fletcher A, Howell TC, McCall SJ, Marks JR, Rogers B, Niedzwiecki D, Allen PJ, Nussbaum DP, Kabiri Z. Prognostic Implications of Codon-Specific KRAS Mutations in Localized and Advanced Stages of Pancreatic Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.03.25321601. [PMID: 39974026 PMCID: PMC11838675 DOI: 10.1101/2025.02.03.25321601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Introduction While KRAS mutations represent the primary oncogenic driver in pancreatic ductal adenocarcinoma (PDAC), the association between codon-specific alterations and patient outcomes remains poorly elucidated, largely due to a lack of datasets coupling genomic profiling with rich clinical annotations across disease stages. Patients and Methods We utilized AACR's GENIE Biopharma Consortium Pancreas v1.2 dataset to test the associating of codon-specific KRAS mutations with clinicogenomic features and patient outcomes in PDAC patients diagnosed with localized (stages I-III) and advanced disease (stage IV). Overall survival was compared using Kaplan-Meier and multivariable Cox proportional hazards methods. Results Among 1,032 eligible patients, 949 (92%) exhibited mutant KRAS . These mutations were predominantly observed at G12D (n=390, 41%), G12V (n=305, 32%), and G12R (n=149, 16%). In the group of patients who presented with localized disease, those with G12V mutation had notably longer survival compared to G12D mutation ( P = 0.002). In contrast, patients with G12V mutation who presented with metastatic disease experienced shorter overall survival compared to those with G12R ( P = 0.005), and G12D mutations ( P = 0.009). Furthermore, no significant differences were observed in the frequencies of co-altered driver genes, including TP53 , CDKN2A , and SMAD4 , across the different KRAS mutations. Conclusions These findings demonstrated that codon-specific KRAS mutations impact PDAC outcomes differently based on disease stage at diagnosis. As studies testing KRAS inhibitors continue to emerge and mature, these data offer important contextual insights regarding survival outcomes associated with codon-specific KRAS mutations based on existing therapeutic approaches.
Collapse
|
4
|
Mouawad A, Habib S, Boutros M, Attieh F, Kourie HR. How to find a needle in a haystack: a systematic review on targeting KRAS wild-type pancreatic cancer. Future Oncol 2024; 20:3539-3547. [PMID: 38861291 PMCID: PMC11776852 DOI: 10.1080/14796694.2024.2355078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Aim: Pancreatic adenocarcinoma is a very aggressive type of cancer, in which targeted therapies have not yet been fully utilized. KRAS wild-type pancreatic adenocarcinoma tumors are associated with different genomic alterations in comparison to KRAS mutated pancreatic adenocarcinoma. Objective: This systematic review aims to provide a one-stop summary of all these alterations, their proposed targeted treatment and their effect on disease progression. Methods: An electronic search strategy was elaborated in the PubMed database between 2020 and January 2024. Results: 21 studies were included, and we found that the most frequent targetable genomic alterations in KRAS wild-type pancreatic adenocarcinoma were BRAF, EGFR, FGFR, MSI-H/dMMR, Her2/ERBB2 amplification, BRCA1/2 and other HRDs, and gene fusions like ALK, NTRK and NRG1.
Collapse
Affiliation(s)
- Antoine Mouawad
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Sofia Habib
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Marc Boutros
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Fouad Attieh
- Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| | - Hampig Raphaël Kourie
- Department of Hematology-Oncology, Université Saint-Joseph de Beyrouth, Beyrouth, 11-5076, Lebanon
| |
Collapse
|
5
|
Bravo AC, Morão B, Luz A, Dourado R, Oliveira B, Guedes A, Moreira-Barbosa C, Fidalgo C, Mascarenhas-Lemos L, Costa-Santos MP, Maio R, Paulino J, Viana Baptista P, Fernandes AR, Cravo M. Bringing Hope to Improve Treatment in Pancreatic Ductal Adenocarcinoma-A New Tool for Molecular Profiling of KRAS Mutations in Tumor and Plasma Samples. Cancers (Basel) 2024; 16:3544. [PMID: 39456638 PMCID: PMC11506488 DOI: 10.3390/cancers16203544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) incidence is rising, and prognosis remains poor due to late diagnosis and limited effective therapies. Currently, patients are treated based on TNM staging, without molecular tumor characterization. This study aimed to validate a technique that combines the amplification refractory mutation system (ARMS) with high-resolution melting analysis (HRMA) for detecting mutations in codon 12 of KRAS in tumor and plasma, and to assess its prognostic value. METHODS Prospective study including patients with newly diagnosed PDAC with tumor and plasma samples collected before treatment. Mutations in codon 12 of KRAS (G12D, G12V, G12C, and G12R) were detected using ARMS-HRMA and compared to Sanger sequencing (SS). Univariate and multivariate analyses were used to evaluate the prognostic significance of these mutations. RESULTS A total of 88 patients, 93% with ECOG-PS 0-1, 57% with resectable disease. ARMS-HRMA technique showed a higher sensitivity than SS, both in tumor and plasma (77% vs. 51%; 25 vs. 0%, respectively). The most frequent mutation was G12D (n = 32, 36%), followed by G12V (n = 22, 25%). On multivariate analysis, patients with G12D and/or G12C mutations, either in tumor or plasma, had lower PFS (HR 1.792, 95% CI 1.061-3.028, p = 0.029; HR 2.081, 95% CI 1.014-4.272, p = 0.046, respectively) and lower OS (HR 1.757, 95% CI 1.013-3.049, p = 0.045; HR 2.229, 95% CI 1.082-4.594, p = 0.030, respectively). CONCLUSIONS ARMS-HRMA is a rapid and cost-effective method for detecting KRAS mutations in PDAC patients, offering the potential for stratifying prognosis and guiding treatment decisions. The presence of G12D and G12C mutations in both tumor and plasma is associated with a poorer prognosis.
Collapse
Affiliation(s)
- Ana Catarina Bravo
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
| | - Bárbara Morão
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
| | - André Luz
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Rúben Dourado
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Beatriz Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Ana Guedes
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz Learning Health, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Catarina Moreira-Barbosa
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz Learning Health, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Catarina Fidalgo
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
| | - Luís Mascarenhas-Lemos
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
- Catolica Medical School, 1649-023 Lisboa, Portugal
| | | | - Rui Maio
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Jorge Paulino
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Pedro Viana Baptista
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Marília Cravo
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
6
|
Hálková T, Bunganič B, Traboulsi E, Minárik M, Zavoral M, Benešová L. Prognostic Role of Specific KRAS Mutations Detected in Aspiration and Liquid Biopsies from Patients with Pancreatic Cancer. Genes (Basel) 2024; 15:1302. [PMID: 39457426 PMCID: PMC11507146 DOI: 10.3390/genes15101302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Although the overall survival prognosis of patients in advanced stages of pancreatic ductal adenocarcinoma (PDAC) is poor, typically ranging from days to months from diagnosis, there are rare cases of patients remaining in therapy for longer periods of time. Early estimations of survival prognosis would allow rational decisions on complex therapy interventions, including radical surgery and robust systemic therapy regimens. Understandably, there is great interest in finding prognostic markers that can be used for patient stratification. We determined the role of various KRAS mutations in the prognosis of PDAC patients using biopsy samples and circulating tumor DNA. Methods: A total of 118 patients with PDAC, clinically confirmed by endoscopic ultrasound-guided fine-needle aspiration biopsy (EUS-FNB), were included in the study. DNA was extracted from cytological slides following a standard cytology evaluation to ensure adequacy (viability and quantity) and to mark the tumor cell fraction. Circulating tumor DNA (ctDNA) was extracted from plasma samples of 45 patients in stage IV of the disease. KRAS mutations in exons 12 and 13 were detected by denaturing capillary electrophoresis (DCE), revealing a minute presence of mutation-specific heteroduplexes. Kaplan-Meier survival curves were calculated for individual KRAS mutation types. Results:KRAS mutations were detected in 90% of tissue (106/118) and 44% of plasma (20/45) samples. All mutations were localized at exon 2, codon 12, with G12D (GGT > GAT) being the most frequent at 44% (47/106) and 65% (13/20), followed by other types including G12V (GGT > GTT) at 31% (33/106) and 10% (2/20), G12R (GGT > CGT) at 17% (18/106) and 10% (2/20), G12C (GGT/TGT) at 5% (5/106) and 0% (0/20) and G12S (GGT/AGT) at 1% (1/106) and 5% (1/20) in tissue and plasma samples, respectively. Two patients had two mutations simultaneously (G12V + G12S and G12D + G12S) in both types of samples (2%, 2/106 and 10%, 2/20 in tissue and plasma samples, respectively). The median survival of patients with the G12D mutation in tissues was less than half that of other patients (median survival 101 days, 95% CI: 80-600 vs. 228 days, 95% CI: 184-602), with a statistically significant overall difference in survival (p = 0.0080, log-rank test), and furthermore it was less than that of all combined patients with other mutation types (101 days, 95% CI: 80-600 vs. 210 days, 95% CI: 161-602, p = 0.0166). For plasma samples, the survival of patients with this mutation was six times shorter than that of patients without the G12D mutation (27 days, 95% CI: 8-334 vs. 161 days, 95% CI: 107-536, p = 0.0200). In contrast, patients with detected KRAS G12R in the tissue survived nearly twice as long as other patients in the aggregate (286 days, 95% CI: 70-602 vs. 162 days, 95% CI: 122-600, p = 0.0374) or patients with other KRAS mutations (286 days, 95% CI: 70-602 vs. 137 days, 95% CI: 107-600, p = 0.0257). Conclusions: Differentiation of specific KRAS mutations in EUS-FNB and ctDNA (above all, the crucial G12D and G12R) is feasible in routine management of PDAC patients and imperative for assessment of prognosis.
Collapse
Affiliation(s)
- Tereza Hálková
- Centre for Applied Genomics of Solid Tumors (CEGES), Genomac Research Institute, Drnovská 1112/60, 161 00 Prague, Czech Republic
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 8/2030, 128 00 Prague, Czech Republic
| | - Bohuš Bunganič
- Department of Medicine, First Faculty of Medicine, Charles University and Military University Hospital, U Vojenské Nemocnice 1200, 169 02 Prague, Czech Republic
| | - Eva Traboulsi
- Department of Pathology, Military University Hospital Prague, U Vojenské Nemocnice 1200, 169 02 Prague, Czech Republic
| | - Marek Minárik
- Department of Analytical Chemistry, Faculty of Science, Charles University, Hlavova 8/2030, 128 00 Prague, Czech Republic;
| | - Miroslav Zavoral
- Department of Medicine, First Faculty of Medicine, Charles University and Military University Hospital, U Vojenské Nemocnice 1200, 169 02 Prague, Czech Republic
| | - Lucie Benešová
- Centre for Applied Genomics of Solid Tumors (CEGES), Genomac Research Institute, Drnovská 1112/60, 161 00 Prague, Czech Republic
| |
Collapse
|
7
|
Wang Y, Dong S, Li H, Yang Y, Guo AL, Chao L. Nomogram for predicting live birth in ovulatory women undergoing frozen-thawed embryo transfer. BMC Pregnancy Childbirth 2024; 24:559. [PMID: 39192200 DOI: 10.1186/s12884-024-06759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Study objectives included the development of a practical nomogram for predicting live birth following frozen-thawed embryo transfers in ovulatory women. METHODS Totally, 2884 patients with regular menstrual cycles in our center were retrospectively enrolled. In an 8:2 ratio, we randomly assigned patients to training and validation cohorts. Then we identified risk factors by multivariate logistic regression and constructed nomogram. Finally, receiver operating characteristic curve analysis, calibration curve and decision curve analysis were performed to assess the calibration and discriminative ability of the nomogram. RESULTS We identified five variables which were related to live birth, including age, anti-Müllerian hormone (AMH), protocol of frozen-thawed embryo transfer (FET), stage of embryos and amount of high-quality embryos. We then constructed nomograms that predict the probabilities of live birth by using those five parameters. Receiver operating characteristic curve analysis (ROC) showed that the area under the curve (AUC) for live birth was 0.666 (95% CI: 0.644-0.688) in the training cohort. The AUC in the subsequent validation cohorts was 0.669 (95% CI, 0.625-0.713). The clinical practicability of this nomogram was demonstrated through calibration curve analysis and decision curve analysis. CONCLUSIONS Our nomogram provides a visual and simple tool in predicting live birth in ovulatory women who received FET. It could also provide advice and guidance for physicians and patients on decision-making during the FET procedure.
Collapse
Affiliation(s)
- Ying Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Shan Dong
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Hengfei Li
- School of Computer Science and Technology, Shandong Jianzhu University, Jinan, Shandong, 250101, P.R. China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - An-Liang Guo
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Lan Chao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China.
| |
Collapse
|
8
|
Casacuberta-Serra S, González-Larreategui Í, Capitán-Leo D, Soucek L. MYC and KRAS cooperation: from historical challenges to therapeutic opportunities in cancer. Signal Transduct Target Ther 2024; 9:205. [PMID: 39164274 PMCID: PMC11336233 DOI: 10.1038/s41392-024-01907-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 08/22/2024] Open
Abstract
RAS and MYC rank amongst the most commonly altered oncogenes in cancer, with RAS being the most frequently mutated and MYC the most amplified. The cooperative interplay between RAS and MYC constitutes a complex and multifaceted phenomenon, profoundly influencing tumor development. Together and individually, these two oncogenes regulate most, if not all, hallmarks of cancer, including cell death escape, replicative immortality, tumor-associated angiogenesis, cell invasion and metastasis, metabolic adaptation, and immune evasion. Due to their frequent alteration and role in tumorigenesis, MYC and RAS emerge as highly appealing targets in cancer therapy. However, due to their complex nature, both oncogenes have been long considered "undruggable" and, until recently, no drugs directly targeting them had reached the clinic. This review aims to shed light on their complex partnership, with special attention to their active collaboration in fostering an immunosuppressive milieu and driving immunotherapeutic resistance in cancer. Within this review, we also present an update on the different inhibitors targeting RAS and MYC currently undergoing clinical trials, along with their clinical outcomes and the different combination strategies being explored to overcome drug resistance. This recent clinical development suggests a paradigm shift in the long-standing belief of RAS and MYC "undruggability", hinting at a new era in their therapeutic targeting.
Collapse
Affiliation(s)
| | - Íñigo González-Larreategui
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Daniel Capitán-Leo
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain
| | - Laura Soucek
- Peptomyc S.L., Barcelona, Spain.
- Models of cancer therapies Laboratory, Vall d'Hebron Institute of Oncology, Cellex Centre, Hospital University Vall d'Hebron Campus, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
- Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
9
|
Nusrat F, Khanna A, Jain A, Jiang W, Lavu H, Yeo CJ, Bowne W, Nevler A. The Clinical Implications of KRAS Mutations and Variant Allele Frequencies in Pancreatic Ductal Adenocarcinoma. J Clin Med 2024; 13:2103. [PMID: 38610868 PMCID: PMC11012482 DOI: 10.3390/jcm13072103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
The KRAS proto-oncogene is a major driver of pancreatic tumorigenesis and is nearly ubiquitously mutated in pancreatic ductal adenocarcinoma (PDAC). KRAS point mutations are detected in over 90% of PDAC cases, and these mutations have been shown to be associated with worse therapy response and overall survival. Pathogenic KRAS mutations are mostly limited to codons 12, 13 and 61, with G12D, G12V, G12R, Q61H, and G13D accounting for approximately 95% of the mutant cases. Emerging data have shown the importance of specific mutant subtypes, as well as KRAS variant allele frequency on clinical prognosis. Furthermore, novel technologies and therapies are being developed to target specific mutant subtypes, with encouraging early results. In this paper, we aim to review the recent studies regarding the relative impact of specific mutant KRAS subtypes on oncologic outcomes, the application of variant allele frequency in next generation sequencing analyses, and the ongoing research into therapies targeting specific mutant KRAS subtypes.
Collapse
Affiliation(s)
- Faria Nusrat
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Akshay Khanna
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aditi Jain
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Wei Jiang
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Harish Lavu
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Charles J Yeo
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Wilbur Bowne
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Avinoam Nevler
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Jefferson Pancreas, Biliary and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
10
|
Khozooei S, Veerappan S, Toulany M. YB-1 activating cascades as potential targets in KRAS-mutated tumors. Strahlenther Onkol 2023; 199:1110-1127. [PMID: 37268766 DOI: 10.1007/s00066-023-02092-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/23/2023] [Indexed: 06/04/2023]
Abstract
Y‑box binding protein‑1 (YB-1) is a multifunctional protein that is highly expressed in human solid tumors of various entities. Several cellular processes, e.g. cell cycle progression, cancer stemness and DNA damage signaling that are involved in the response to chemoradiotherapy (CRT) are tightly governed by YB‑1. KRAS gene with about 30% mutations in all cancers, is considered the most commonly mutated oncogene in human cancers. Accumulating evidence indicates that oncogenic KRAS mediates CRT resistance. AKT and p90 ribosomal S6 kinase are downstream of KRAS and are the major kinases that stimulate YB‑1 phosphorylation. Thus, there is a close link between the KRAS mutation status and YB‑1 activity. In this review paper, we highlight the importance of the KRAS/YB‑1 cascade in the response of KRAS-mutated solid tumors to CRT. Likewise, the opportunities to interfere with this pathway to improve CRT outcome are discussed in light of the current literature.
Collapse
Affiliation(s)
- Shayan Khozooei
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Soundaram Veerappan
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Mahmoud Toulany
- Division of Radiobiology and Molecular Environmental Research, Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
11
|
Schepis T, De Lucia SS, Pellegrino A, del Gaudio A, Maresca R, Coppola G, Chiappetta MF, Gasbarrini A, Franceschi F, Candelli M, Nista EC. State-of-the-Art and Upcoming Innovations in Pancreatic Cancer Care: A Step Forward to Precision Medicine. Cancers (Basel) 2023; 15:3423. [PMID: 37444534 PMCID: PMC10341055 DOI: 10.3390/cancers15133423] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Pancreatic cancer remains a social and medical burden despite the tremendous advances that medicine has made in the last two decades. The incidence of pancreatic cancer is increasing, and it continues to be associated with high mortality and morbidity rates. The difficulty of early diagnosis (the lack of specific symptoms and biomarkers at early stages), the aggressiveness of the disease, and its resistance to systemic therapies are the main factors for the poor prognosis of pancreatic cancer. The only curative treatment for pancreatic cancer is surgery, but the vast majority of patients with pancreatic cancer have advanced disease at the time of diagnosis. Pancreatic surgery is among the most challenging surgical procedures, but recent improvements in surgical techniques, careful patient selection, and the availability of minimally invasive techniques (e.g., robotic surgery) have dramatically reduced the morbidity and mortality associated with pancreatic surgery. Patients who are not candidates for surgery may benefit from locoregional and systemic therapy. In some cases (e.g., patients for whom marginal resection is feasible), systemic therapy may be considered a bridge to surgery to allow downstaging of the cancer; in other cases (e.g., metastatic disease), systemic therapy is considered the standard approach with the goal of prolonging patient survival. The complexity of patients with pancreatic cancer requires a personalized and multidisciplinary approach to choose the best treatment for each clinical situation. The aim of this article is to provide a literature review of the available treatments for the different stages of pancreatic cancer.
Collapse
Affiliation(s)
- Tommaso Schepis
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Sara Sofia De Lucia
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Antonio Pellegrino
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Angelo del Gaudio
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Rossella Maresca
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Gaetano Coppola
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Michele Francesco Chiappetta
- Section of Gastroenterology and Hepatology, Promise, Policlinico Universitario Paolo Giaccone, 90127 Palermo, Italy;
- IBD-Unit, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Antonio Gasbarrini
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Anesthesiological and Reanimation Sciences, Fondazione Universitaria Policlinico Agostino Gemelli di Roma, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy; (F.F.); (M.C.)
| | - Marcello Candelli
- Department of Emergency Anesthesiological and Reanimation Sciences, Fondazione Universitaria Policlinico Agostino Gemelli di Roma, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy; (F.F.); (M.C.)
| | - Enrico Celestino Nista
- Center for Diagnosis and Treatment of Digestive Diseases, CEMAD, Gastroenterology Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (T.S.); (S.S.D.L.); (A.P.); (A.d.G.); (R.M.); (G.C.); (A.G.)
- Department of Translational Medicine and Surgery, School of Medicine, Catholic University of the Sacred Heart of Rome, 00168 Rome, Italy
| |
Collapse
|