1
|
Roazzi L, Patelli G, Bencardino KB, Amatu A, Bonazzina E, Tosi F, Amoruso B, Bombelli A, Mariano S, Stabile S, Porta C, Siena S, Sartore-Bianchi A. Ongoing Clinical Trials and Future Research Scenarios of Circulating Tumor DNA for the Treatment of Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:295-308. [PMID: 38519391 DOI: 10.1016/j.clcc.2024.02.001] [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: 09/14/2023] [Revised: 01/04/2024] [Accepted: 02/11/2024] [Indexed: 03/24/2024]
Abstract
Liquid biopsy using circulating tumor DNA (ctDNA) has emerged as a minimally invasive, timely approach to provide molecular diagnosis and monitor tumor evolution in patients with cancer. Since the molecular landscape of metastatic colorectal cancer (mCRC) is substantially heterogeneous and dynamic over space and time, ctDNA holds significant advantages as a biomarker for this disease. Numerous studies have demonstrated that ctDNA broadly recapitulates the molecular profile of the primary tumor and metastases, and have mainly focused on the genotyping of RAS and BRAF, that is propaedeutic for anti-EGFR treatment selection. However, ctDNA soon broadened its scope towards the assessment of early tumor response, as well as the identification of drug resistance biomarkers to drive potential molecular actionability. In this review article, we provide an overview of the current state-of-the-art of this methodology and its applications, focusing on ongoing clinical trials that employ ctDNA to prospectively guide treatment in patients with mCRC.
Collapse
Affiliation(s)
- Laura Roazzi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Katia Bruna Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Brunella Amoruso
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Anna Bombelli
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sara Mariano
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Stefano Stabile
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Camillo Porta
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| |
Collapse
|
2
|
Pesola G, Epistolio S, Cefalì M, Trevisi E, De Dosso S, Frattini M. Neo-RAS Wild Type or RAS Conversion in Metastatic Colorectal Cancer: A Comprehensive Narrative Review. Cancers (Basel) 2024; 16:3923. [PMID: 39682112 DOI: 10.3390/cancers16233923] [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: 09/06/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
The management of metastatic colorectal cancer in patients harboring RAS mutations primarily involves chemotherapy, often combined with bevacizumab, as a standard first-line treatment. However, emerging evidence suggests that tumors in a subset of these patients may experience a conversion from RAS-mutant status to RAS wild type (wt) during or after chemotherapy, a process referred to as "RAS conversion" or "neo-RAS wt". Understanding the mechanisms driving the neo-RAS wt phenomenon is crucial for its application in personalized medicine. Hypotheses suggest that selective pressure from chemotherapy may lead to a decrease in the number of mutant RAS clones or an outgrowth of pre-existing RAS wt clones. Further research is needed to validate these mechanisms and understand the impact of the neo-RAS wt phenomenon on long-term outcomes, such as overall survival and progression-free survival. This review provides a comprehensive overview of the current understanding of the neo-RAS wt phenomenon, including its incidence, potential mechanisms, and clinical implications.
Collapse
Affiliation(s)
- Guido Pesola
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Samantha Epistolio
- Laboratory of Genetics and Molecular Pathology, Istituto Cantonale di Patologia EOC, 6600 Locarno, Switzerland
| | - Marco Cefalì
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Elena Trevisi
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | - Sara De Dosso
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Milo Frattini
- Laboratory of Genetics and Molecular Pathology, Istituto Cantonale di Patologia EOC, 6600 Locarno, Switzerland
| |
Collapse
|
3
|
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
|
4
|
Albuquerque J, Neto da Silva D, Padrão T, Leal-Costa L, Bizarro R, Correia J, Baptista C, Machete M, Prazeres G, Margarido I, Fernandes G, Simões P, Timóteo T, Lopes F, Godinho J, Moreira-Pinto J, Rodrigues T, Faria A, Pulido C, Cirnes L, Teixeira JA, Passos Coelho JL. Loss of RAS Mutations in Liquid Biopsies of Patients With Multi-Treated Metastatic Colorectal Cancer. Oncologist 2024; 29:e337-e344. [PMID: 38071748 PMCID: PMC10911918 DOI: 10.1093/oncolo/oyad299] [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: 07/17/2023] [Accepted: 10/17/2023] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Liquid biopsy (LB) is a non-invasive tool to evaluate the heterogeneity of tumors. Since RAS mutations (RAS-mut) play a major role in resistance to antiepidermal growth factor receptor inhibitors (EGFR) monoclonal antibodies (Mabs), serial monitoring of RAS-mut with LB may be useful to guide treatment. The main aim of this study was to evaluate the prognostic value of the loss of RAS-mut (NeoRAS-wt) in LB, during the treatment of metastatic colorectal cancer (mCRC). METHODS A retrospective study was conducted on patients with mCRC between January 2018 and December 2021. RAS-mut were examined in tissue biopsy, at mCRC diagnosis, and with LB, during treatment. RESULTS Thirty-nine patients with RAS-mut mCRC were studied. LB was performed after a median of 3 lines (0-7) of systemic treatment including anti-vascular endothelial growth factor (anti-VEGF) Mabs. NeoRAS-wt was detected in 13 patients (33.3%); 9 (69.2%) of them received further treatment with anti-EGFR Mabs with a disease control rate of 44.4%. Median overall survival (OS), from the date of LB testing, was 20 months in the NeoRAS-wt group and 9 months in the persistent RAS-mut group (log-rank 2.985; P = .08), with a 12-month OS of 84.6% and 57.7%, respectively. NeoRAS-wt was identified as a predictor of survival (HR = 0.29; P = .007), with an 11-month improvement in median OS and a 71% decrease in risk of death, in heavily pretreated patients. CONCLUSIONS In conclusion, monitoring clonal evolution in mCRC by LB may provide an additional treatment line for patients with NeoRAS-wt in advanced disease.
Collapse
Affiliation(s)
| | | | - Teresa Padrão
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | - Luísa Leal-Costa
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Rita Bizarro
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Jorge Correia
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | - Carlota Baptista
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Madalena Machete
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Gil Prazeres
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | - Inês Margarido
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | | | - Pedro Simões
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Teresa Timóteo
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | - Fábio Lopes
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - João Godinho
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - João Moreira-Pinto
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Tânia Rodrigues
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | - Ana Faria
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Catarina Pulido
- Department of Medical Oncology, Hospital da Luz, Lisboa, Portugal
| | - Luís Cirnes
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
| | - José A Teixeira
- Department of Medical Oncology, Hospital Beatriz Ângelo, Loures, Portugal
| | | |
Collapse
|
5
|
Gramaça J, Fernandes IG, Trabulo C, Gonçalves J, dos Santos RG, Baptista A, Pina I. Emerging role of liquid biopsy in rat sarcoma virus mutated metastatic colorectal cancer: A case report. World J Gastrointest Oncol 2024; 16:234-243. [PMID: 38292846 PMCID: PMC10824108 DOI: 10.4251/wjgo.v16.i1.234] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/28/2023] [Accepted: 11/08/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND In patients with metastatic colorectal cancer (mCRC), the treatment options are limited and have been proved to be affected by rat sarcoma virus (RAS) mutational status. In RAS wild-type (wt) patients, the combination of anti-epidermal growth factor receptor (EGFR) monoclonal antibodies with chemotherapy (CT) is more effective than CT alone. On the other hand, RAS-mutated patients are not eligible for treatment with anti-EGFR antibodies. CASE SUMMARY Eleven patients with initially RAS-mutated mCRC were followed from diagnosis to May 2022. At the time of cell-free DNA determination, five patients had undergone one CT line, five patients had undergone two CT lines, and one patient had undergone three CT lines (all in combination with bevacizumab). At the second and third treatment lines [second line (2L), third line (3L)], patients with neo-RAS wt received a combination of CT and cetuximab. In neo-RAS wt patients treated with anti-EGFR, our findings indicated an increase in progression-free survival for both 2L and 3L (14.5 mo, P = 0.119 and 3.9 mo, P = 0.882, respectively). Regarding 2L overall survival, we registered a slight increase in neo-RAS wt patients treated with anti-EGFR (33.6 mo vs 32.4 mo, P = 0.385). At data cut-off, two patients were still alive: A RAS-mutated patient undergoing 3L treatment and a neo-RAS wt patient who received 2L treatment with anti-EGFR (ongoing). CONCLUSION Our case series demonstrated that monitoring RAS mutations in mCRC by liquid biopsy may provide an additional treatment line for neo-RAS wt patients.
Collapse
Affiliation(s)
- João Gramaça
- Centro Hospitalar Barreiro Montijo, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| | - Isabel Gomes Fernandes
- Medical Oncology Unit, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| | - Carolina Trabulo
- Medical Oncology Unit, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| | - Joana Gonçalves
- Medical Oncology Unit, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| | - Rita Gameiro dos Santos
- Medical Oncology Unit, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| | - Adriano Baptista
- Medical Oncology Unit, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| | - Idília Pina
- Medical Oncology Unit, Centro Hospitalar Barreiro Montijo, Setúbal, Barreiro 2830-003, Portugal
| |
Collapse
|
6
|
Callesen LB, Boysen AK, Andersen CSA, Pallisgaard N, Spindler KLG. The Importance of Feasibility Assessment in the Design of ctDNA Guided Trials - Results From the OPTIPAL II Study. Clin Colorectal Cancer 2023; 22:421-430.e1. [PMID: 37586928 DOI: 10.1016/j.clcc.2023.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Both quantitative and molecular changes in ctDNA can hold important information when treating metastatic colorectal cancer (mCRC), but its clinical utility is yet to be established. Before conducting a large-scale randomized trial, it is essential to test feasibility. This study investigates whether ctDNA is feasible for detecting patients who will benefit from treatment with epidermal growth factor receptor inhibitors and the prognostic value of circulating tumor DNA (ctDNA) response. MATERIALS AND METHODS Patients with mCRC, who were considered for systemic palliative treatment and were eligible for ctDNA analysis. Mutational testing on cell-free DNA (cfDNA) was done by ddPCR. ctDNA response from baseline to the third treatment cycle was evaluated in patients with detectable ctDNA at baseline. ctDNA maximum response was defined as undetectable ctDNA at the third treatment cycle, ctDNA partial response as any decrease in the ctDNA level, and ctDNA progression as any increase in the ctDNA level. RESULTS Forty-nine patients were included. The time to test results for mutational testing on cfDNA was significantly shorter than on tumor tissue (p < .001). Progression-free survival were 11.2 months (reference group), 7.5 months (HR = 10.7, p= .02), and 4.6 months (HR = 11.4, p= .02) in patients with ctDNA maximum response, partial response, and progression, respectively. Overall survival was 31.2 months (reference group), 15.2 months (HR = 4.1, p= .03), and 9.0 months (HR = 2.6, p= .03) in patients with ctDNA maximum response, partial response, and progression, respectively. CONCLUSION Pretreatment mutational testing on cfDNA in daily clinic is feasible and can be applied in randomized clinical trials evaluating the clinical utility of ctDNA. Early dynamics in ctDNA during systemic treatment hold prognostic value.
Collapse
Affiliation(s)
- Louise Bach Callesen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark; Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | | | - Christina Søs Auður Andersen
- Department of Pathology, Zealand University Hospital, Næstved, Denmark; Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Niels Pallisgaard
- Department of Pathology, Zealand University Hospital, Næstved, Denmark; Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Karen-Lise Garm Spindler
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark; Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
7
|
Harada K, Yuki S, Kawamoto Y, Nakamura T, Kaneko S, Ishida K, Sakamoto N, Komatsu Y. Anti-epidermal growth factor receptor treatment for patients with Neo RAS wild-type metastatic colorectal cancer: a case report of two cases. Ther Adv Med Oncol 2023; 15:17588359231216090. [PMID: 38033418 PMCID: PMC10685759 DOI: 10.1177/17588359231216090] [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: 06/30/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
The NeoRAS phenomenon is defined as the conversion of tumor RAS status from mutant-type (MT) to wild-type (WT) after systemic chemotherapy in metastatic colorectal cancer (mCRC). Cetuximab, an anti-epidermal growth factor receptor (EGFR) antibody, is effective in patients with RAS WT mCRC but ineffective in those with RAS MT mCRC; however, its outcome in patients with NeoRAS WT mCRC is unclear. Herein, we report two cases of NeoRAS WT mCRC that responded clinically to anti-EGFR treatment. The first was a 40-year-old man with synchronous peritoneal metastatic rectosigmoid cancer. The first RAS testing on tumor tissue revealed a KRAS G12C mutation, which was converted to RAS WT after two lines of chemotherapy, as assessed by liquid biopsy. After initiating irinotecan plus cetuximab treatment, a computed tomography (CT) scan revealed that malignant ascites had resolved. The treatment was discontinued after 4 months because of disease progression. The second was a 68-year-old male patient with synchronous liver metastasis from sigmoid colon cancer. The KRAS G12D mutation, initially detected in tumor tissue, was not detected by liquid biopsy after six lines of chemotherapy. Cetuximab monotherapy was initiated, and the liver metastases shrank significantly. The patient continued cetuximab monotherapy for 8 months without disease progression. Our cases demonstrate the efficacy of anti-EGFR therapy for NeoRAS WT mCRC and highlight the importance of capturing the gene mutation profile throughout the clinical course for optimal treatment selection.
Collapse
Affiliation(s)
- Kazuaki Harada
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Takeaki Nakamura
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Shiho Kaneko
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Koichi Ishida
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| |
Collapse
|
8
|
Ros J, Matito J, Villacampa G, Comas R, Garcia A, Martini G, Baraibar I, Saoudi N, Salvà F, Martin Á, Antista M, Toledo R, Martinelli E, Pietrantonio F, Boccaccino A, Cremolini C, Dientsmann R, Tabernero J, Vivancos A, Elez E. Plasmatic BRAF-V600E allele fraction as a prognostic factor in metastatic colorectal cancer treated with BRAF combinatorial treatments. Ann Oncol 2023; 34:543-552. [PMID: 36921693 DOI: 10.1016/j.annonc.2023.02.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Combination of a BRAF inhibitor (BRAFi) and an anti-epidermal growth factor receptor (EGFR), with or without a MEK inhibitor (MEKi), improves survival in BRAF-V600E-mutant metastatic colorectal cancer (mCRC) over standard chemotherapy. However, responses are heterogeneous and there are no available biomarkers to assess patient prognosis or guide doublet- or triplet-based regimens. In order to better characterize the clinical heterogeneity observed, we assessed the prognostic and predictive role of the plasmatic BRAF allele fraction (AF) for these combinations. PATIENTS AND METHODS A prospective discovery cohort including 47 BRAF-V600E-mutant patients treated with BRAFi-anti-EGFR ± MEKi in clinical trials and real-world practice was evaluated. Results were validated in an independent multicenter cohort (n= 29). Plasmatic BRAF-V600E AF cut-off at baseline was defined in the discovery cohort with droplet digital PCR (ddPCR). All patients had tissue-confirmed BRAF-V600E mutations. RESULTS Patients with high AF have major frequency of liver metastases and more metastatic sites. In the discovery cohort, median progression-free survival (PFS) and overall survival (OS) were 4.4 and 10.1 months, respectively. Patients with high BRAF AF (≥2%, n = 23) showed worse PFS [hazard ratio (HR) 2.97, 95% confidence interval (CI) 1.55-5.69; P = 0.001] and worse OS (HR 3.28, 95% CI 1.58-6.81; P = 0.001) than low-BRAF AF patients (<2%, n = 24). In the multivariable analysis, BRAF AF levels maintained independent significance. In the validation cohort, high BRAF AF was associated with worse PFS (HR 3.83, 95% CI 1.60-9.17; P = 0.002) and a trend toward worse OS was observed (HR 1.86, 95% CI 0.80-4.34; P = 0.15). An exploratory analysis of predictive value showed that high-BRAF AF patients (n = 35) benefited more from triplet therapy than low-BRAF AF patients (n = 41; PFS and OS interaction tests, P < 0.01). CONCLUSIONS Plasmatic BRAF AF determined by ddPCR is a reliable surrogate of tumor burden and aggressiveness in BRAF-V600E-mutant mCRC treated with a BRAFi plus an anti-EGFR with or without a MEKi and identifies patients who may benefit from treatment intensification. Our results warrant further validation of plasmatic BRAF AF to refine clinical stratification and guide treatment strategies.
Collapse
Affiliation(s)
- J Ros
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Matito
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - G Villacampa
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; The Institute of Cancer Research, London, UK
| | - R Comas
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Garcia
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - G Martini
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - I Baraibar
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - N Saoudi
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - F Salvà
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Á Martin
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - M Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - R Toledo
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - E Martinelli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - F Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - A Boccaccino
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - C Cremolini
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - R Dientsmann
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Vivancos
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - E Elez
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
9
|
Nicolazzo C, Magri V, Marino L, Belardinilli F, Di Nicolantonio F, De Renzi G, Caponnetto S, De Meo M, Giannini G, Santini D, Cortesi E, Gazzaniga P. Genomic landscape and survival analysis of ctDNA “neo-RAS wild-type” patients with originally RAS mutant metastatic colorectal cancer. Front Oncol 2023; 13:1160673. [PMID: 37064137 PMCID: PMC10093715 DOI: 10.3389/fonc.2023.1160673] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundThe term “neo-RAS wild-type” refers to the switch to RAS wild-type disease in plasma circulating tumor DNA (ctDNA) from originally RAS mutant colorectal cancers. Consistently, the hypothesis to re-determine RAS mutational status in ctDNA at disease progression in RAS mutant mCRC opened to a new perspective for clinically-based selection of patients to be treated with EGFR inhibitors. Currently, the genomic landscape of “neo-RAS wild-type” is unknown. This is a prospective study aimed to investigate clinical and genomic features associated with RAS mutation clearance in a large cohort of RAS mutant mCRC patients who converted to RAS wild- type in liquid biopsy at failure of first-line treatments. Secondary aim was to investigate the long term prognostic significance of “true neo-RAS wild- type”.Patients and methods70 patients with stage IV RAS mutant colorectal cancer were prospectively enrolled. Plasma samples were collected at progression from first-line treatment. RAS/BRAF mutations in plasma were assessed by RT-PCR. In RAS/BRAF wild-type samples, ctDNA was used to generate libraries using a 17 genes panel whose alteration has clinical relevance. To investigate the prognostic significance of RAS mutation clearance, test curves for PFS and OS were represented by Kaplan-Meier estimator plot and Log-rank test.ResultsThe most commonly detected actionable mutations in “neo-RAS wild-type” were: PIK3CA (35.7%); RET (11.9%); IDH1 (9.5%); KIT (7%); EGFR (7%); MET (4.7%); ERBB2 (4.7%); FGFR3 (4.7%). Both OS and post-progression survival were longer in patients with “neo-RAS wild-type” compared to those who remained RAS mutant (p<0.001 for both).ConclusionsDe-novo-targetable mutations occured in a large percentage of “neo-RAS wild-type”, being PIK3CA the most commonly detected. RAS mutation clearance in ctDNA is associated with long- term improvement of overall survival.
Collapse
Affiliation(s)
- Chiara Nicolazzo
- Lab. Liquid Biopsy, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Valentina Magri
- Department of Radiology, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Luca Marino
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Rome, Italy
| | - Francesca Belardinilli
- Lab. Liquid Biopsy, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Gianluigi De Renzi
- Lab. Liquid Biopsy, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Salvatore Caponnetto
- Department of Radiology, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Michela De Meo
- Lab. Liquid Biopsy, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Giannini
- Lab. Liquid Biopsy, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniele Santini
- Department of Radiology, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Enrico Cortesi
- Department of Radiology, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Paola Gazzaniga
- Lab. Liquid Biopsy, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- *Correspondence: Paola Gazzaniga,
| |
Collapse
|
10
|
Udagawa S, Ooki A, Shinozaki E, Fukuda K, Yamaguchi K, Osumi H. Circulating Tumor DNA: The Dawn of a New Era in the Optimization of Chemotherapeutic Strategies for Metastatic Colo-Rectal Cancer Focusing on RAS Mutation. Cancers (Basel) 2023; 15:1473. [PMID: 36900264 PMCID: PMC10001242 DOI: 10.3390/cancers15051473] [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: 01/03/2023] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Genotyping of tumor tissues to assess RAS and BRAF V600E mutations enables us to select optimal molecularly targeted therapies when considering treatment strategies for patients with metastatic colorectal cancer. Tissue-based genetic testing is limited by the difficulty of performing repeated tests, due to the invasive nature of tissue biopsy, and by tumor heterogeneity, which can limit the usefulness of the information it yields. Liquid biopsy, represented by circulating tumor DNA (ctDNA), has attracted attention as a novel method for detecting genetic alterations. Liquid biopsies are more convenient and much less invasive than tissue biopsies and are useful for obtaining comprehensive genomic information on primary and metastatic tumors. Assessing ctDNA can help track genomic evolution and the status of alterations in genes such as RAS, which are sometimes altered following chemotherapy. In this review, we discuss the potential clinical applications of ctDNA, summarize clinical trials focusing on RAS, and present the future prospects of ctDNA analysis that could change daily clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Hiroki Osumi
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| |
Collapse
|
11
|
Thrombotic Microangiopathy in the Setting of Colorectal Cancer: A Therapeutic Challenge with a Bad Prognosis. Hematol Rep 2023; 15:9-16. [PMID: 36648880 PMCID: PMC9844426 DOI: 10.3390/hematolrep15010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/08/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
While most cases of thrombotic microangiopathic hemolytic anemias are idiopathic, some can occur in the setting of a malignancy. Differentiating both conditions is crucial to initiate the appropriate treatment. In this case report and literature review, we discuss the occurrence of a thrombotic microangiopathy in a 61-year-old male patient with a treatment-refractory metastatic colorectal cancer invading his bone marrow. Plasmapheresis does not constitute the mainstay of treatment in this setting, as targeting the primary disease is the ultimate management. Treating the condition of our patient has been challenging as multiple lines of treatments of his primary disease had been exhausted. The discrepancy in KRAs status obtained between PCR and later NGS offered a new treatment line with Cetuximab. In this article, we will discuss the different factors that differentiate between idiopathic and cancer-induced microangiopathy. We will emphasize on the fact that the treatment of the primary disease constitutes the most important step in the treatment of cancer-induced thrombotic microangiopathy. We will also raise several explanations to target the disagreement in KRAS status obtained by the different technical modalities.
Collapse
|
12
|
Osumi H, Ishizuka N, Takashima A, Kumekawa Y, Nakano D, Shiozawa M, Denda T, Sawada R, Ouchi K, Wakatsuki T, Narikazu B, Kato K, Yamaguchi K, Shinozaki E. Multicentre single-arm phase II trial evaluating the safety and effiCacy of Panitumumab and iRinOtecan in NeoRAS Wild-type mEtaStatic colorectal cancer patientS (C-PROWESS trial): study protocol. BMJ Open 2022; 12:e063071. [PMID: 36581973 PMCID: PMC9438189 DOI: 10.1136/bmjopen-2022-063071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION A new concept of 'NeoRAS wild-type (WT)', which means conversion of RAS status from RAS mutant to RAS WT after treatment, has been reported. Previous observational and proof-of-concept studies have demonstrated the efficacy of epidermal growth factor receptor inhibitors in patients with NeoRAS WT metastatic colorectal cancer (mCRC). Moreover, posthoc biomarker analyses of these studies have suggested that not only the RAS status in the circulating tumour DNA (ctDNA) but also other gene mutational status may be useful as biomarkers of epidermal growth factor receptor inhibitors for NeoRAS WT mCRC. METHODS AND ANALYSIS This trial is a multicentre, single-arm, phase II trial to assess the efficacy and safety of panitumumab plus irinotecan therapy for patients with NeoRAS mCRC. The key eligibility criteria include RAS mutant mCRC initially proven in tumour tissue refractory or intolerant to fluoropyrimidine, oxaliplatin and irinotecan; RAS WT in ctDNA (defined as plasma mutant allele frequencies of all RAS ≤0.1%) within 28 days before enrolment and Eastern Cooperative Oncology Group performance status ≤2. The primary endpoint is the response rate. The target sample size is 30 patients. Biomarker analyses are planned to be performed using next-generation sequencing-based ctDNA analysis. ETHICS AND DISSEMINATION This study was approved by the certified review board of National Cancer Center Hospital. The main results of the trial will be presented in international meetings and in medical journals. TRIAL REGISTRATION NUMBER s031210565.
Collapse
Affiliation(s)
- Hiroki Osumi
- Department of Gastroenterology, Cancer Institute Hospital Gastroenterology Center, Koto-ku, Tokyo, Japan
| | - Naoki Ishizuka
- Department of Clinical Trial Planning and Management, Cancer Institute Hospital Gastroenterology Center, Koto-ku, Tokyo, Japan
| | - Atsuo Takashima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Yosuke Kumekawa
- Divison of Gastrointestinal Oncology, Saitama Cancer Center, Saitama, Japan
| | - Daisuke Nakano
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Manabu Shiozawa
- Department of Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Ryoichi Sawada
- Division of Gastroenterology and Hepatology, Jikei University Hospital, Tokyo, Japan
| | - Kota Ouchi
- Department of Medical Oncology, Tohoku University Hospital, Miyagi, Japan
| | - Takeru Wakatsuki
- Department of Gastroenterology, Cancer Institute Hospital Gastroenterology Center, Koto-ku, Tokyo, Japan
| | - Boku Narikazu
- Department of Medical Oncology and General Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ken Kato
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterology, Cancer Institute Hospital Gastroenterology Center, Koto-ku, Tokyo, Japan
| | - Eiji Shinozaki
- Department of Gastroenterology, Cancer Institute Hospital Gastroenterology Center, Koto-ku, Tokyo, Japan
| |
Collapse
|
13
|
Callesen LB, Hamfjord J, Boysen AK, Pallisgaard N, Guren TK, Kure EH, Spindler KLG. Circulating tumour DNA and its clinical utility in predicting treatment response or survival in patients with metastatic colorectal cancer: a systematic review and meta-analysis. Br J Cancer 2022; 127:500-513. [PMID: 35440666 PMCID: PMC9345951 DOI: 10.1038/s41416-022-01816-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We investigate the current knowledge on circulating tumour DNA (ctDNA) and its clinical utility in predicting outcomes in patients with metastatic colorectal cancer (mCRC). METHODS PubMed, Embase, Cochrane Database of Systematic Reviews and Cochrane Central Register of Controlled Trials were searched. Last search 16/12/2020. We included studies on patients with mCRC reporting the predictive or prognostic value of ctDNA. We performed separate random-effects meta-analyses to investigate if baseline ctDNA and early changes in ctDNA levels during treatment were associated with survival. The risk of bias was assessed according to the Quality in Prognosis Studies tool. RESULTS Seventy-one studies were included with 6930 patients. Twenty-four studies were included in meta-analyses. High baseline ctDNA level was associated with short progression-free survival (PFS) (HR = 2.2; 95% CI 1.8-2.8; n = 509) and overall survival (OS) (HR = 2.4; 95% CI 1.9-3.1; n = 1336). A small or no early decrease in ctDNA levels during treatment was associated with short PFS (HR = 3.0; 95% CI 2.2-4.2; n = 479) and OS (HR = 2.8; 95% CI 2.1-3.9; n = 583). Results on clonal evolution and lead-time were inconsistent. A majority of included studies (n = 50/71) had high risk of bias in at least one domain. CONCLUSIONS Plasma ctDNA is a strong prognostic biomarker in mCRC. However, true clinical utility is lacking.
Collapse
Affiliation(s)
- Louise B Callesen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Julian Hamfjord
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anders K Boysen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Pallisgaard
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Tormod K Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Elin H Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Natural Sciences and Environmental Health, Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Campus Bø, Bø, Norway
| | - Karen-Lise G Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
14
|
Fischer LE, Stintzing S, Heinemann V, Keilholz U, Keune D, Vollbrecht C, Burmeister T, Kind A, Weiss L, Horst D, Kirchner T, Klauschen F, Jung A, Westphalen CB, Jelas I. Liquid Biopsy in Colorectal Cancer: Quo Vadis? Implementation of Liquid Biopsies in Routine Clinical Patient Care in Two German Comprehensive Cancer Centers. Front Oncol 2022; 12:870411. [PMID: 35646657 PMCID: PMC9134071 DOI: 10.3389/fonc.2022.870411] [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: 02/06/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives The use of liquid biopsies (LB) in patients with solid malignancies enables comprehensive genomic profiling (CGP) of circulating tumor DNA (ctDNA) and has the potential to guide therapy stratification and support disease monitoring. To examine clinical uptake of LB in a real-world setting, LB implementation was analyzed at two German cancer centers (LMU Munich and Charité - Universitätsmedizin Berlin) between 2017 and 2021, with focus on colorectal cancer (CRC) patients. Methods In this retrospective analysis, all patients who received a LB between January 2017 and December 2021 as part of routine clinical management were included. To provide adequate context, we collected disease characteristics and technical specifications of the LB methods applied. Additionally, we examined the concordance of RAS status in tumor tissue and LB. Finally, we discuss the potential of LB as a diagnostic tool to drive personalized treatment in CRC patients and how to implement LB in clinical routine. Results In total, our cohort included 86 CRC patients and 161 LB conducted in these patients between 2017 and 2021. In 59 patients, comparison between tissue-based and liquid-based molecular diagnostics, revealed a divergence in 23 (39%) of the evaluable samples. Conclusion Our real-world data analysis indicates that the possibilities of LB are not yet exploited in everyday clinical practice. Currently, the variety of methods and lack of standardization, as well as restricted reimbursement for liquid based CGP hinder the use of LB in clinical routine. To overcome these issues, prospective clinical trials are needed to provide evidence driving the implementation of LB into the management of CRC patients and to support their implementation into clinical guidelines.
Collapse
Affiliation(s)
- Laura E Fischer
- Department of Medicine III, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Ulrich Keilholz
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany.,Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dietmar Keune
- Charité Comprehensive Cancer Center, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudia Vollbrecht
- Institute of Pathology Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Burmeister
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Labor Berlin - Charité Vivantes, GmbH, Molekulardiagnostik - Hämatologie, Berlin, Germany
| | - Andreas Kind
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lena Weiss
- Department of Medicine III, University Hospital, Munich, Germany
| | - David Horst
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany.,Institute of Pathology Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Kirchner
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.,Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, München, Germany
| | - Frederick Klauschen
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.,Institute of Pathology Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, München, Germany
| | - Andreas Jung
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany.,Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, München, Germany
| | - Christoph Benedikt Westphalen
- Department of Medicine III, University Hospital, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital Munich, Munich, Germany
| | - Ivan Jelas
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany
| |
Collapse
|
15
|
RAS Mutation Conversion in Bevacizumab-Treated Metastatic Colorectal Cancer Patients: A Liquid Biopsy Based Study. Cancers (Basel) 2022; 14:cancers14030802. [PMID: 35159069 PMCID: PMC8833999 DOI: 10.3390/cancers14030802] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Recent evidence has been provided that the clonal evolution of mutant RAS colorectal tumors may lead to the negative selection of mutant RAS clones, with the appearance of a time window characterized by the disappearance of RAS mutant clones in plasma. We demonstrate here for the first time that the use of bevacizumab in the first-line treatment is the most significant factor for RAS conversion from mutant to wild type in plasma. The frequent appearance of this “RAS wild-type * window” in patients treated with a first line treatment containing bevacizumab could possibly present them as candidates for second line treatment with anti-EGFR monoclonal antibodies, which are otherwise precluded. Abstract Liquid biopsies have shown that, in RAS mutant colorectal cancer, the conversion to RAS wild-type * status during the course of the disease is a frequent event, supporting the concept that the evolutionary landscape of colorectal cancer can lead to an unexpected negative selection of RAS mutant clones. The aim of the present study was to clarify whether the negative selection of RAS mutation in plasma might be drug-dependent. For this purpose, we used liquid biopsy to compare the rate of conversion from RAS mutant to RAS wild-type * in two groups of originally RAS mutant mCRC patients: the first treated with chemotherapy alone, while the second was treated with chemotherapy combined with bevacizumab. Serial liquid biopsies were performed at 3 months (T1), 6 months (T2), 9 months (T3), and 12 months (T4) after starting first line treatments. We found that the only independent variable significantly associated to RAS status conversion was the use of bevacizumab. RAS conversion was not found associated to tumor burden reduction, although bevacizumab-treated patients who converted to RAS wild-type * had a significantly longer PFS compared to patients who remained RAS mutant. The appearance of a “RAS wild-type * window”, mainly in bevacizumab-treated patients, might present them as candidates for second line treatment with anti-EGFR, which was otherwise precluded.
Collapse
|
16
|
Gazzaniga P, Nicolazzo C, Caponnetto S, Cortesi E. About RAS Mutation Clearance in Plasma ctDNA From RAS-Mutant Colorectal Cancer Patients. JCO Precis Oncol 2022; 5:389-390. [PMID: 34994600 DOI: 10.1200/po.20.00376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Paola Gazzaniga
- Paola Gazzaniga, PhD and Chiara Nicolazzo, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy and Salvatore Caponnetto, PhD and Enrico Cortesi, MD, Department of Radiology, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Chiara Nicolazzo
- Paola Gazzaniga, PhD and Chiara Nicolazzo, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy and Salvatore Caponnetto, PhD and Enrico Cortesi, MD, Department of Radiology, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Salvatore Caponnetto
- Paola Gazzaniga, PhD and Chiara Nicolazzo, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy and Salvatore Caponnetto, PhD and Enrico Cortesi, MD, Department of Radiology, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Enrico Cortesi
- Paola Gazzaniga, PhD and Chiara Nicolazzo, PhD, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy and Salvatore Caponnetto, PhD and Enrico Cortesi, MD, Department of Radiology, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
17
|
Macedo F, Monteiro J, Pereira TC, Monteiro A, Felix Soares R, Bonito N, Sousa G. Therapeutic impact of determination of RAS mutations in the plasma of patient with colorectal cancer. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2022; 15:93-98. [PMID: 35611253 PMCID: PMC9123636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/29/2021] [Indexed: 11/21/2022]
Abstract
Stage IV colorectal cancer treatment includes targeted therapy depending on RAS status. During disease progression, loss or gain of RAS mutations could happen, supporting the hypothesis of the evolutionary pressure of therapy. Circulating tumor DNA (ctDNA) are nucleic acids released to the bloodstream by the tumor during its development and may be detected by liquid biopsy. The Idylla© Biocartis, a fully automated real-time-PCR-based molecular diagnostic system, was used in a patient with metastatic colorectal cancer with a NRAS mutation in progression after several therapeutic lines. The ctDNA mutational analysis was performed and revealed the absence of mutations in the KRAS, NRAS, and BRAF genes. The patient started the third line of palliative chemotherapy with irinotecan + cetuximab and achieved a partial response for the first time. The authors describe a case in which liquid biopsy determined the higher progression-free survival achieved.
Collapse
|
18
|
Bouchahda M, Saffroy R, Karaboué A, Hamelin J, Innominato P, Saliba F, Bosselut N, Lemoine A, Lévi F. Reply to P. Gazzaniga et al. JCO Precis Oncol 2021; 5:391-392. [DOI: 10.1200/po.20.00465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Mohamed Bouchahda
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Raphael Saffroy
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Abdoulaye Karaboué
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Jocelyne Hamelin
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Pasquale Innominato
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Faouzi Saliba
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Nelly Bosselut
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Antoinette Lemoine
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| | - Francis Lévi
- Mohamed Bouchahda, MD, Medical Oncology Department, Paul Brousse Hospital, Villejuif, France; UPR “Chronotherapy, Cancer and Transplantation,” Medical School, Paris-Saclay University, Villejuif, France; Medical Oncology Unit, Clinique Saint Jean L'Ermitage, Melun, France; Medical Oncology Unit, Clinique du Mousseau, Evry, France; Raphael Saffroy, MD, Oncogenetics Department, APHP, Université Paris-Saclay, Paul Brousse Hospital, Villejuif, France; Abdoulaye Karaboué, MD, UPR “Chronotherapy, Cancer and
| |
Collapse
|
19
|
Clinical Applications of Minimal Residual Disease Assessments by Tumor-Informed and Tumor-Uninformed Circulating Tumor DNA in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184547. [PMID: 34572774 PMCID: PMC8471730 DOI: 10.3390/cancers13184547] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Circulating tumor DNA, or ctDNA, are fragments of tumor DNA that can be detected in the blood of patients with colorectal cancer. Measuring ctDNA levels in the blood has shown the potential to provide important information that can be helpful in the clinical care of patients with colorectal cancer. For example, in patients with colon cancer that has been removed by surgery, measuring ctDNA in the blood can predict the likelihood of cancer recurrence, while in those with metastatic colorectal cancer, measuring ctDNA can inform the clinician whether chemotherapy is effective at earlier timepoints than currently available tests. In this review, we discuss the results from ongoing studies describing the utility of ctDNA measurements across all stages of colorectal cancer. We also discuss the various clinical scenarios that ctDNA may have the most immediate impact in colorectal cancer management. Abstract Emerging data suggest that circulating tumor DNA (ctDNA) can detect colorectal cancer (CRC)-specific signals across both non-metastatic and metastatic settings. With the development of multiple platforms, including tumor-informed and tumor-agnostic ctDNA assays and demonstration of their provocative analytic performance to detect minimal residual disease, there are now ongoing, phase III randomized clinical trials to evaluate their role in the management paradigm of CRC. In this review, we highlight landmark studies that have formed the basis for ongoing studies on the clinically applicability of plasma ctDNA assays in resected, stage I–III CRC and metastatic CRC. We discuss clinical settings by which ctDNA may have the most immediate impact in routine clinical practice. These include the potential for ctDNA to (1) guide surveillance and intensification or de-intensification strategies of adjuvant therapy in resected, stage I–III CRC, (2) predict treatment response to neoadjuvant therapy in locally advanced rectal cancer inclusive of total neoadjuvant therapy (TNT), and (3) predict response to systemic and surgical therapies in metastatic disease. We end by considering clinical variables that can influence our ability to reliably interpret ctDNA dynamics in the clinic.
Collapse
|
20
|
DE Santiago BG, López-Gómez M, Delgado-López PD, Gordo AJ, Neria F, Thuissard-Vasallo IJ, Gómez-Raposo C, Tevar FZ, Moreno-Rubio J, Hernández AM, Iglesias I, Casado E. RAS Mutational Status in Advanced Colorectal Adenocarcinoma Treated With Anti-angiogenics: Preliminary Experience With Liquid Biopsy. In Vivo 2021; 35:2841-2844. [PMID: 34410976 PMCID: PMC8408740 DOI: 10.21873/invivo.12571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023]
Abstract
AIM To determinate molecular changes in the downstream epidermal growth factor receptor signaling pathway using serial liquid biopsies in patients with metastatic colorectal tumors (mCRC) under anti-angiogenic treatment. PATIENTS AND METHODS Determination of RAS mutation in primary tissue samples from colorectal tumors was performed in the 23 patients included in the study at diagnosis using quantitative-polymerase chain reaction. Sequential mutations were studied in circulating tumor (ct) DNA obtained from plasma samples. RESULTS Twenty-three patients with RAS-mutated primary tumors were included. In the first ctDNA determination, 17 of these patients were found to have wild-type RAS status. Remarkably, three out of these 17 wild-type cases changed to RAS-mutated in subsequent ctDNA assays. CONCLUSION Serial liquid biopsies in patients with mCRC might be a useful tool for identifying changes in the RAS mutation status in patients who had undergone previous anti-angiogenic therapy. The understanding of these changes might help to better define the landscape of mCRC and be the path to future randomized studies.
Collapse
Affiliation(s)
| | - Miriam López-Gómez
- Department of Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain
| | | | - Ana Jiménez Gordo
- Department of Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain
| | - Fernando Neria
- Faculty of Biomedical Science and Health, Universidad Europea, Madrid, Spain
| | | | - César Gómez-Raposo
- Department of Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain
| | | | - Juan Moreno-Rubio
- Clinical Oncology Group, IMDEA Food Institute, CEI UAM+CSIC, Madrid, Spain
| | | | - Irene Iglesias
- Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, Complutense University Madrid, Madrid, Spain
| | - Enrique Casado
- Department of Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain
| |
Collapse
|
21
|
Osumi H, Vecchione L, Keilholz U, Vollbrecht C, Alig AHS, von Einem JC, Stahler A, Striefler JK, Kurreck A, Kind A, Modest DP, Stintzing S, Jelas I. NeoRAS wild-type in metastatic colorectal cancer: Myth or truth?-Case series and review of the literature. Eur J Cancer 2021; 153:86-95. [PMID: 34153718 DOI: 10.1016/j.ejca.2021.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022]
Abstract
Upfront KRAS and NRAS gene testing ('RAS') is the standard of care for metastatic colorectal cancer (mCRC), to guide first-line treatment. The presence of RAS mutation (MT) is a negative predictor for the efficacy of anti-EGFR antibodies and the use of cetuximab and panitumumab is restricted to RAS wild-type (WT) mCRC. Conversion from RAS WT to RAS MT mCRC after treatment with anti-EGFR antibodies is a known and well-described acquired resistance mechanism. The by far less frequent 'NeoRAS wild-type' phenomenon (reversion from RAS MT to RAS WT) has recently drawn attention. The proposed effect of chemotherapy on RAS status in mCRC patients is not fully understood. Because of the intriguing biological consequence of a RAS MT to RAS WT reversion, subsequent treatment of NeoRAS WT patients with anti-EGFR antibodies is increasingly being discussed. Here, we report three clinical cases of NeoRAS WT mCRC patients, which received standard-of-care regimens for RAS MT mCRC. Anti-EGFR antibodies were used in two out of three patients after progression of the disease. One of the patients had a long-term response. In line with our observations, NeoRAS WT phenomenon occurs in clinical practice. Retesting of RAS status during treatment should be discussed in patients with unusual long-term clinical courses of RAS MT mCRC to optimise treatment strategy and to evaluate the use of anti-EGFR antibodies.
Collapse
Affiliation(s)
- Hiroki Osumi
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Loredana Vecchione
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Vollbrecht
- Institute of Pathology Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Annabel H S Alig
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jobst C von Einem
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Arndt Stahler
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jana K Striefler
- Department of Hematology, Oncology, and Tumor Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Annika Kurreck
- Department of Hematology, Oncology, and Tumor Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Kind
- Department of Hematology, Oncology, and Tumor Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dominik P Modest
- Department of Hematology, Oncology, and Tumor Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ivan Jelas
- Department of Hematology, Oncology, and Tumor Immunology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Hematology, Oncology, and Tumor Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
22
|
Nicolazzo C, Barault L, Caponnetto S, Macagno M, De Renzi G, Gradilone A, Belardinilli F, Cortesi E, Di Nicolantonio F, Gazzaniga P. Circulating Methylated DNA to Monitor the Dynamics of RAS Mutation Clearance in Plasma from Metastatic Colorectal Cancer Patients. Cancers (Basel) 2020; 12:E3633. [PMID: 33291569 PMCID: PMC7761880 DOI: 10.3390/cancers12123633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/02/2020] [Indexed: 01/20/2023] Open
Abstract
The clearance of RAS mutations in plasma circulating tumor DNA (ctDNA) from originally RAS-mutant metastatic colorectal cancer (mCRC) has been recently demonstrated. Clinical trials investigating whether RAS mutant mCRC who "convert" to wild-type in plasma might benefit from EGFR blockade are ongoing. Detection of tumor-specific DNA methylation alterations in ctDNA has been suggested as a specific tool to confirm the tumoral origin of cell-free DNA. We monitored RAS clearance in plasma from patients with RAS-mutant mCRC at baseline (pre-treatment) (T0); after 4 months of first-line therapy (T1); at the time of first (T2) and second (T3) progression. A five-gene methylation panel was used to confirm the presence of ctDNA in samples in which RAS mutation clearance was detected. At T1, ctDNA analysis revealed wild-type RAS status in 83% of samples, all not methylated, suggesting at this time point the lack of ctDNA shedding. At T2, ctDNA analysis revealed wild-type RAS status in 83% of samples, of which 62.5% were found methylated. At T3, 50% of wild-type RAS samples were found methylated. Non-methylated samples were found in patients with lung or brain metastases. This five-gene methylation test might be useful to confirm the presence of ctDNA in RAS wild-type plasma samples.
Collapse
Affiliation(s)
- Chiara Nicolazzo
- Liquid Biopsy Unit, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (G.D.R.); (A.G.)
| | - Ludovic Barault
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; (L.B.); (F.D.N.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy;
| | - Salvatore Caponnetto
- Department of Radiology, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy; (S.C.); (E.C.)
| | - Marco Macagno
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy;
| | - Gianluigi De Renzi
- Liquid Biopsy Unit, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (G.D.R.); (A.G.)
| | - Angela Gradilone
- Liquid Biopsy Unit, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (G.D.R.); (A.G.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | | | - Enrico Cortesi
- Department of Radiology, Oncology and Pathology, Sapienza University of Rome, 00161 Rome, Italy; (S.C.); (E.C.)
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, 10060 Candiolo, TO, Italy; (L.B.); (F.D.N.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy;
| | - Paola Gazzaniga
- Liquid Biopsy Unit, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (G.D.R.); (A.G.)
| |
Collapse
|