1
|
Kokori E, Olatunji G, Ogieuhi IJ, Ajayi YI, Akinmoju O, Akinboade A, Irumudomon JG, Omoworare OT, Ezeano C, Adebayo YA, Oyewo O, Aderinto N. The emerging role of Sotorasib plus Panitumumab combination therapy in colorectal cancer treatment. Int J Clin Oncol 2025; 30:867-877. [PMID: 40080361 DOI: 10.1007/s10147-025-02736-y] [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: 07/26/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025]
Abstract
Colorectal cancer (CRC) poses a substantial global health challenge, ranking as the third most commonly diagnosed and second most fatal cancer worldwide. With an increasing incidence, particularly in older populations, CRC demands innovative therapeutic approaches to address the limitations of existing treatments. One critical target in CRC is the KRAS gene, which is frequently mutated and implicated in various cancer-related processes. This narrative review explores the promising role of Sotorasib plus Panitumumab combination therapy in CRC treatment. Combining Sotorasib with Panitumumab, an EGFR antagonist, offers a synergistic approach to comprehensively block KRAS and EGFR pathways, potentially overcoming resistance mechanisms observed in monotherapies. The review discusses the evolution of CRC treatment from traditional chemotherapy to the advent of targeted therapies like Bevacizumab and Cetuximab. It highlights the limitations of existing therapies, including resistance and toxicities, emphasising the urgency for innovative approaches. The CodeBreak clinical trials, specifically CodeBreak 101 and CodeBreak 300, provide a focal point for evaluating the efficacy of Sotorasib plus Panitumumab in patients with refractory KRAS G12C-mutated mCRC. Preliminary results demonstrate significant improvements in progression-free survival (PFS) and objective response rates, suggesting a paradigm shift in CRC treatment. The preliminary findings from the CodeBreak 300 trial signify a transformative impact of Sotorasib plus Panitumumab in refractory KRAS G12C-mutated mCRC. With a notable increase in PFS and objective response rates and a well-tolerated safety profile, this combination therapy emerges as a potential new standard of care. The results present an optimistic outlook for patients resistant to conventional therapies.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Yusuf Ismaila Ajayi
- Department of Medicine and Surgery, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Olumide Akinmoju
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeola Akinboade
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | | | - Chimezirim Ezeano
- Health Science Center, University of North Texas, Fort Worth, Texas, USA
| | | | | | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
2
|
Yildirim HC, Gunenc D, Almuradova E, Sutcuoglu O, Yalcin S. A Narrative Review of RAS Mutations in Early-Stage Colorectal Cancer: Mechanisms and Clinical Implications. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:408. [PMID: 40142219 PMCID: PMC11944112 DOI: 10.3390/medicina61030408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/28/2025]
Abstract
Colorectal cancer (CRC) is the third-most common cancer globally and a leading cause of cancer-related deaths. While the prognostic and predictive roles of RAS mutations in advanced CRC are well-established, their significance in early-stage CRC remains a topic of debate. Studies have been conducted for many years on clinical and pathological parameters that may be associated with RAS mutation, and there are inconsistent results in this regard. Currently, the only biomarker used in early-stage CRC is microsatellite status. KRAS mutations are detected in 40-50% of patients with colorectal cancer. RAS activating mutations cause loss of EGFR regulation by acting on the RAS/RAF/MAPK signaling pathways. In advanced colorectal cancer, these mechanisms cause a decrease in the effectiveness of EGFR inhibitors. However, studies on patients with early-stage colorectal cancer have inconsistent results. This review highlights the prognostic and clinical significance of KRAS mutations in early-stage CRC, particularly in MSS tumors. In the MSS group, KRAS mutations were associated with shorter TTR and OS compared to DWT patients. In contrast, in the MSI-H group, KRAS mutations showed no prognostic effect in TTR and OS. However. KRAS mutations were associated with shorter SAR in both MSI-H and MSS groups of patients. The findings underscore the need for routine molecular profiling, including KRAS and MSI status, to refine risk stratification and guide adjuvant therapy decisions. Further studies are warranted to explore targeted therapeutic approaches for KRAS-mutant CRC in the adjuvant setting.
Collapse
Affiliation(s)
- Hasan Cagri Yildirim
- Department of Medical Oncology, Nigde Training and Research Hospital, 51100 Nigde, Turkey
| | - Damla Gunenc
- Department of Medical Oncology, Ege University Faculty of Medicine, 35040 Izmir, Turkey;
| | | | - Osman Sutcuoglu
- Department of Medical Oncology, Etlik City Hospital, 06170 Ankara, Turkey;
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, 06230 Ankara, Turkey;
| |
Collapse
|
3
|
Ciracì P, Studiale V, Taravella A, Antoniotti C, Cremolini C. Late-line options for patients with metastatic colorectal cancer: a review and evidence-based algorithm. Nat Rev Clin Oncol 2025; 22:28-45. [PMID: 39558030 DOI: 10.1038/s41571-024-00965-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/20/2024]
Abstract
Over the past few years, several novel systemic treatments have emerged for patients with treatment-refractory metastatic colorectal cancer, thus making selection of the most effective later-line therapy a challenge for medical oncologists. Over the past decade, regorafenib and trifluridine-tipiracil were the only available drugs and often provided limited clinical benefit compared to best supportive care. Results from subsequent practice-changing trials opened several novel therapeutic avenues, both for unselected patients (such as trifluridine-tipiracil plus bevacizumab or fruquintinib) and for subgroups defined by the presence of actionable alterations in their tumours (such as HER2-targeted therapies or KRASG12C inhibitors) or with no acquired mechanisms of resistance to the previously received targeted agents in circulating tumour DNA (such as retreatment with anti-EGFR antibodies). In this Review, we provide a comprehensive overview of advances in the field over the past few years and offer a practical perspective on translation of the most relevant results into the daily management of patients with metastatic colorectal cancer using an evidence-based algorithm. Finally, we discuss some of the most appealing ongoing areas of research and highlight approaches with the potential to further expand the therapeutic armamentarium.
Collapse
Affiliation(s)
- Paolo Ciracì
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Vittorio Studiale
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ada Taravella
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Carlotta Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy.
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
4
|
Moretto R, Rossini D, Murgioni S, Ciracì P, Nasca V, Germani MM, Calegari MA, Vetere G, Intini R, Taravella A, Studiale V, Boccaccio C, Passardi A, Tamburini E, Zaniboni A, Salvatore L, Pietrantonio F, Lonardi S, Masi G, Cremolini C. KRASG12D-Mutated Metastatic Colorectal Cancer: Clinical, Molecular, Immunologic, and Prognostic Features of a New Emerging Targeted Alteration. JCO Precis Oncol 2024; 8:e2400329. [PMID: 39509668 DOI: 10.1200/po.24.00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/12/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
PURPOSE KRASG12D mutation (mut) occurs in about 10%-12% of metastatic colorectal cancer (mCRC). Recently, novel KRASG12D inhibitors have been developed and are currently under investigation in phase I/II clinical trials in solid tumors including mCRC. We aimed at performing a comprehensive characterization of clinical, molecular, immunologic, and prognostic features of KRASG12D-mutated mCRC to inform the design and the interpretation of future trials. METHODS We performed a pooled analysis of phase III TRIBE and TRIBE2 studies comparing 5-fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI)/bevacizumab (bev) to doublets (5-fluorouracil, leucovorin, and oxaliplatin or 5-fluorouracil, leucovorin, and irinotecan)/bev. RESULTS One hundred and thirty-six (16%) of 854 patients with available KRASG12D mutational status were KRASG12D mutated. KRASG12D-mutated patients had more frequently right-sided primary tumor and were less likely to present liver-only metastases with respect to other RAS mutated and all-wild-type (wt) patients. Compared with the BRAFV600E-mutated group, KRASG12D-mutated patients had more frequently left-sided primary tumor, resected primary tumor at the time of diagnosis, and Eastern Cooperative Oncology Group performance status 0. KRASG12D-mutated patients had better prognosis than BRAFV600E-mutated and worse prognosis than all wt patients. No prognostic difference was evident between KRASG12D mut and other RAS mut patients overall or according to other specific KRAS or NRAS hotspot mutations. No interaction effect was observed between KRASG12D mut and the benefit provided by FOLFOXIRI/bev compared with doublets/bev. PIK3CA mut were reported more frequently among KRASG12D-mutated tumors compared with both other RAS mut and all wt. CONCLUSION A detail estimation of KRASG12D mut mCRC patients' characteristics and expected outcomes may be useful when planning future studies in this subgroup. The high prevalence of PI3K/PTEN/Akt pathway activating alterations may affect the efficacy of targeted strategies.
Collapse
Affiliation(s)
- Roberto Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Daniele Rossini
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Sabina Murgioni
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Paolo Ciracì
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Vincenzo Nasca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maria Alessandra Calegari
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Guglielmo Vetere
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Rossana Intini
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Ada Taravella
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Vittorio Studiale
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Boccaccio
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, Meldola, Italy
| | - Emiliano Tamburini
- Department of Oncology and Palliative Care, Cardinale G Panico, Tricase City Hospital, Tricase, Italy
| | | | - Lisa Salvatore
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Gianluca Masi
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Ugai S, Yao Q, Takashima Y, Zhong Y, Matsuda K, Kawamura H, Imamura Y, Okadome K, Mima K, Arima K, Kosumi K, Song M, Meyerhardt JA, Giannakis M, Nowak JA, Ugai T, Ogino S. Clinicopathological, molecular, and prognostic features of colorectal carcinomas with KRAS c.34G>T (p.G12C) mutation. Cancer Sci 2024; 115:3455-3465. [PMID: 39039804 PMCID: PMC11448363 DOI: 10.1111/cas.16262] [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: 03/28/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/24/2024] Open
Abstract
Evidence indicates that combinations of anti-EGFR antibodies and KRAS p.G12C (c.34G>T) inhibitors can be an effective treatment strategy for advanced colorectal cancer. We hypothesized that KRAS c.34G>T (p.G12C)-mutated colorectal carcinoma might be a distinct tumor subtype. We utilized a prospective cohort incident tumor biobank (including 1347 colorectal carcinomas) and detected KRAS c.34G>T (p.G12C) mutation in 43 cases (3.2%) and other KRAS mutations (in codon 12, 13, 61, or 146) in 467 cases (35%). The CpG island methylator phenotype (CIMP)-low prevalence was similarly higher in KRAS c.34G>T mutants (52%) and other KRAS mutants (49%) than in KRAS-wild-type tumors (31%). KRAS c.34G>T mutants showed higher CIMP-high prevalence (14%) and lower CIMP-negative prevalence (33%) compared with other KRAS mutants (6% and 45%, respectively; p = 0.0036). Similar to other KRAS mutants, KRAS c.34G>T-mutated tumors were associated with cecal location, non-microsatellite instability (MSI)-high status, BRAF wild type, and PIK3CA mutation when compared with KRAS-wild-type tumors. Compared with BRAF-mutated tumors, KRAS c.34G>T mutants showed more frequent LINE-1 hypomethylation, a biomarker for early-onset colorectal carcinoma. KRAS c.34G>T mutants were not associated with other features, including the tumor tissue abundance of Fusobacterium nucleatum (F. animalis), pks+ Escherichia coli, Bifidobacterium, or (enterotoxigenic) Bacteroides fragilis. Among 1122 BRAF-wild-type colorectal carcinomas, compared with KRAS-wild-type tumors, multivariable-adjusted colorectal cancer-specific mortality hazard ratios (95% confidence interval) were 1.82 (1.05-3.17) in KRAS c.34G>T (p.G12C)-mutated tumors (p = 0.035) and 1.57 (1.22-2.02) in other KRAS-mutated tumors (p = 0.0004). Our study provides novel evidence for clinical and tumor characteristics of KRAS c.34G>T (p.G12C)-mutated colorectal carcinoma.
Collapse
Affiliation(s)
- Satoko Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Qian Yao
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yasutoshi Takashima
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer Institute and Harvard MedicalBostonMassachusettsUSA
| | - Yuxue Zhong
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Kosuke Matsuda
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Hidetaka Kawamura
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
- Department of SurgeryFukushima Medical UniversityFukushimaJapan
| | - Yu Imamura
- Department of Esophageal SurgeryThe Cancer Institute Hospital of the Japanese Foundation of Cancer ResearchTokyoJapan
| | - Kazuo Okadome
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kota Arima
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Mingyang Song
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of NutritionHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Clinical and Translational Epidemiology UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Division of GastroenterologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Jeffrey A. Meyerhardt
- Department of Medical OncologyDana‐Farber Cancer Institute and Harvard MedicalBostonMassachusettsUSA
| | - Marios Giannakis
- Department of Medical OncologyDana‐Farber Cancer Institute and Harvard MedicalBostonMassachusettsUSA
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Department of MedicineBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Jonathan A. Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer Institute and Harvard MedicalBostonMassachusettsUSA
| | - Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of PathologyBrigham and Women's Hospital, and Harvard Medical SchoolBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Cancer Immunology ProgramDana‐Farber/Harvard Cancer CenterBostonMassachusettsUSA
- Tokyo Medical and Dental University (Institute of Science Tokyo)TokyoJapan
| |
Collapse
|
6
|
Xiao A, Fakih M. KRAS G12C Inhibitors in the Treatment of Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:199-206. [PMID: 38825433 DOI: 10.1016/j.clcc.2024.05.004] [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: 04/15/2024] [Accepted: 05/11/2024] [Indexed: 06/04/2024]
Abstract
KRAS mutations contribute substantially to the overall colorectal cancer burden and have long been a focus of drug development efforts. After a lengthy preclinical road, KRAS inhibition via the G12C allele has finally become a therapeutic reality. Unlike in NSCLC, early studies of KRAS inhibitors in CRC struggled to demonstrate single agent activity. Investigation into these tissue-specific treatment differences has led to a deeper understanding of the complexities of MAPK signaling and the diverse adaptive feedback responses to KRAS inhibition. EGFR reactivation has emerged as a principal resistance mechanism to KRAS inhibitor monotherapy. Thus, the field has pivoted to dual EGFR/KRAS blockade with promising efficacy. Despite significant strides in the treatment of KRAS G12C mutated CRC, new challenges are on the horizon. Alternative RTK reactivation and countless acquired molecular resistance mechanisms have shifted the treatment goalpost. This review focuses on the historical and contemporary clinical strategies of targeting KRAS G12C alterations in CRC and highlights future directions to overcome treatment challenges.
Collapse
Affiliation(s)
- Annie Xiao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd. Duarte, CA
| | - Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Rd. Duarte, CA.
| |
Collapse
|
7
|
Martínez-Pérez J, Valladares-Ayerbes M. KRAS G12C inhibitors in metastatic colorectal cancer: a tale of two targets. Transl Gastroenterol Hepatol 2024; 9:54. [PMID: 39503027 PMCID: PMC11535810 DOI: 10.21037/tgh-24-48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/29/2024] [Indexed: 11/08/2024] Open
Affiliation(s)
- Julia Martínez-Pérez
- Medical Oncology Department, University Hospital Virgen del Rocio, Seville, Spain
- Institute of Biomedicine (IBIS, HUVR/University of Sevilla/CSIC), Seville, Spain
| | - Manuel Valladares-Ayerbes
- Medical Oncology Department, University Hospital Virgen del Rocio, Seville, Spain
- Institute of Biomedicine (IBIS, HUVR/University of Sevilla/CSIC), Seville, Spain
| |
Collapse
|
8
|
Osumi H, Shinozaki E, Nakamura Y, Esaki T, Yasui H, Taniguchi H, Satake H, Sunakawa Y, Komatsu Y, Kagawa Y, Denda T, Shiozawa M, Satoh T, Nishina T, Goto M, Takahashi N, Kato T, Bando H, Yamaguchi K, Yoshino T. Clinical features associated with NeoRAS wild-type metastatic colorectal cancer A SCRUM-Japan GOZILA substudy. Nat Commun 2024; 15:5885. [PMID: 39003289 PMCID: PMC11246505 DOI: 10.1038/s41467-024-50026-4] [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: 10/31/2023] [Accepted: 06/11/2024] [Indexed: 07/15/2024] Open
Abstract
"NeoRAS WT" refers to the loss of RAS mutations (MTs) following first-line treatment in metastatic colorectal cancer (mCRC). We evaluate the incidence and clinicopathological characteristics of NeoRAS WT mCRC using next-generation sequencing of plasma circulating tumor DNA. Patients with mCRC enrolled in the GOZILA study initially diagnosed with tissue RAS MT mCRC and received subsequent systemic therapy are eligible. NeoRAS WT is defined as the absence of detectable RAS MT in plasma and assessed in all eligible patients (Group A) and in a subgroup with at least one somatic alteration detected in plasma (Group B). Overall, 478 patients are included. NeoRAS WT prevalence is 19.0% (91/478) in Group A and 9.8% (42/429) in Group B. Absence of liver or lymph node metastasis and tissue RAS MTs other than KRAS exon 2 MTs are significantly associated with NeoRAS WT emergence. Overall, 1/6 and 2/6 patients with NeoRAS WT treated with anti-EGFR monoclonal antibodies (mAbs) show partial response and stable disease for ≥6 months, respectively. NeoRAS WT mCRC is observed at a meaningful prevalence, and anti-EGFR mAb-based therapy may be effective.
Collapse
Affiliation(s)
- Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kochi Medical School, Kochi, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshito Komatsu
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshinori Kagawa
- Department of Gastroenterological Surgery, Osaka General Medical Center, Osaka, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Manabu Shiozawa
- Department of Gastroenterological Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Taroh Satoh
- Palliative and Supportive Care Center, Osaka University Hospital, Suita, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Masahiro Goto
- Department of Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Osaka, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
9
|
Yaeger R, Uboha NV, Pelster MS, Bekaii-Saab TS, Barve M, Saltzman J, Sabari JK, Peguero JA, Paulson AS, Jänne PA, Cruz-Correa M, Anderes K, Velastegui K, Yan X, Der-Torossian H, Klempner SJ, Kopetz SE. Efficacy and Safety of Adagrasib plus Cetuximab in Patients with KRASG12C-Mutated Metastatic Colorectal Cancer. Cancer Discov 2024; 14:982-993. [PMID: 38587856 PMCID: PMC11152245 DOI: 10.1158/2159-8290.cd-24-0217] [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: 02/22/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
Adagrasib, an irreversible, selective KRASG12C inhibitor, may be an effective treatment in KRASG12C-mutated colorectal cancer, particularly when combined with an anti-EGFR antibody. In this analysis of the KRYSTAL-1 trial, patients with previously treated KRASG12C-mutated unresectable or metastatic colorectal cancer received adagrasib (600 mg twice daily) plus cetuximab. The primary endpoint was objective response rate (ORR) by blinded independent central review. Ninety-four patients received adagrasib plus cetuximab. With a median follow-up of 11.9 months, ORR was 34.0%, disease control rate was 85.1%, and median duration of response was 5.8 months (95% confidence interval [CI], 4.2-7.6). Median progression-free survival was 6.9 months (95% CI, 5.7-7.4) and median overall survival was 15.9 months (95% CI, 11.8-18.8). Treatment-related adverse events (TRAEs) occurred in all patients; grade 3-4 in 27.7% and no grade 5. No TRAEs led to adagrasib discontinuation. Exploratory analyses suggest circulating tumor DNA may identify features of response and acquired resistance. SIGNIFICANCE Adagrasib plus cetuximab demonstrates promising clinical activity and tolerable safety in heavily pretreated patients with unresectable or metastatic KRASG12C-mutated colorectal cancer. These data support a potential new standard of care and highlight the significance of testing and identification of KRASG12C mutations in patients with colorectal cancer. This article is featured in Selected Articles from This Issue, p. 897.
Collapse
Affiliation(s)
- Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nataliya V. Uboha
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin, Madison, Wisconsin
| | | | | | - Minal Barve
- Mary Crowley Cancer Research Center, Dallas, Texas
| | - Joel Saltzman
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Joshua K. Sabari
- Division of Medical Oncology, Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | | | - Andrew Scott Paulson
- Department of Medical Oncology, Texas Oncology – Baylor Charles A. Sammons Cancer Center, Dallas, Texas
| | - Pasi A. Jänne
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | | | - Kenna Anderes
- Mirati Therapeutics, a wholly owned subsidiary of Bristol Myers Squibb Company, San Diego, California
| | - Karen Velastegui
- Mirati Therapeutics, a wholly owned subsidiary of Bristol Myers Squibb Company, San Diego, California
| | - Xiaohong Yan
- Mirati Therapeutics, a wholly owned subsidiary of Bristol Myers Squibb Company, San Diego, California
| | - Hirak Der-Torossian
- Mirati Therapeutics, a wholly owned subsidiary of Bristol Myers Squibb Company, San Diego, California
| | - Samuel J. Klempner
- Division of Hematology-Oncology, Massachusetts General Cancer Center, Boston, Massachusetts
| | - Scott E. Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
10
|
Kavgaci G, Dizdar O, Yalcin S. Twelve-month progression-free survival with sotorasib and panitumumab in KRAS G12C mutant metastatic colorectal cancer. Anticancer Drugs 2024; 35:459-461. [PMID: 38451823 DOI: 10.1097/cad.0000000000001587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Colorectal cancer (CRC) ranks third in global cancer prevalence, with 40% presenting as metastatic colorectal cancer (mCRC). KRAS mutations in mCRC patients confer resistance to anti-EGFR treatments. Promising inhibitors such as sotorasib and adagrasib targeting KRASG12C mutations have demonstrated efficacy. Herein, we present a heavily pretreated mCRC case with a progression-free survival of 12 months with sotorasib and panitumumab. In 2017, a 27-year-old male presented with abdominal pain and received a diagnosis of stage IIIC KRAS G12C mutant CRC. Following surgery and adjuvant chemotherapy, he developed metastases in the liver and lungs in 2020. Treatment with FOLFIRINOX and bevacizumab, and later FOLFIRI and bevacizumab, with surgeries and local interventions resulted in partial responses. Upon disease progression, sotorasib and panitumumab were initiated, achieving a complete metabolic response. After 12 months of progression-free survival, oligoprogressive liver lesions were surgically resected. This case highlights the remarkable outcome of a heavily treated KRAS G12C mutant mCRC patient. The combination of sotorasib and panitumumab, along with multidisciplinary approaches including surgery and local interventions, played an important role in our patient's survival.
Collapse
Affiliation(s)
- Gozde Kavgaci
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | | | | |
Collapse
|
11
|
Contreras-Toledo D, Jiménez-Fonseca P, López CL, Montes AF, López Muñoz AM, Vázquez Rivera F, Alonso V, Alcaide J, Salvà F, Covela Rúa M, Guillot M, Martín Carnicero A, Jimeno Mate R, Cameselle García S, Asensio Martínez E, González Astorga B, Fernandez-Diaz AB, González Villaroel P, Virgili Manrique AC, Melián Sosa M, Alonso B, Cousillas Castiñeiras A, Castañón López C, Aparicio J, Carmona-Bayonas A. Dynamic nature of BRAF or KRAS p.G12C mutations in second-line therapy for advanced colorectal cancer patients: do early and late effects exist? Br J Cancer 2024; 130:777-787. [PMID: 38191609 PMCID: PMC10912758 DOI: 10.1038/s41416-023-02563-w] [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: 05/10/2023] [Revised: 12/08/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024] Open
Abstract
INTRODUCTION The mitogen-activated protein kinase (MAPK) signalling network aberrations in metastatic colorectal cancer (mCRC) generate intrinsic dynamic effects and temporal variations that are crucial but often overlooked in clinical trial populations. Here, we investigate the time-varying impact of MAPK pathway mutation genotype on each treatment line's contribution to the overall clinical course. METHODS The PROMETEO study focused on mCRC patients undergoing second-line treatment at 20 hospitals. We evaluated genotypes and employed flexible models to analyse the dynamic effect of each mutation. RESULTS We examined data derived from 1160 patients. The effects of KRAS G12C or G12V, and BRAF V600E are clearly time-varying, with unexpected consequences such as the deleterious effect of BRAF V600E vs other genotypes dissipating over time when subjects receive antiangiogenics, or KRAS G12V and G12C showing increasing aggressiveness over time. Thus, contrary to expectations, the 12-month survival rate from the second line for those who survived >6 months was 49.9% (95% CI, 32.7-67.3) for KRAS G12C and 59% (95% CI, 38.5-80.6) for BRAF V600E. CONCLUSIONS The dynamic perspective is essential for understanding the behaviour of tumours with specific genotypes, especially from the second line onward. This may be relevant in patient monitoring and treatment decision-making, particularly in cases with distinct mutations.
Collapse
Affiliation(s)
- Débora Contreras-Toledo
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Universidad de Oviedo, Oviedo, Spain.
| | - Paula Jiménez-Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Universidad de Oviedo, Oviedo, Spain
| | - Carlos López López
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Universidad de Cantabria (UNICAN), Santander, Spain
| | - Ana Fernández Montes
- Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | | | - Francisca Vázquez Rivera
- Department of Medical Oncology, Hospital Universitario de Santiago, Santiago de Compostela, Spain
| | - Vicente Alonso
- Department of Medical Oncology, Hospital Universitario Miguel Servet, IISA, Zaragoza, Spain
| | - Julia Alcaide
- Department of Medical Oncology, Hospital Costa del Sol, Marbella, Medical Oncology Intercenter Unit, Hospital Universitario Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Francesc Salvà
- Department of Medical Oncology, Hospital Universitario Vall D'Hebrón, Vall D´Hebrón Institute of Oncology (VHIO), Barcelona, Spain
| | - Marta Covela Rúa
- Department of Medical Oncology, Hospital Universitario Lucus Augusti, Lugo, Spain
| | - Mónica Guillot
- Department of Medical Oncology, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | | | - Raquel Jimeno Mate
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | | | | | | | | | | | | | - Marcos Melián Sosa
- Department of Medical Oncology, Instituto Valenciano de Oncología (IVO), Valencia, Spain
| | - Beatriz Alonso
- Department of Medical Oncology, Hospital Universitario de Canarias, Tenerife, Spain
| | | | | | - Jorge Aparicio
- Department of Medical Oncology, Hospital Universitario y Politécnico La Fe de Valencia, Valencia, Spain
| | - Alberto Carmona-Bayonas
- Department of Medical Oncology, Hospital Universitario Morales Meseguer, Universidad de Murcia, IMIB, Murcia, Spain.
| |
Collapse
|
12
|
Sunagua Aruquipa M, D'Alpino Peixoto R, Jacome A, Cesar F, Lorandi V, Dienstmann R. Association of KRAS G12C Status with Age at Onset of Metastatic Colorectal Cancer. Curr Issues Mol Biol 2024; 46:1374-1382. [PMID: 38392206 PMCID: PMC10887791 DOI: 10.3390/cimb46020088] [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: 12/13/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
The association of age at the onset of CRC and the prevalence of a KRAS G12C mutation is unclear. A retrospective, multicenter study evaluating metastatic CRC patients from January 2019 to July 2023, treated at the Oncoclinicas units and tested for tissue based KRAS/NRAS and BRAF mutations in a centralized genomics lab. A mismatch repair (MMR) status was retrieved from different labs and electronic medical records, as were patient demographics (age, gender) and tumor sidedness. The chi-square test was used to examine the association between clinical and molecular variables, with p value < 0.05 being statistically significant. A total of 858 cases were included. The median age was 63.7 years (range 22-95) and 17.4% were less than 50 years old at the diagnosis of metastatic CRC. Male patients represented 50.3% of the population. The sidedness distribution was as follows: left side 59.2%, right side 36.8% and not specified 4%. The prevalence of the KRAS mutation was 49.4% and the NRAS mutation was 3.9%. Among KRAS mutated tumors, the most common variants were G12V (27.6%) and G12D (23.5%), while KRAS G12C was less frequent (6.4%), which represented 3.1% of the overall population. The BRAF mutant cases were 7.3% and most commonly V600E. Only five (<1%) non-V600E mutations were detected. MSI-high or dMMR was present in 14 cases (1.6%). In the age-stratified analysis, left-sidedness (p < 0.001) and a KRAS G12C mutation (p = 0.046) were associated with a younger age (<50 years). In the sidedness-stratified analysis, a BRAF mutation (p = 0.001) and MSI-high/dMMR status (p = 0.009) were more common in right-sided tumors. Our data suggest that KRAS G12C mutations are more frequent in early-onset metastatic CRC. To the best of our knowledge, this is the largest cohort in the Latin American population with metastatic CRC reporting RAS, BRAF and MSI/MMR status.
Collapse
Affiliation(s)
| | | | - Alexandre Jacome
- Oncoclinicas Gastrointestinal Oncology Department, Belo Horizonte 34006-059, MG, Brazil
| | - Fernanda Cesar
- Oncoclinicas Gastrointestinal Oncology Department, Vitoria 29050-400, ES, Brazil
| | - Vinicius Lorandi
- Oncoclinicas Gastrointestinal Oncology Deparment, Porto Alegre 90610-001, RS, Brazil
| | | |
Collapse
|
13
|
Xu M, Zhao X, Wen T, Qu X. Unveiling the role of KRAS in tumor immune microenvironment. Biomed Pharmacother 2024; 171:116058. [PMID: 38171240 DOI: 10.1016/j.biopha.2023.116058] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/03/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Kirsten rats sarcoma viral oncogene (KRAS), the first discovered human oncogene, has long been recognized as "undruggable". KRAS mutations frequently occur in multiple human cancers including non-small cell lung cancer(NSCLC), colorectal cancer(CRC) and pancreatic ductal adenocarcinoma(PDAC), functioning as a "molecule switch" determining the activation of various oncogenic signaling pathways. Except for its intrinsic pro-tumorigenic role, KRAS alteration also exhibits an unique immune signature characterized by elevated PD-L1 level and high tumor mutational burden(TMB). KRAS mutation shape an immune suppressive microenvironment by impeding effective T cells infiltration and recruiting suppressive immune cells including myeloid-derived suppressor cells(MDSCs), regulatory T cells(Tregs), cancer associated fibroblasts(CAFs). In immune checkpoint inhibitor(ICI) era, NSCLC patients with mutated KRAS tend to be more responsive to ICI than patients with intact KRAS. The hallmark for KRAS mutation is the existence of multiple kinds of co-mutations. Different types of co-alterations have distinct tumor microenvironment(TME) signatures and responses to ICI. TP53 co-mutation possess a "hot" TME and achieve higher response to immunotherapy while other loss of function mutation correlated with a "colder" TME and a poor outcome to ICI-based therapy. The groundbreaking discovery of KRAS G12C inhibitors significantly improved outcomes for this KRAS subtype even though efficacy was limited to NSCLC patients. KRAS G12C inhibitors also restore the suppressive TME, creating an opportunity for combinations with ICI. However, an inevitable challenge to KRAS inhibitors is drug resistance. Promising combination strategies such as combination with SHP2 is an approach deserve further exploration because of their immune modulatory effect.
Collapse
Affiliation(s)
- Miao Xu
- Department of Medical Oncology, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Provinces, The First Hospital of China Medical University, Shenyang, Liaoning, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning, China
| | - Xing Zhao
- Department of Pediatrics, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Provinces, The First Hospital of China Medical University, Shenyang, Liaoning, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Provinces, The First Hospital of China Medical University, Shenyang, Liaoning, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning, China.
| |
Collapse
|
14
|
Underwood PW, Ruff SM, Pawlik TM. Update on Targeted Therapy and Immunotherapy for Metastatic Colorectal Cancer. Cells 2024; 13:245. [PMID: 38334637 PMCID: PMC10854977 DOI: 10.3390/cells13030245] [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: 12/27/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Metastatic colorectal cancer remains a deadly malignancy and is the third leading cause of cancer-related death. The mainstay of treatment for metastatic colorectal cancer is chemotherapy, but unfortunately, even with recent progress, overall survival is still poor. Colorectal cancer is a heterogeneous disease, and the underlying genetic differences among tumors can define the behavior and prognosis of the disease. Given the limitations of cytotoxic chemotherapy, research has focused on developing targeted therapy based on molecular subtyping. Since the early 2000s, multiple targeted therapies have demonstrated efficacy in treating metastatic colorectal cancer and have received FDA approval. The epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), and DNA mismatch repair pathways have demonstrated promising results for targeted therapies. As new gene mutations and proteins involved in the oncogenesis of metastatic colorectal cancer are identified, new targets will continue to emerge. We herein provide a summary of the updated literature regarding targeted therapies for patients with mCRC.
Collapse
Affiliation(s)
| | | | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Suite 670, Columbus, OH 43210, USA; (P.W.U.); (S.M.R.)
| |
Collapse
|
15
|
Kuboki Y, Fakih M, Strickler J, Yaeger R, Masuishi T, Kim EJ, Bestvina CM, Kopetz S, Falchook GS, Langer C, Krauss J, Puri S, Cardona P, Chan E, Varrieur T, Mukundan L, Anderson A, Tran Q, Hong DS. Sotorasib with panitumumab in chemotherapy-refractory KRAS G12C-mutated colorectal cancer: a phase 1b trial. Nat Med 2024; 30:265-270. [PMID: 38177853 PMCID: PMC11135132 DOI: 10.1038/s41591-023-02717-6] [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: 06/19/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024]
Abstract
The current third-line (and beyond) treatment options for RAS-mutant metastatic colorectal cancer have yielded limited efficacy. At the time of study start, the combination of sotorasib, a KRAS (Kirsten rat sarcoma viral oncogene homolog)-G12C inhibitor, and panitumumab, an epidermal growth factor receptor (EGFR) inhibitor, was hypothesized to overcome treatment-induced resistance. This phase 1b substudy of the CodeBreaK 101 master protocol evaluated sotorasib plus panitumumab in patients with chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. Here, we report the results in a dose-exploration cohort and a dose-expansion cohort. Patients received sotorasib (960 mg, once daily) plus panitumumab (6 mg kg-1, once every 2 weeks). The primary endpoints were safety and tolerability. Secondary endpoints included efficacy and pharmacokinetics. Exploratory biomarkers at baseline were assessed. Forty-eight patients (dose-exploration cohort, n = 8; dose-expansion cohort, n = 40) were treated. Treatment-related adverse events of any grade and grade ≥3 occurred in 45 (94%) and 13 (27%) patients, respectively. In the dose-expansion cohort, the confirmed objective response rate was 30.0% (95% confidence interval (CI) 16.6%, 46.5%). Median progression-free survival was 5.7 months (95% CI 4.2, 7.7 months). Median overall survival was 15.2 months (95% CI 12.5 months, not estimable). Prevalent genomic coalterations included APC (84%), TP53 (74%), SMAD4 (33%), PIK3CA (28%) and EGFR (26%). Sotorasib-panitumumab demonstrated acceptable safety with promising efficacy in chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. ClinicalTrials.gov identifier: NCT04185883 .
Collapse
Affiliation(s)
| | - Marwan Fakih
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Edward J Kim
- UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Corey Langer
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sonam Puri
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | | | | | | | | | | | - Qui Tran
- Amgen Inc., Thousand Oaks, CA, USA
| | - David S Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
16
|
Bhandari NR, Gilligan AM, Myers J, Ale-Ali A, Smolen L. Integrated budget impact model to estimate the impact of introducing selpercatinib as a tumor-agnostic treatment option for patients with RET-altered solid tumors in the US. J Med Econ 2024; 27:348-358. [PMID: 38334069 DOI: 10.1080/13696998.2024.2317120] [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: 10/31/2023] [Accepted: 02/07/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVE To estimate the potential budget impact on US third party payers (commercial or Medicare) associated with addition of selpercatinib as a tumor-agnostic treatment for patients with Rearranged during Transfection (RET)-altered solid tumors. METHODS An integrated budget impact model (iBIM) with 3-year (Y) time horizon was developed for 19 RET-altered tumors. It is referred to as an integrated model because it is a single model that integrated results across multiple tumor types (as opposed to tumor-specific models developed traditionally). The model estimated eligible patient populations and included tumor-specific comparator treatments for each tumor type. Estimated annual total costs (2022USD, $) included costs of drug, administration, supportive care, and toxicity. For a one-million-member plan, the number of patients with RET-altered tumors eligible for treatment, incremental total costs, and incremental per-member per-month (PMPM) costs associated with introduction of selpercatinib treatment were estimated. Uncertainty associated with model parameters was assessed using various sensitivity analyses. RESULTS Commercial perspective estimated 11.68 patients/million with RET-altered tumors as treatment-eligible annually, of which 7.59 (Y1), 8.17 (Y2), and 8.76 (Y3) patients would be selpercatinib-treated (based on forecasted market share). The associated incremental total and PMPM costs (commercial) were estimated to be: $873,099 and $0.073 (Y1), $2,160,525 and $0.180 (Y2), and $2,561,281 and $0.213 (Y3), respectively. The Medicare perspective estimated 55.82 patients/million with RET-altered tumors as treatment-eligible annually, of which 36.29 (Y1), 39.08 (Y2), and 41.87 (Y3) patients would be selpercatinib-treated. The associated incremental total and PMPM costs (Medicare) were estimated to be: $4,447,832 and $0.371 (Y1), $11,076,422 and $0.923 (Y2), and $12,637,458 and $1.053 (Y3), respectively. One-way sensitivity analyses across both perspectives identified drug costs, selpercatinib market share, incidence of RET, and treatment duration as significant drivers of incremental costs. CONCLUSIONS Three-year incremental PMPM cost estimates suggest a modest impact on payer-budgets associated with introduction of tumor-agnostic selpercatinib treatment.
Collapse
Affiliation(s)
| | | | - Julie Myers
- Medical Decision Modeling Inc, Indianapolis, IN, USA
| | | | - Lee Smolen
- Medical Decision Modeling Inc, Indianapolis, IN, USA
| |
Collapse
|
17
|
Fakih MG, Salvatore L, Esaki T, Modest DP, Lopez-Bravo DP, Taieb J, Karamouzis MV, Ruiz-Garcia E, Kim TW, Kuboki Y, Meriggi F, Cunningham D, Yeh KH, Chan E, Chao J, Saportas Y, Tran Q, Cremolini C, Pietrantonio F. Sotorasib plus Panitumumab in Refractory Colorectal Cancer with Mutated KRAS G12C. N Engl J Med 2023; 389:2125-2139. [PMID: 37870968 DOI: 10.1056/nejmoa2308795] [Citation(s) in RCA: 140] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
BACKGROUND KRAS G12C is a mutation that occurs in approximately 3 to 4% of patients with metastatic colorectal cancer. Monotherapy with KRAS G12C inhibitors has yielded only modest efficacy. Combining the KRAS G12C inhibitor sotorasib with panitumumab, an epidermal growth factor receptor (EGFR) inhibitor, may be an effective strategy. METHODS In this phase 3, multicenter, open-label, randomized trial, we assigned patients with chemorefractory metastatic colorectal cancer with mutated KRAS G12C who had not received previous treatment with a KRAS G12C inhibitor to receive sotorasib at a dose of 960 mg once daily plus panitumumab (53 patients), sotorasib at a dose of 240 mg once daily plus panitumumab (53 patients), or the investigator's choice of trifluridine-tipiracil or regorafenib (standard care; 54 patients). The primary end point was progression-free survival as assessed by blinded independent central review according to the Response Evaluation Criteria in Solid Tumors, version 1.1. Key secondary end points were overall survival and objective response. RESULTS After a median follow-up of 7.8 months (range, 0.1 to 13.9), the median progression-free survival was 5.6 months (95% confidence interval [CI], 4.2 to 6.3) and 3.9 months (95% CI, 3.7 to 5.8) in the 960-mg sotorasib-panitumumab and 240-mg sotorasib-panitumumab groups, respectively, as compared with 2.2 months (95% CI, 1.9 to 3.9) in the standard-care group. The hazard ratio for disease progression or death in the 960-mg sotorasib-panitumumab group as compared with the standard-care group was 0.49 (95% CI, 0.30 to 0.80; P = 0.006), and the hazard ratio in the 240-mg sotorasib-panitumumab group was 0.58 (95% CI, 0.36 to 0.93; P = 0.03). Overall survival data are maturing. The objective response was 26.4% (95% CI, 15.3 to 40.3), 5.7% (95% CI, 1.2 to 15.7), and 0% (95% CI, 0.0 to 6.6) in the 960-mg sotorasib-panitumumab, 240-mg sotorasib-panitumumab, and standard-care groups, respectively. Treatment-related adverse events of grade 3 or higher occurred in 35.8%, 30.2%, and 43.1% of patients, respectively. Skin-related toxic effects and hypomagnesemia were the most common adverse events observed with sotorasib-panitumumab. CONCLUSIONS In this phase 3 trial of a KRAS G12C inhibitor plus an EGFR inhibitor in patients with chemorefractory metastatic colorectal cancer, both doses of sotorasib in combination with panitumumab resulted in longer progression-free survival than standard treatment. Toxic effects were as expected for either agent alone and resulted in few discontinuations of treatment. (Funded by Amgen; CodeBreaK 300 ClinicalTrials.gov number, NCT05198934.).
Collapse
Affiliation(s)
- Marwan G Fakih
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Lisa Salvatore
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Taito Esaki
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Dominik P Modest
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - David P Lopez-Bravo
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Julien Taieb
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Michalis V Karamouzis
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Erika Ruiz-Garcia
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Tae-Won Kim
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Yasutoshi Kuboki
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Fausto Meriggi
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - David Cunningham
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Kun-Huei Yeh
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Emily Chan
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Joseph Chao
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Yaneth Saportas
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Qui Tran
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Chiara Cremolini
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| | - Filippo Pietrantonio
- From Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte (M.G.F.), and Amgen, Thousand Oaks (E.C., J.C., Y.S., Q.T.) - both in California; Oncologia Medica, Università Cattolica del Sacro Cuore, and Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome (L.S.), the Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia (F.M.), the Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa (C.C.), and the Medical Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori, Milan (F.P.) - all in Italy; the Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka (T.E.), and the Experimental Therapeutics and GI Oncology Department, National Cancer Center Hospital East, Kashiwa (Y.K.) - both in Japan; the Medicine Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin (D.P.M.); the Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona (D.P.L.-B.); Université Paris Cité, Site de Recherche Intégrée sur le Cancer, Cancer Research for Personalized Medicine Comprehensive Cancer Center, Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges-Pompidou, Paris (J.T.); the Department of Biological Chemistry, National and Kapodistrian University of Athens-School of Medicine, Athens (M.V.K.); Gastrointestinal Oncology Department and Translational Medicine Laboratory, Instituto Nacional de Cancerologia, Mexico City (E.R.-G.); the Oncology Department, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (T.W.K.); the Medicine Department, Royal Marsden Hospital, London (D.C.); and the Department of Oncology, National Taiwan University Hospital, and the Graduate Institute of Oncology, National Taiwan University College of Medicine - both in Taipei (K.H.Y.)
| |
Collapse
|
18
|
Strickler JH, Yoshino T, Stevinson K, Eichinger CS, Giannopoulou C, Rehn M, Modest DP. Prevalence of KRAS G12C Mutation and Co-mutations and Associated Clinical Outcomes in Patients With Colorectal Cancer: A Systematic Literature Review. Oncologist 2023; 28:e981-e994. [PMID: 37432264 PMCID: PMC10628573 DOI: 10.1093/oncolo/oyad138] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/20/2023] [Indexed: 07/12/2023] Open
Abstract
PURPOSE A systematic literature review was conducted to estimate the global prevalence of Kirsten rat sarcoma virus gene (KRAS) mutations, with an emphasis on the clinically significant KRAS G12C mutation, and to estimate the prognostic significance of these mutations in patients with colorectal cancer (CRC). DESIGN Relevant English-language publications in the Embase, MEDLINE, and the Cochrane Library databases (from 2009 to 2021) and congress presentations (from 2016 to 2021) were reviewed. Eligible studies were those that reported the prevalence and clinical outcomes of the KRAS G12C mutation in patients with CRC. RESULTS A total of 137 studies (interventional [n = 8], post hoc analyses of randomized clinical trials [n = 6], observational [n = 122], and longitudinal [n =1]) were reviewed. Sixty-eight studies reported the prevalence of KRAS mutations (KRASm) in 42 810 patients with CRC. The median global prevalence of KRASm was 38% (range, 13.3%-58.9%) and that of the KRAS G12C mutation (KRAS G12C) 3.1% (range, 0.7%-14%). Available evidence suggests that KRASm are possibly more common in tumors that develop on the right side of the colon. Limited evidence suggests a lower objective response rate and inferior disease-free/relapse-free survival in patients with KRAS G12C compared with patients with KRASwt or other KRASm. CONCLUSION Our analysis reveals that KRAS G12C is prevalent in 3% of patients with CRC. Available evidence suggests a poor prognosis for patients with KRAS G12C. Right-sided tumors were more likely to harbor KRASm; however, their role in determining clinical outcomes needs to be investigated further.
Collapse
Affiliation(s)
- John H Strickler
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kendall Stevinson
- Health Economics and Outcomes Researc, Amgen Inc., Thousand Oaks, CA, USA
| | | | | | - Marko Rehn
- Global Medical Affairs, Amgen Inc., Thousand Oaks, CA, USA
| | - Dominik Paul Modest
- Department for Hematology, Oncology and Cancer Immunology (CVK), Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
19
|
Taieb J, Sinicrope FA, Pederson L, Lonardi S, Alberts SR, George TJ, Yothers G, Van Cutsem E, Saltz L, Ogino S, Kerr R, Yoshino T, Goldberg RM, André T, Laurent-Puig P, Shi Q. Different prognostic values of KRAS exon 2 submutations and BRAF V600E mutation in microsatellite stable (MSS) and unstable (MSI) stage III colon cancer: an ACCENT/IDEA pooled analysis of seven trials. Ann Oncol 2023; 34:1025-1034. [PMID: 37619846 PMCID: PMC10938565 DOI: 10.1016/j.annonc.2023.08.006] [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: 06/27/2023] [Revised: 08/05/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The prognostic value of KRAS and BRAFV600E mutations in stage III colon cancer (CC) remains controversial and has never been clearly analyzed in patients with microsatellite instability-high (MSI-H) tumors due to sample size limitations. Data are also lacking for KRAS submutations and prognosis. PATIENTS AND METHODS We examined clinicopathological variables and prognosis in patients with surgically resected stage III CC who participated in seven clinical trials from the ACCENT/IDEA databases. Associations between KRAS exon 2 and BRAFV600E mutations and time to recurrence (TTR), overall survival (OS), and survival after recurrence (SAR) were assessed using a Cox model. We also analyzed the prognostic value of KRAS exon 2 submutations. RESULTS Among 8460 patients, 11.4% had MSI-H status. In the MSI-H group, BRAFV600E, KRAS exon 2 mutants, and double-wild-type statuses were detected in 40.6%, 18.1%, and 41.3%, respectively, whereas and in the microsatellite stable (MSS) group, these were detected in 7.7%, 38.6%, and 53.8%, respectively. In the MSS group, 5-year TTR rates of 61.8%, 66.3%, and 72.9% were observed among patients with BRAFV600E, KRAS exon 2 mutants, and those who were DWT, respectively [adjusted hazard ratio (HR) = 1.58 and 1.31, both P < 0.001]. In the MSI-H group, 5-year TTR rates did not differ significantly among the mutated subgroups. Similar results were found for OS. However, survival after relapse was significantly shorter in the KRAS exon 2- and BRAFV600E-mutated patients in both MSS (adjusted HR = 2.06 and 1.15; both P < 0.05) and MSI-H (adjusted HR = 1.99 and 1.81; both P < 0.05) groups. In the MSS group, KRAS exon 2 mutations were associated with TTR, but only p.G12C, p.G12D, and p.G13D were associated with poor outcomes after disease recurrence. CONCLUSIONS Testing for both KRAS and BRAFV600E mutations in stage III patients should be considered as they can better define individual patient prognosis, and may also enable patient selection for (neo)adjuvant trials dedicated to specific molecular subtypes with poor prognosis.
Collapse
Affiliation(s)
- J Taieb
- Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Université Paris Cité, AP-HP, SIRIC CARPEM, Paris, France; Department of Oncology, Mayo Clinic, Rochester, USA.
| | | | - L Pederson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, USA
| | - S Lonardi
- Department of Oncology, Veneto Institute of Oncology IRCCS, Padua, Italy
| | - S R Alberts
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - T J George
- Department of Oncology, University of Florida and the University of Florida Health Cancer Center, Gainesville, USA
| | - G Yothers
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, USA
| | - E Van Cutsem
- Department of Digestive Oncology, University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| | - L Saltz
- Department of Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - S Ogino
- Department of Pathology, Brigham & Women's Hospital, Boston, USA
| | - R Kerr
- Department of Oncology, Oxford University, Oxford, UK
| | - T Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - R M Goldberg
- Department of Oncology, West Virginia University Cancer Institute, Morgantown, USA; Mary Babb Randolph Cancer Center, Morgantown, USA
| | - T André
- Sorbonne Université, Department of Medical Oncology, Hôpital Saint-Antoine, Paris, France
| | - P Laurent-Puig
- Institut du cancer Paris CARPEM, AP-HP, Université Paris Cité, Paris, France; Hôpital Européen Georges Pompidou, Department of Tumor and Cancer Genomic Medicine, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris Cité, Team Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France
| | - Q Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, USA
| |
Collapse
|
20
|
Rasola C, Laurent-Puig P, André T, Falcoz A, Lepage C, Aparicio T, Bouché O, Lievre A, Mineur L, Bennouna J, Louvet C, Bachet JB, Borg C, Vernerey D, Lonardi S, Taieb J. Time to recurrence and its relation to survival after recurrence in patients resected for stage III colon cancer. Eur J Cancer 2023; 194:113321. [PMID: 37797388 DOI: 10.1016/j.ejca.2023.113321] [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: 06/25/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND It is intuitively thought that early relapse is associated with poor survival after recurrence (SAR) in resected colon cancer (CC) patients, but this has never been formally studied. METHODS We pooled data from stage III patients treated with oxaliplatin-based adjuvant therapy in two phase III trials, to analyse time to recurrence (TTR) and its relationship with SAR. TTR and SAR were also studied according to molecular status (mismatch repair (MMR), RAS, and BRAFV600E). Early relapsing patients were defined as patients having a TTR event within 12 months after starting adjuvant chemotherapy. RESULTS 4548 stage III CC patients were included in the present analysis. Deficient MMR (dMMR) CC patients experienced fewer recurrences than proficient (p)MMR CC patients (18.8% versus 27.6%) but had a significantly shorter median TTR (mTTR; 0.74 versus 1.40 years, p < 0.0001). In pMMR patients, BRAF and RAS mutations were also associated with earlier mTTR as compared to double wild-type (WT) patients (0.99 versus 1.38 versus 1.54 years, respectively, p < 0.0001). Early recurrence occurred in 397 patients and was associated with a median SAR (2.2 versus 3.3 years, p = 0.0007). However, this association was mainly due to pMMR/RAS and BRAFV600E mutated tumours and was not confirmed in dMMR and pMMR/double WT subgroups. CONCLUSION In resected stage III CC treated with standard oxaliplatin-based adjuvant therapy, TTR varies between dMMR, pMMR/RAS, or BRAFV600E mutated and pMMR/double WT tumours. In addition, early relapse is associated with poor survival, mainly due to patients resected for a pMMR/RAS or BRAFV600E mutated tumour.
Collapse
Affiliation(s)
- Cosimo Rasola
- Department of Gastroenterology and GI Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Université Paris-Cité, Paris, France; Department of Oncology, Veneto Institute of Oncology IRCCS, Padua, Italy; Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris Cité, team Personalized Medicine, Phamacogenomics and Therapeutic Optimization, Paris, France; Institut du Cancer Paris CARPEM, AP-HP,Centre Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Thierry André
- Sorbonne Université and Medical Oncology Department, Hôpital Saint-Antoine, Paris, France
| | - Antoine Falcoz
- University Hospital of Besançon, Methodology and Quality of Life Unit in Oncology, Besançon, France; INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Come Lepage
- Gastroenterology and Digestive Oncology, Hôpital Universitaire Le Bocage, Dijon, France
| | - Thomas Aparicio
- Université Paris-Cité, Gastroenterology Department, Hôpital Saint Louis, APHP, Paris, France
| | | | - Astrid Lievre
- Digestive Unit, Hôpital Universitaire de Pontchaillou, Rennes, France
| | - Laurent Mineur
- Oncology Department, Clinique Sainte-Catherine, Avignon, France
| | - Jaafar Bennouna
- Department of Medical Oncology, Hôpital Foch, Suresnes, France
| | - Christophe Louvet
- Department of Medical Oncology, Institute Mutualiste Montsouris, Paris, France
| | - Jean Baptiste Bachet
- Sorbonne University, Hepatogastroenterology and Digestive Oncology Department, Pitié Salpêtrière hospital, APHP, Paris, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, France
| | - Dewi Vernerey
- University Hospital of Besançon, Methodology and Quality of Life Unit in Oncology, Besançon, France; INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IRCCS, Padua, Italy; Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Julien Taieb
- Department of Gastroenterology and GI Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Université Paris-Cité, Paris, France.
| |
Collapse
|
21
|
Qunaj L, May MS, Neugut AI, Herzberg BO. Prognostic and therapeutic impact of the KRAS G12C mutation in colorectal cancer. Front Oncol 2023; 13:1252516. [PMID: 37790760 PMCID: PMC10543081 DOI: 10.3389/fonc.2023.1252516] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
KRAS G12C mutations are critical in the pathogenesis of multiple cancer types, including non-small cell lung (NSCLC), pancreatic ductal adenocarcinoma (PDAC), and colorectal (CRC) cancers. As such, they have increasingly become a target of novel therapies in the management of these malignancies. However, the therapeutic success of KRAS G12C inhibitors to date has been far more limited in CRC and PDAC than NSCLC. In this review, we briefly summarize the biochemistry of KRAS targeting and treatment resistance, highlight differences in the epidemiology of various G12C-mutated cancers, and provide an overview of the published data on KRAS G12C inhibitors for various indications. We conclude with a summary of ongoing clinical trials in G12C-mutant CRC and a discussion of future directions in the management of this disease. KRAS G12C mutation, targeted therapies, colorectal cancer, non-small cell lung cancer, pancreatic cancer, drug development.
Collapse
Affiliation(s)
- Lindor Qunaj
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York, NY, United States
| | - Michael S. May
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York, NY, United States
| | - Alfred I. Neugut
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, United States
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Benjamin O. Herzberg
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
22
|
Ottaiano A, Sabbatino F, Perri F, Cascella M, Sirica R, Patrone R, Capuozzo M, Savarese G, Ianniello M, Petrillo N, Circelli L, Granata V, Berretta M, Santorsola M, Nasti G. KRAS p.G12C Mutation in Metastatic Colorectal Cancer: Prognostic Implications and Advancements in Targeted Therapies. Cancers (Basel) 2023; 15:3579. [PMID: 37509241 PMCID: PMC10377118 DOI: 10.3390/cancers15143579] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
KRAS is frequently mutated in tumors. It is mutated in approximately 30% of all cancer cases and in nearly 50% of cases of metastatic colorectal cancer (CRC), which is the third leading cause of cancer-related deaths worldwide. Recent advancements in understanding CRC biology and genetics have highlighted the significance of KRAS mutations in the progression of CRC. The KRAS gene encodes a small GTPase (Guanosine TriPhosphatases) that plays a key role in signaling pathways associated with important proteins involved in amplifying growth factor and receptor signals. Mutations in KRAS are frequently observed in codons 12 and 13, and these mutations have oncogenic properties. Abnormal activation of KRAS proteins strongly stimulates signals associated with various cancer-related processes in CRC, including cell proliferation, migration and neoangiogenesis. In this review, we explore the distinct prognostic implications of KRAS mutations. Specifically, the KRAS p.G12C mutation is associated with a worse prognosis in metastatic CRC. The correlation between structure, conformation and mutations is visually presented to emphasize how alterations in individual amino acids at the same position in a single protein can unexpectedly exhibit complex involvement in cancer. Last, KRAS p.G12C is discussed as an emerging and promising therapeutic target in metastatic CRC, providing a concise overview of available clinical data regarding the use of new inhibitors.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, 84081 Salerno, Italy;
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (R.S.); (G.S.); (M.I.); (N.P.); (L.C.)
| | - Renato Patrone
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| | | | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (R.S.); (G.S.); (M.I.); (N.P.); (L.C.)
| | - Monica Ianniello
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (R.S.); (G.S.); (M.I.); (N.P.); (L.C.)
| | - Nadia Petrillo
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (R.S.); (G.S.); (M.I.); (N.P.); (L.C.)
| | - Luisa Circelli
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (R.S.); (G.S.); (M.I.); (N.P.); (L.C.)
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.P.); (M.C.); (R.P.); (V.G.); (M.S.); (G.N.)
| |
Collapse
|
23
|
|
24
|
Ciardiello D, Maiorano BA, Martinelli E. Targeting KRAS G12C in colorectal cancer: the beginning of a new era. ESMO Open 2023; 8:100745. [PMID: 36549128 PMCID: PMC9800313 DOI: 10.1016/j.esmoop.2022.100745] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 12/24/2022] Open
Abstract
RAS mutation is considered one of the most relevant oncogenic drivers in human cancers. Unfortunately, for more than three decades, RAS has been considered an undruggable target. Recently, the discovery of selective and potent KRASG12C inhibitors represented a light at the end of the tunnel. Indeed, sotorasib and adagrasib proved clinical activity in patients with refractory metastatic colorectal cancer harboring KRASG12C mutation; however, responses are lower than expected, suggesting the presence of intrinsic resistance. Consequently, novel combinatory strategies to disrupt the RAS signaling pathways are under clinical investigation. This review aims to discuss the current knowledge and novel routes of KRASG12C inhibition in metastatic colorectal cancer.
Collapse
Affiliation(s)
- D Ciardiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo; Medical Oncology Unit, Department of Precision Medicine, 'Luigi Vanvitelli' University of Campania, Naples.
| | - B A Maiorano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo; Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - E Martinelli
- Medical Oncology Unit, Department of Precision Medicine, 'Luigi Vanvitelli' University of Campania, Naples
| |
Collapse
|
25
|
Yaeger R, Weiss J, Pelster MS, Spira AI, Barve M, Ou SHI, Leal TA, Bekaii-Saab TS, Paweletz CP, Heavey GA, Christensen JG, Velastegui K, Kheoh T, Der-Torossian H, Klempner SJ. Adagrasib with or without Cetuximab in Colorectal Cancer with Mutated KRAS G12C. N Engl J Med 2023; 388:44-54. [PMID: 36546659 PMCID: PMC9908297 DOI: 10.1056/nejmoa2212419] [Citation(s) in RCA: 256] [Impact Index Per Article: 128.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Adagrasib, an oral small-molecule inhibitor of mutant KRAS G12C protein, has shown clinical activity in pretreated patients with several tumor types, including colorectal cancer. Preclinical studies suggest that combining a KRAS G12C inhibitor with an epidermal growth factor receptor antibody could be an effective clinical strategy. METHODS In this phase 1-2, open-label, nonrandomized clinical trial, we assigned heavily pretreated patients with metastatic colorectal cancer with mutant KRAS G12C to receive adagrasib monotherapy (600 mg orally twice daily) or adagrasib (at the same dose) in combination with intravenous cetuximab once a week (with an initial loading dose of 400 mg per square meter of body-surface area, followed by a dose of 250 mg per square meter) or every 2 weeks (with a dose of 500 mg per square meter). The primary end points were objective response (complete or partial response) and safety. RESULTS As of June 16, 2022, a total of 44 patients had received adagrasib, and 32 had received combination therapy with adagrasib and cetuximab, with a median follow-up of 20.1 months and 17.5 months, respectively. In the monotherapy group (43 evaluable patients), a response was reported in 19% of the patients (95% confidence interval [CI], 8 to 33). The median response duration was 4.3 months (95% CI, 2.3 to 8.3), and the median progression-free survival was 5.6 months (95% CI, 4.1 to 8.3). In the combination-therapy group (28 evaluable patients), the response was 46% (95% CI, 28 to 66). The median response duration was 7.6 months (95% CI, 5.7 to not estimable), and the median progression-free survival was 6.9 months (95% CI, 5.4 to 8.1). The percentage of grade 3 or 4 treatment-related adverse events was 34% in the monotherapy group and 16% in the combination-therapy group. No grade 5 adverse events were observed. CONCLUSIONS Adagrasib had antitumor activity in heavily pretreated patients with metastatic colorectal cancer with mutant KRAS G12C, both as oral monotherapy and in combination with cetuximab. The median response duration was more than 6 months in the combination-therapy group. Reversible adverse events were common in the two groups. (Funded by Mirati Therapeutics; KRYSTAL-1 ClinicalTrials.gov number, NCT03785249.).
Collapse
Affiliation(s)
- Rona Yaeger
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Jared Weiss
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Meredith S Pelster
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Alexander I Spira
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Minal Barve
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Sai-Hong I Ou
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Ticiana A Leal
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Tanios S Bekaii-Saab
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Cloud P Paweletz
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Grace A Heavey
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - James G Christensen
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Karen Velastegui
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Thian Kheoh
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Hirak Der-Torossian
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| | - Samuel J Klempner
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York (R.Y.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill (J.W.); Sarah Cannon Research Institute, Tennessee Oncology, Nashville (M.S.P.); Virginia Cancer Specialists, NEXT Oncology-Virginia, Fairfax (A.I.S.); US Oncology Research, the Woodlands (A.I.S.), and Mary Crowley Cancer Research, Dallas (M.B.) - both in Texas; the University of California Irvine School of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.), and Mirati Therapeutics, San Diego (J.G.C., K.V., T.K., H.D.-T.) - all in California; the Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta (T.A.L.); Medical Oncology, Mayo Clinic, Phoenix, Arizona (T.S.B.-S.); Belfer Center for Applied Cancer Science and the Department of Medical Oncology, Dana-Farber Cancer Institute (C.P.P., G.A.H.), and the Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital (S.J.K.) - both in Boston
| |
Collapse
|
26
|
Doleschal B, Petzer A, Rumpold H. Current concepts of anti-EGFR targeting in metastatic colorectal cancer. Front Oncol 2022; 12:1048166. [PMID: 36465407 PMCID: PMC9714621 DOI: 10.3389/fonc.2022.1048166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/26/2022] [Indexed: 11/07/2023] Open
Abstract
Anti-EGFR targeting is one of the key strategies in the treatment of metastatic colorectal cancer (mCRC). For almost two decades oncologists have struggled to implement EGFR antibodies in the mCRC continuum of care. Both sidedness and RAS mutational status rank high among the predictive factors for the clinical efficacy of EGFR inhibitors. A prospective phase III trial has recently confirmed that anti-EGFR targeting confers an overall survival benefit only in left sided RAS-wildtype tumors when given in first line. It is a matter of discussion if more clinical benefit can be reached by considering putative primary resistance mechanisms (e.g., HER2, BRAF, PIK3CA, etc.) at this early stage of treatment. The value of this procedure in daily routine clinical utility has not yet been clearly delineated. Re-exposure to EGFR antibodies becomes increasingly crucial in the disease journey of mCRC. Yet re- induction or re-challenge strategies have been problematic as they relied on mathematical models that described the timely decay of EGFR antibody resistant clones. The advent of liquid biopsy and the implementation of more accurate next-generation sequencing (NGS) based high throughput methods allows for tracing of EGFR resistant clones in real time. These displays the spatiotemporal heterogeneity of metastatic disease compared to the former standard radiographic assessment and re-biopsy. These techniques may move EGFR inhibition in mCRC into the area of precision medicine in order to apply EGFR antibodies with the increase or decrease of EGFR resistant clones. This review critically discusses established concepts of tackling the EGFR pathway in mCRC and provides insight into the growing field of liquid biopsy guided personalized approaches of EGFR inhibition in mCRC.
Collapse
Affiliation(s)
- Bernhard Doleschal
- Department of Internal Medicine I for Hematology With Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Austria
| | - Andreas Petzer
- Department of Internal Medicine I for Hematology With Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz, Linz, Austria
| | - Holger Rumpold
- Gastrointestinal Cancer Center, Ordensklinikum Linz, Linz, Austria
- Johannes Kepler University Linz, Medical Faculty, Linz, Austria
| |
Collapse
|
27
|
Ciardiello D, Chiarazzo C, Famiglietti V, Damato A, Pinto C, Zampino MG, Castellano G, Gervaso L, Zaniboni A, Oneda E, Rapisardi S, Bordonaro R, Zichi C, De Vita F, Di Maio M, Parisi A, Giampieri R, Berardi R, Lavacchi D, Antonuzzo L, Tamburini E, Maiorano BA, Parrella P, Latiano TP, Normanno N, De Stefano A, Avallone A, Martini G, Napolitano S, Troiani T, Martinelli E, Ciardiello F, De Vita F, Maiello E. Clinical efficacy of sequential treatments in KRASG12C-mutant metastatic colorectal cancer: findings from a real-life multicenter Italian study (CRC-KR GOIM). ESMO Open 2022; 7:100567. [PMID: 35994791 PMCID: PMC9588891 DOI: 10.1016/j.esmoop.2022.100567] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The presence of KRASG12C mutation in metastatic colorectal cancer (mCRC) correlates with poor outcome. Although different selective inhibitors are under clinical development, the optimal treatment remains uncertain. Thus, we conducted a retrospective analysis in a large cohort of patients with KRASG12C mCRC treated in 12 Italian oncology units. PATIENTS AND METHODS Patients with unresectable mCRC harboring KRASG12C mutation receiving a first-line chemotherapy doublet or triplet between 2011 and 2021 were included in the study. Evaluation of overall response rate (ORR), progression-free survival (PFS) and overall survival (OS) analysis was carried out. RESULTS A total of 256/6952 (3.7%) patients with mCRC displayed KRASG12C mutation; of these, 111 met the inclusion criteria. The ORR of first-line therapy was 38.7% (43/111). Median PFS (mPFS) was 9 months [95% confidence interval (CI) 7.5-10.5 months]. After progression, only 62% and 36% of the patients are fit to receive second or third lines of treatment, with limited clinical benefit. Median OS (mOS) was 21 months (95% CI 17.4-24.6 months). In patients receiving first-line triplet chemotherapy, ORR was 56.3% (9/16), mPFS was 13 months (95% CI 10.3-15.7 months) and mOS was 32 months (95% CI 7.7-56.3 months). For irinotecan-based doublets, ORR was 34.5 (10/29), mPFS was 9 months (95% CI 6.4-11.6 months) and mOS was 22 months (95% CI 16.0-28.0 months). With oxaliplatin-based doublets ORR was 36.4% (24/62), mPFS was 7 months (95% CI 4.6-9.4 months) and mOS was 18 months (95% CI, 13.6-22.4 months). CONCLUSION Patients with KRASG12C-mutant mCRC had a disappointing response to standard treatments. Within the limitations of a retrospective study, these results suggest that first-line chemotherapy intensification with FOLFOXIRI is a valid option in fit patients.
Collapse
Affiliation(s)
- D Ciardiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy; Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - C Chiarazzo
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - V Famiglietti
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - A Damato
- Medical Oncology Unit, Comprhensive Cancer Center, AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - C Pinto
- Medical Oncology Unit, Comprhensive Cancer Center, AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - M G Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - G Castellano
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - L Gervaso
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - A Zaniboni
- Medical Oncology Unit, Poliambulanza Foundation, Brescia, Italy
| | - E Oneda
- Medical Oncology Unit, Poliambulanza Foundation, Brescia, Italy
| | - S Rapisardi
- Medical Oncology Unit, ARNAS Garibaldi, Catania, Italy
| | - R Bordonaro
- Medical Oncology Unit, ARNAS Garibaldi, Catania, Italy
| | - C Zichi
- Department of Oncology, University of Turin, A.O. Ordine Mauriziano, Turin, Italy
| | - F De Vita
- Department of Oncology, University of Turin, A.O. Ordine Mauriziano, Turin, Italy
| | - M Di Maio
- Department of Oncology, University of Turin, A.O. Ordine Mauriziano, Turin, Italy
| | - A Parisi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy; Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - R Giampieri
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - R Berardi
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona, Italy
| | - D Lavacchi
- Clinical Oncology Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - L Antonuzzo
- Clinical Oncology Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - E Tamburini
- Oncology Department and Palliative Care, Cardinale Panico, Tricase City Hospital, Tricase, Italy
| | - B A Maiorano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy; Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| | - P Parrella
- Oncology Laboratory, Foundation Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo, Foggia, Italy
| | - T P Latiano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - N Normanno
- Cellular Biology and Biotherapy, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - A De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - A Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Naples, Italy
| | - G Martini
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - S Napolitano
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - T Troiani
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - E Martinelli
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - F Ciardiello
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy.
| | - F De Vita
- Medical Oncology Unit, Department of Precision Medicine, "Luigi Vanvitelli" University of Campania, Naples, Italy
| | - E Maiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
28
|
Ardito F, Razionale F, Campisi A, Carlino A, Vellone M, Vani S, Larocca LM, Giuliante F. The Impact of KRAS Mutational Status on Long-Term Survival following Liver Resection for Hilar Cholangiocarcinoma. Cancers (Basel) 2022; 14:4370. [PMID: 36139531 PMCID: PMC9496723 DOI: 10.3390/cancers14184370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
KRAS mutation is reportedly associated with poor prognosis in patients with different cancer types. However, mutational data on hilar cholangiocarcinoma are few and controversial. The aim of this study was to evaluate the rate of KRAS mutations in a single-center homogeneous population resected for hilar cholangiocarcinoma and the subsequent impact on prognosis. KRAS mutation status was evaluated in 54 patients undergoing major hepatectomy combined with resection of the main biliary confluence and regional lymphadenectomy for hilar cholangiocarcinoma between 2001 and 2019. Among these 54 patients, 12 (22.2%) had a KRAS mutation. KRAS mutation was not related with pathologic characteristics of the tumor. Five-year overall survival (OS) in patients with KRAS mutation was significantly lower than that observed in patients with KRAS wild type (0 vs. 49.2%, respectively; p = 0.003). In the multivariable analysis; independent predictors of poor OS were KRAS mutation (HR = 5.384; p = 0.003) and lymph node metastases (HR = 2.805; p = 0.023). The results of our study suggested that KRAS mutation in hilar cholangiocarcinoma was not rarely observed. KRAS mutation was an independent strong predictor of poor OS. KRAS mutation analysis should be included in the routine pathologic evaluation of resected hilar cholangiocarcinoma in order to better stratify prognosis.
Collapse
Affiliation(s)
- Francesco Ardito
- Hepatobiliary Surgery Unit, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Razionale
- Hepatobiliary Surgery Unit, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Campisi
- Hepatobiliary Surgery Unit, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Angela Carlino
- Department of Pathology, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Maria Vellone
- Hepatobiliary Surgery Unit, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Simone Vani
- Hepatobiliary Surgery Unit, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Luigi M. Larocca
- Department of Pathology, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Felice Giuliante
- Hepatobiliary Surgery Unit, Foundation Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|