1
|
Guo X, Liu Y, Wang YT, Liu K, Ding H. Combined BRAF G469A mutation and echinoderm microtubule associated protein like-4-anaplastic lymphoma kinase rearrangement with resistance: A case report and review of literature. World J Clin Oncol 2025; 16:98812. [DOI: 10.5306/wjco.v16.i2.98812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/13/2024] [Accepted: 10/24/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Through deeper understanding of targetable driver mutations in non-small-cell lung cancer (NSCLC) over the past years, some patients with driver mutations have benefited from the targeted molecular therapies. Although the anaplastic lymphoma kinase and BRAF mutations are not frequent subtypes in NSCLC, the availability of several targeted-drugs has been confirmed through a series of clinical trials. But little is clear about the proper strategy in rare BRAF G469A mutation, not to mention co-exhibition of anaplastic lymphoma kinase and BRAF G469A mutations, which is extremely rare in NSCLC.
CASE SUMMARY We present a patient to stage IVA lung adenocarcinoma with coexisting echinoderm microtubule associated protein like-4 rearrangement and BRAF G469A mutation. She received several targeted drugs with unintended resistance and suffered from unbearable adverse events.
CONCLUSION Due to the rarity of co-mutations, the case not only enriches the limited literature on NSCLC harbouring BRAF G469A and echinoderm microtubule associated protein like-4 mutations, but also suggests the efficacy and safety of specific multiple-drug therapy in such patients.
Collapse
Affiliation(s)
- Xuan Guo
- Department of Pulmonary and Critical Care Medicine, Yixing People’s Hospital of Jiangsu University, Yixing 214200, Jiangsu Province, China
| | - Yan Liu
- Department of Pulmonary and Critical Care Medicine, Yixing People’s Hospital of Jiangsu University, Yixing 214200, Jiangsu Province, China
| | - Yu-Ting Wang
- Department of Pulmonary and Critical Care Medicine, Yixing People’s Hospital of Jiangsu University, Yixing 214200, Jiangsu Province, China
| | - Kan Liu
- Department of Pulmonary and Critical Care Medicine, Yixing People’s Hospital of Jiangsu University, Yixing 214200, Jiangsu Province, China
| | - Hui Ding
- Department of Pulmonary and Critical Care Medicine, Yixing People’s Hospital of Jiangsu University, Yixing 214200, Jiangsu Province, China
| |
Collapse
|
2
|
Dandapath I, Das S, Charan BD, Garg A, Suri A, Kedia S, Sharma MC, Sarkar C, Khonglah Y, Ahmed S, Suri V. Evaluation of KIAA1549::BRAF fusions and clinicopathological insights of pilocytic astrocytomas. Ann Diagn Pathol 2024; 72:152318. [PMID: 38733671 DOI: 10.1016/j.anndiagpath.2024.152318] [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: 02/14/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Pilocytic astrocytoma (PAs) represents a significant portion of childhood primary brain tumors, with distinct histological and radiological features. The prevalence of KIAA1549::BRAF fusion in PAs has been well-established, this study aims to assess the prevalence of KIAA1549::BRAF fusions and explore their associations with tumor characteristics, radiological findings, and patient outcomes in PAs. METHODS Histologically confirmed cases of PAs from a 5-year period were included in the study. Demographic, histopathological, and radiological data were collected, and immunohistochemistry was performed to characterize tumor markers. FISH and qRT-PCR assays were employed to detect KIAA1549::BRAF fusions. Statistical analyses were conducted to examine associations between fusion status and various other parameters. RESULTS Histological analysis revealed no significant differences in tumor features based on fusion status. However, younger age groups showed higher fusion prevalence. Radiologically, fusion-positive cases were distributed across different tumor subtypes SE, CWE and NCWE. Survival analysis did not demonstrate a significant impact of fusion status on overall survival, however most cases with recurrence and death harboured KIAA1549::BRAF fusion. Of 200 PAs, KIAA1549::BRAF fusions were detected in 64 % and 74 % of cases via qRT-PCR and FISH, respectively. Concordance between the two platforms was substantial (86 %). CONCLUSION KIAA1549::BRAF fusions are prevalent in PAs and can be reliably detected using both FISH and qRT-PCR assays. Cost considerations suggest qRT-PCR as a more economical option for fusion detection in routine clinical practice.
Collapse
Affiliation(s)
- Iman Dandapath
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Sumanta Das
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Bheru Dan Charan
- Department of Neuroradiology, All, India Institute of Medical Science, New Delhi, India
| | - Ajay Garg
- Department of Neuroradiology, All, India Institute of Medical Science, New Delhi, India
| | - Ashish Suri
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Shweta Kedia
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar Chand Sharma
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Yookarin Khonglah
- Department of Pathology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences, Shillong, Meghalaya, India
| | - Shabnam Ahmed
- Department of Pathology, GNRC Hospitals, Dispur, Assam, India
| | - Vaishali Suri
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
3
|
Girod M, Dalle S, Mortier L, Dalac S, Leccia MT, Dutriaux C, Montaudié H, de Quatrebarbes J, Lesimple T, Brunet-Possenti F, Saiag P, Maubec E, Legoupil D, Stoebner PE, Arnault JP, Lefevre W, Lebbe C, Dereure O. Non-V600E/K BRAF Mutations in Metastatic Melanoma: Molecular Description, Frequency, and Effectiveness of Targeted Therapy in a Large National Cohort. JCO Precis Oncol 2022; 6:e2200075. [DOI: 10.1200/po.22.00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Mitogen-activating protein kinase inhibitors (MAPKis) are largely used in V600E/K BRAF–mutated metastatic melanomas, but data regarding effectiveness of targeted therapy in patients with rare BRAF mutations and molecular description of these infrequent mutations are scarce. PATIENTS AND METHODS A multicenter study was conducted on patients with metastatic melanoma harboring a well-identified mutation of BRAF and enrolled from March 2013 to June 2021 in the French nationwide prospective cohort MelBase. The molecular BRAF mutation pattern, response to MAPKis when applicable, and survival data were analyzed. RESULTS Of 856 selected patients, 51 (6%) harbored a non-V600E/K BRAF mutation involving codons V600 (24 of 51, 47%; V600G 27.4%, V600R 15.6%), K601 (6 of 51, 11.7%), and L597 (4 of 51, 7.8%). An objective response to MAPKis either BRAF inhibitor (BRAFi) alone or combined with MEK inhibitor was achieved in 56% (353 of 631) of V600E/K, 58% (11 of 19) of non-E/K V600, and 22% (2 of 9) of non-V600 BRAF-mutated patients, with a median progression-free survival of 7.7, 7.8, and 2.8 months, respectively. Overall, objective response rate was higher with BRAFi + MEK inhibitor combination than with BRAFi in monotherapy for each subset. CONCLUSION Rare BRAF mutations are not anecdotal in the metastatic melanoma population. Although data interpretation must remain careful owing to the limited size of some subsets of patients, non-E/K V600 BRAF mutations seem to confer a high sensitivity to targeted therapy, whereas MAPKis seem less effective in patients with non-V600 BRAF mutations. However, this strategy may be used as an alternative option in the case of immunotherapy failure in the latter population.
Collapse
Affiliation(s)
- Manon Girod
- Department of Dermatology, University of Montpellier, Montpellier, France
| | - Stéphane Dalle
- Department of Dermatology, Service de dermatologie, Hôpital Lyon Sud, Centre de recherche en cancérologie de Lyon, Université Claude Bernard Lyon 1, Hospices Civils de Lyon, Lyon, France
| | | | - Sophie Dalac
- Department of Dermatology, Hôpital du bocage, Dijon, France
| | | | - Caroline Dutriaux
- Department of Dermatology, Centre Hospitalier Universitaire, Bordeaux, France
| | - Henri Montaudié
- Department of Dermatology, University Hospital of Nice, Université Côte d'Azur and INSERM U1065, Centre Méditerranéen de Médecine Moléculaire, Université Côte d'Azur, Nice, France
| | | | - Thierry Lesimple
- Department of Medical Oncology, Centre Régional de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | | | - Philippe Saiag
- Department of General and Oncologic Dermatology, Ambroise-Paré Hospital, APHP & EA3440 "Biomarkers in Cancerology and Hemato-Oncology”, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Eve Maubec
- Department of Dermatology, Hôpital Avicenne, Bobigny, France
| | - Delphine Legoupil
- Department of Dermatology, Centre Hospitalier Universitaire, Brest, France
| | | | | | - Wendy Lefevre
- Department of Dermatology, MelBase, Hôpital Saint-Louis, Paris, France
| | - Celeste Lebbe
- Department of Dermatology, DMU ICARE, AP-HP Hôpital Saint Louis and INSERM U976, Université de Paris, Paris, France
| | - Olivier Dereure
- Department of Dermatology, University of Montpellier, Montpellier, France
- INSERM U1058 Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, Montpellier, France
| |
Collapse
|
4
|
Toutain G, Min V, Rome A, Andre N. Trametinib for a BRAF G469A missense mutation in a neuroblastoma unveiled by liquid biopsy. Pediatr Blood Cancer 2022; 69:e29742. [PMID: 35652680 DOI: 10.1002/pbc.29742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Gauthier Toutain
- Department of Pediatric Oncology, La Timone University Hospital of Marseille, APHM, Marseille, France
| | - Victoria Min
- Department of Pediatric Oncology, La Timone University Hospital of Marseille, APHM, Marseille, France
| | - Angélique Rome
- Department of Pediatric Oncology, La Timone University Hospital of Marseille, APHM, Marseille, France
| | - Nicolas Andre
- Department of Pediatric Oncology, La Timone University Hospital of Marseille, APHM, Marseille, France.,Centre d'Essais Précoces en Cancérologie de Marseille (CEPCM), CLIPP2, APHM, Marseille, France.,SMARTc Unit, CRCM Inserm 1068-CNRS UMR 7258 Aix-Marseille University, Marseille, France
| |
Collapse
|
5
|
Argani P, Tickoo SK, Matoso A, Pratilas CA, Mehra R, Tretiakova M, Sibony M, Meeker AK, Lin MT, Reuter VE, Epstein JI, Gagan J, Palsgrove DN. Adult Wilms Tumor: Genetic Evidence of Origin of a Subset of Cases From Metanephric Adenoma. Am J Surg Pathol 2022; 46:988-999. [PMID: 35184066 PMCID: PMC9310085 DOI: 10.1097/pas.0000000000001864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The genetics of nephroblastoma (Wilms tumor) occurring in adults is largely unknown, as studies have largely been limited to isolated case reports. We, therefore, studied 14 adult Wilms tumors for genetic alterations, using expanded targeted sequencing on 11 cases. The patients ranged from 17 to 46 years of age (mean and median, 31 y), and there were 8 males and 6 females. Five Wilms tumors harbored BRAF V600E mutations. All of these had better-differentiated areas identical to metanephric adenoma, as has previously been described. In 3 such cases, microdissection studies revealed that the BRAF V600E mutation was present in both the metanephric adenoma and Wilms tumor areas; however, additional genetic alterations (including TERT promoter mutations in 2 cases, ASLX1/ATR mutations in 1 other case) were limited to the Wilms tumor component. These findings suggest that the Wilms tumor developed from the metanephric adenoma. Other adult Wilms tumors harbored genetic alterations previously reported in the more common pediatric Wilms tumors, including WT1 mutations (2 cases), ASLX1 mutations (3 additional cases), NSD2 mutation (1 additional case), and 11p loss (3 cases). In summary, a significant subset of adult Wilms tumors (specifically those of epithelial type with differentiated areas) harbor targetable BRAF V600E mutations and appear to arise from metanephric adenomas as a consequence of additional acquired genetic alterations. Other adult Wilms tumors often harbor genetic alterations found in their more common pediatric counterparts, suggesting at least some similarities in their pathogenesis.
Collapse
Affiliation(s)
| | - Satish K. Tickoo
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Andres Matoso
- Departments of Pathology
- Departments of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Rohit Mehra
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI
| | - Maria Tretiakova
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA
| | | | - Alan K. Meeker
- Departments of Pathology
- Departments of Oncology
- Departments of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Victor E. Reuter
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jonathan I. Epstein
- Departments of Pathology
- Departments of Oncology
- Departments of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Doreen N. Palsgrove
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
6
|
Dandapath I, Chakraborty R, Kaur K, Mahajan S, Singh J, Sharma MC, Sarkar C, Suri V. Molecular alterations of low-grade gliomas in young patients: Strategies and platforms for routine evaluation. Neurooncol Pract 2021; 8:652-661. [PMID: 34777834 PMCID: PMC8579091 DOI: 10.1093/nop/npab053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In recent years, it has been established that molecular biology of pediatric low-grade gliomas (PLGGs) is entirely distinct from adults. The majority of the circumscribed pediatric gliomas are driven by mitogen-activated protein kinase (MAPK) pathway, which has yielded important diagnostic, prognostic, and therapeutic biomarkers. Further, the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT) Steering Committee in their fourth meeting, suggested including a panel of molecular markers for integrated diagnosis in "pediatric-type" diffuse gliomas. However, a designated set of platforms for the evaluation of these alterations has yet not been mentioned for easier implementation in routine molecular diagnostics. Herein, we have reviewed the relevance of analyzing these markers and discussed the strategies and platforms best apposite for clinical laboratories.
Collapse
Affiliation(s)
- Iman Dandapath
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Kavneet Kaur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Mahajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotsna Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar C Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
7
|
Detection of Circulating Tumor DNA with a Single-Molecule Sequencing Analysis Validated for Targeted and Immunotherapy Selection. Mol Diagn Ther 2020; 23:521-535. [PMID: 31209714 PMCID: PMC6675782 DOI: 10.1007/s40291-019-00406-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Comprehensive genetic cancer profiling using circulating tumor DNA has enabled the detection of National Comprehensive Cancer Network (NCCN) guideline-recommended somatic alterations from a single, non-invasive blood draw. However, reliably detecting somatic variants at low variant allele fractions (VAFs) remains a challenge for next-generation sequencing (NGS)-based tests. We have developed the single-molecule sequencing (SMSEQ) platform to address these challenges. METHODS The OncoLBx assay utilizes the SMSEQ platform to optimize cell-free DNA extraction and library preparation with variant type-specific calling algorithms to improve sensitivity and specificity. OncoLBx is a pan-cancer panel for solid tumors targeting 75 genes and five microsatellite sites analyzing five classes of NCCN-recommended somatic variants: single-nucleotide variants (SNVs), insertions and deletions (indels), copy number variants (CNVs), fusions and microsatellite instability (MSI). Circulating DNA was extracted from plasma, followed by library preparation using SMSEQ. Analytical validation was performed according to recently published American College of Medical Genetics and Genomics (ACMG)/Association for Molecular Pathology (AMP) guidelines and established the limit of detection (LOD), sensitivity, specificity, accuracy and reproducibility using 126 gold-standard reference samples, healthy donor samples verified by whole-exome sequencing by an external College of American Pathologists (CAP) reference lab and cell lines with known variants. Results were analyzed using a locus-specific modeling algorithm. RESULTS We have demonstrated that OncoLBx detects VAFs of ≥ 0.1% for SNVs and indels, ≥ 0.5% for fusions, ≥ 4.5 copies for CNVs and ≥ 2% for MSI, with all variant types having specificity ≥ 99.999%. Diagnostic performance of paired samples displays 80% sensitivity and > 99.999% clinical specificity. Clinical utility and performance were assessed in 416 solid tumor samples. Variants were detected in 79% of samples, for which 87.34% of positive samples had available targeted therapy.
Collapse
|
8
|
Castillo P, Marginet M, Jares P, García M, Gonzalvo E, Arance A, García A, Alos L, Teixido C. Implementation of an NGS panel for clinical practice in paraffin-embedded tissue samples from locally advanced and metastatic melanoma patients. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:101-108. [PMID: 36046072 PMCID: PMC9400780 DOI: 10.37349/etat.2020.00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
Aim: Single biomarker diagnostic test of BRAFV600 locus in metastatic melanoma is mandatory for treatment decision; however, multiple-gene based techniques, such as targeted next-generation sequencing (NGS) are being used to maximize the number of patients that can benefit from a targeted therapy. The main objective of this study is to investigate whether an NGS panel could be adopted in routine clinical care for advanced melanoma. Methods: Patients diagnosed with advanced melanoma at our center from 2017 to 2019 were included. Presence of genetic alterations was performed using two methods: real-time polymerase chain reaction-based Idylla test (Biocartis) and NGS with the oncomine solid tumor DNA kit (Thermo Fisher Scientific). Total genomic DNA was extracted from formalin-fixed and paraffin embedded samples for sequencing. Results: A total of 155 samples were evaluated for molecular analysis but 40 samples (25.8%) were inadequate for sequencing. The clinical utility of BRAFV600 real-time polymerase chain reaction and targeted-NGS was compared in 29 samples and a very good concordance was observed (Kappa = 0.89, 95% confidence interval 0.68 ± 1.05). An oncogenic mutation by NGS was found in 75 samples (65%)–53% of whom were candidates for personalized therapies. The most prevalent mutated genes were BRAF (39%), TP53 (23%), and NRAS (14%). Other genes identified at lower incidence (< 5%) were: PIK3CA, ERBB4, CTNNB1, STK11, FGFR1, SMAD4, KRAS, FGFR3, PTEN and AKT. Co-occurrence of oncogenic mutations was detected in 40% of the samples. Among the mutations identified, TP53 was significantly more prevalent in men (men 31.8% versus women 12.2%, P = 0.03) and NRAS in women (men 9.1% versus women 24.4%, P = 0.03). Conclusions: Targeted-NGS testing is a feasible technique to implement in the routine clinical practice. Based on our results, NGS has provided more information on target-genes than RT-PCR technique, maximizing the benefit for patients with advanced melanoma.
Collapse
Affiliation(s)
- Paola Castillo
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Marta Marginet
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Pedro Jares
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Mireia García
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Elena Gonzalvo
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Adriana García
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Llucia Alos
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Cristina Teixido
- Department of Pathology, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
9
|
Kim SY, Kim TW. Current challenges in the implementation of precision oncology for the management of metastatic colorectal cancer. ESMO Open 2020; 5:e000634. [PMID: 32188714 PMCID: PMC7078672 DOI: 10.1136/esmoopen-2019-000634] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/28/2019] [Accepted: 01/02/2020] [Indexed: 12/21/2022] Open
Abstract
Over the last few decades, molecularly targeted agents have been used for the treatment of metastatic colorectal cancer. They have made remarkable contributions to prolonging the lives of patients. The emergence of several biomarkers and their introduction to the clinic have also aided in guiding such treatment. Recently, next-generation sequencing (NGS) has enabled clinicians to identify these biomarkers more easily and reliably. However, there is considerable uncertainty in interpreting and implementing the vast amount of information from NGS. The clinical relevance of biomarkers other than NGS are also subjects of debate. This review covers controversial issues and recent findings on such therapeutics and their molecular targets, including VEGF, EGFR, BRAF, HER2, RAS, actionable fusions, Wnt pathway and microsatellite instability for comprehensive understanding of obstacles on the road to precision oncology in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Songpa-gu, Seoul, Republic of Korea
| | - Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Songpa-gu, Seoul, Republic of Korea
| |
Collapse
|
10
|
Cope NJ, Novak B, Liu Z, Cavallo M, Gunderwala AY, Connolly M, Wang Z. Analyses of the oncogenic BRAF D594G variant reveal a kinase-independent function of BRAF in activating MAPK signaling. J Biol Chem 2020; 295:2407-2420. [PMID: 31929109 DOI: 10.1074/jbc.ra119.011536] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/07/2020] [Indexed: 12/19/2022] Open
Abstract
Class 3 mutations in B-Raf proto-oncogene, Ser/Thr kinase (BRAF), that result in kinase-impaired or kinase-dead BRAF have the highest mutation frequency in BRAF gene in lung adenocarcinoma. Several studies have reported that kinase-dead BRAF variants amplify mitogen-activated protein kinase (MAPK) signaling by dimerizing with and activating WT C-Raf proto-oncogene, Ser/Thr kinase (CRAF). However, the structural and functional principles underlying their activation remain elusive. Herein, using cell biology and various biochemical approaches, we established that variant BRAFD594G, a kinase-dead representative of class 3 mutation-derived BRAF variants, has a higher dimerization potential as compared with WT BRAF. Molecular dynamics simulations uncovered that the D594G substitution orients the αC-helix toward the IN position and extends the activation loop within the kinase domain, shifting the equilibrium toward the active, dimeric conformation, thus priming BRAFD594G as an effective allosteric activator of CRAF. We found that B/CRAF heterodimers are the most thermodynamically stable RAF dimers, suggesting that RAF heterodimers, and not homodimers, are the major players in determining the amplitude of MAPK signaling in cells. Additionally, we show that BRAFD594G:CRAF heterodimers bypass autoinhibitory P-loop phosphorylation, which might contribute to longer duration of MAPK pathway signaling in cancer cells. Last, we propose that the dimer interface of the BRAFD594G:CRAF heterodimer may represent a promising target in the design of novel anticancer therapeutics.
Collapse
Affiliation(s)
- Nicholas J Cope
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Borna Novak
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Zhiwei Liu
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Maria Cavallo
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Amber Y Gunderwala
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Matthew Connolly
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104
| | - Zhihong Wang
- Department of Chemistry & Biochemistry, University of the Sciences, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
11
|
Liu X, Zhong D. [Research Progress of Immune Checkpoint Inhibitor Therapy for BRAF Mutation
in Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:583-589. [PMID: 31526463 PMCID: PMC6754571 DOI: 10.3779/j.issn.1009-3419.2019.09.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
在非小细胞肺癌(non-small cell lung cancer, NSCLC)患者中,约2%-4%有BRAF基因突变,该型肿瘤恶性程度高、化疗有效率低、预后差。尽管BRAF抑制剂及MEK抑制剂联合治疗在BRAF V600E突变的晚期NSCLC患者中成效显著,已被写入美国国家综合癌症网络(National Comprehensive Cancer Network, NCCN)指南,但两药联合副作用发生率高,耐药之后尚无有效治疗策略,且针对非V600E突变患者仍缺乏靶向治疗方案。本文将针对BRAF突变型NSCLC免疫标志物表达情况以及免疫检查点抑制剂(immune checkpoint inhibitor, ICI)疗效相关研究做一综述,为延长患者生存提供更多选择方案。
Collapse
Affiliation(s)
- Xia Liu
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Diansheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
12
|
Cornejo KM, Cosar EF, Paner GP, Yang P, Tomaszewicz K, Meng X, Mehta V, Sirintrapun SJ, Barkan GA, Hutchinson L. Mutational Profile Using Next-Generation Sequencing May Aid in the Diagnosis and Treatment of Urachal Adenocarcinoma. Int J Surg Pathol 2019; 28:51-59. [PMID: 31496327 DOI: 10.1177/1066896919872535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Objectives. The rare urachal adenocarcinoma (UAC) of the bladder has striking morphologic and immunohistochemical overlap with colorectal adenocarcinoma (CAC) and bladder adenocarcinoma (BAC). To date, the mutational status in UAC and BAC has not been well investigated. Methods. We retrospectively evaluated 34 UACs (mucinous, n = 9; intestinal, n = 3; signet ring cell, n = 1; not otherwise specified, n = 21) and 4 BACs (n = 4). Next-generation sequencing analysis of 50 cancer "hotspot" gene mutations using the Ampliseq Cancer Hotspot Panel v2 was performed. Two UAC cases did not have adequate DNA quality with poor sequencing coverage and were excluded from the study. Results. RAS mutations were identified in 16 of 32 (50%) UACs (15 KRAS; 1 NRAS) and none of the BACs (0%). TP53 mutations were found in both UACs (18/32; 56%) and BACs (4/4; 100%). GNAS (n = 4), SMAD4 (n = 3), and BRAF (n = 1) mutations were only found in UACs. In contrast, APC (n = 2) mutations were only found in BACs. The mucinous subtype of UAC contained a SMAD4 mutation in 33% of cases (3/9), which was not identified in any other subtype (0/23; 0%) (P = .0169). The only BRAF mutation was identified in the single signet ring cell subtype of UAC. There were no other differences in the mutation profile when comparing histologic subtypes of UAC. Conclusions. In summary, UAC and BAC have overlapping but distinct mutation profiles and these differences may aid in separating these 2 entities. Next-generation sequencing to identify therapeutic targets or resistance markers may aid treatment decisions.
Collapse
Affiliation(s)
- Kristine M Cornejo
- University of Massachusetts Medical School, UMass Memorial Medical Center, Worcester, MA, USA
| | - Ediz F Cosar
- University of Massachusetts Medical School, UMass Memorial Medical Center, Worcester, MA, USA
| | | | - Ping Yang
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Keith Tomaszewicz
- University of Massachusetts Medical School, UMass Memorial Medical Center, Worcester, MA, USA
| | - Xiuling Meng
- University of Massachusetts Medical School, UMass Memorial Medical Center, Worcester, MA, USA
| | - Vikas Mehta
- Mount Sinai Hospital Medical Center, Chicago, IL, USA
| | | | | | - Lloyd Hutchinson
- University of Massachusetts Medical School, UMass Memorial Medical Center, Worcester, MA, USA
| |
Collapse
|
13
|
|
14
|
Lin Q, Zhang H, Ding H, Qian J, Lizaso A, Lin J, Han-Zhang H, Xiang J, Li Y, Zhu H. The association between BRAF mutation class and clinical features in BRAF-mutant Chinese non-small cell lung cancer patients. J Transl Med 2019; 17:298. [PMID: 31470866 PMCID: PMC6716889 DOI: 10.1186/s12967-019-2036-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/18/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND BRAF mutations occur in 2-4% non-small cell lung cancer (NSCLC) patients and can be categorized into three functional classes based on signaling mechanism and kinase activity: RAS-independent kinase-activating V600 monomers (class 1), RAS-independent kinase-activating dimers (class 2) and RAS-dependent kinase-inactivating heterodimers (class 3). The association between functional classes and clinical features in Chinese NSCLC patients remains unexplored. Our multi-center study aimed to survey the BRAF mutation rate and analyze the associated clinical features in this population. METHODS Capture-based sequencing data of either plasma or tissue samples obtained from 8405 Chinese stage I-IV NSCLC patients were retrospectively analyzed. RESULTS BRAF mutations were detected in 238 patients, revealing an overall mutation rate of 2.8%. Among them, 32%, 21% and 13% had BRAF mutant class 1, 2 and 3 respectively. The remaining 34% had other BRAF mutations. V600 (32%) and G469 (13%) were the two most predominant BRAF mutations. Patients with class 2 and 3 mutations were more likely to have concurrent KRAS mutations (P = 0.001). Collectively, BRAF mutations, including non-class 1-3 mutations, were more likely to occur in males (P < 0.01). However, females were more likely to harbor class 1 mutations (P < 0.02). We also compared the overall survival (OS) of first-line chemotherapy-treated advanced-stage patients and revealed comparable OS among the three groups. CONCLUSION Our study revealed a 2.8% BRAF mutation rate in Chinese NSCLC patients. Our data also showed a male predominance when all BRAF mutations were considered collectively, and a female predominance for class 1 mutations. Furthermore, BRAF V600E is less likely to have concurrent KRAS mutations comparing to the other two classes.
Collapse
Affiliation(s)
- Quan Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Campus, Ouhai District, Wenzhou, 325015 Zhejiang China
| | - Haoran Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000 Anhui China
| | - Huaxin Ding
- Ningbo Diagnostic Pathology Center, Ningbo, 315000 Zhejiang China
| | - Jun Qian
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001 Jiangsu China
| | - Analyn Lizaso
- Burning Rock Biotech, Guangzhou, 510300 Guangdong China
| | - Jing Lin
- Burning Rock Biotech, Guangzhou, 510300 Guangdong China
| | - Han Han-Zhang
- Burning Rock Biotech, Guangzhou, 510300 Guangdong China
| | | | - Yuping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Campus, Ouhai District, Wenzhou, 325015 Zhejiang China
| | - Hong Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Gusu District, Suzhou, 215006 Jiangsu China
| |
Collapse
|
15
|
Nassereddine H, Sannier A, Brosseau S, Rodier JM, Khalil A, Msika S, Danel C, Couvelard A, Théou-Anton N, Cazes A. Clinicopathological and Molecular Study of Peritoneal Carcinomatosis Associated with Non-Small Cell Lung Carcinoma. Pathol Oncol Res 2019; 26:2795-2800. [PMID: 31407221 DOI: 10.1007/s12253-019-00713-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/06/2019] [Indexed: 11/24/2022]
Abstract
To retrospectively characterize the molecular features of Non-Small Cell Lung Carcinomas (NSCLC) with peritoneal carcinomatosis (PC), clinicopathological data of 12 patients diagnosed with NSCLC and PC between 2007 and 2016 were collected. Immunohistochemistry and Next Generation Sequencing (NGS) were performed on cases with available material. PC was the initial presentation of NSCLC in 17% of the cases. Overall, patients with PC displayed a poor median survival of 12 weeks. Histology was adenocarcinoma in 11 cases. 37.5% of cases showed PD-L1 immunostaining positivity (50% cut-off). ALK and ROS1 immunostainings were negative. Using NGS, we identified 17 molecular alterations in 9 genes (TP53, KRAS, STK11, BRAF, EGFR, DDR2, ERBB4, SMAD4, CTNNB1) in 88.9% of adenocarcinomas. To the best of our knowledge, 5 of these variants are not referenced in the literature. In conclusion, PC might be the initial presentation of NSCLC. Molecular profiling of our cases did not find any effective targetable alteration, except from high PD-L1 expression.
Collapse
Affiliation(s)
- Hussein Nassereddine
- Assistance Publique - Hôpitaux de Paris, département de pathologie, Hôpital Bichat-Claude Bernard, Paris, France. .,Department of pathology, AP-HP, Bichat- Hospital, 46 rue Henri Huchard 75877, 18, Paris Cedex, France.
| | - Aurélie Sannier
- Assistance Publique - Hôpitaux de Paris, département de pathologie, Hôpital Bichat-Claude Bernard, Paris, France.,Université de Paris, Paris, France
| | - Solenn Brosseau
- Université de Paris, Paris, France.,APHP, service d'Oncologie Thoracique, Hôpital Bichat-Claude Bernard, Paris, France
| | | | - Antoine Khalil
- Université de Paris, Paris, France.,APHP, service de Radiologie, Hôpital Bichat-Claude Bernard, Paris, France
| | - Simon Msika
- Université de Paris, Paris, France.,APHP, service de Chirurgie Générale et Digestive, Hôpital Bichat-Claude Bernard, Paris, France
| | - Claire Danel
- Assistance Publique - Hôpitaux de Paris, département de pathologie, Hôpital Bichat-Claude Bernard, Paris, France.,Université de Paris, Paris, France
| | - Anne Couvelard
- Assistance Publique - Hôpitaux de Paris, département de pathologie, Hôpital Bichat-Claude Bernard, Paris, France.,Université de Paris, Paris, France
| | | | - Aurélie Cazes
- Assistance Publique - Hôpitaux de Paris, département de pathologie, Hôpital Bichat-Claude Bernard, Paris, France.,Université de Paris, Paris, France
| |
Collapse
|
16
|
Man RJ, Zhang YL, Jiang AQ, Zhu HL. A patent review of RAF kinase inhibitors (2010–2018). Expert Opin Ther Pat 2019; 29:675-688. [DOI: 10.1080/13543776.2019.1651842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ruo-Jun Man
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, People’s Republic of China
- College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, People’s Republic of China
| | - Ya-Liang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, People’s Republic of China
| | - Ai-Qin Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, People’s Republic of China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
17
|
Lokhandwala PM, Tseng LH, Rodriguez E, Zheng G, Pallavajjalla A, Gocke CD, Eshleman JR, Lin MT. Clinical mutational profiling and categorization of BRAF mutations in melanomas using next generation sequencing. BMC Cancer 2019; 19:665. [PMID: 31277584 PMCID: PMC6612071 DOI: 10.1186/s12885-019-5864-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022] Open
Abstract
Background Analysis of melanomas for actionable mutations has become the standard of care. Recently, a classification scheme has been proposed that categorizes BRAF mutations based on their mechanisms for activation of the MAPK pathway. Methods In this analysis BRAF, KIT, NRAS, and PIK3CA mutations were examined by next generation sequencing (NGS) in 446 melanomas in a clinical diagnostic setting. KRAS and HRAS were also analyzed to elucidate coexisting BRAF and RAS mutations. BRAF mutations were categorized into class-1 (kinase-activated, codon 600), class-2 (kinase-activated, non-codon 600) and class-3 (kinase-impaired), based on the newly proposed classification scheme. Results NGS demonstrated high analytic sensitivity. Among 355 mutations detected, variant allele frequencies were 2–5% in 21 (5.9%) mutations and 2–10% in 47 (13%) mutations. Mutations were detected in BRAF (42%), NRAS (25%), KIT (4.9%) and PIK3CA (2.7%). The incidence of class-1, class-2 and class-3 mutations were 33% (26% p.V600E and 6.1% p.V600K), 3.1 and 4.9% respectively. With a broader reportable range of NGS, class-1, class-2 and class-3 mutations accounted for 77, 7.4 and 12% of all BRAF mutations. Class-3 mutations, commonly affecting codons 594, 466 and 467, showed a higher incidence of coexisting RAS mutations, consistent with their RAS-dependent signaling. Significant association with old age and primary tumors of head/neck/upper back suggest chronic solar damage as a contributing factor for melanomas harboring BRAF p.V600K or class-3 mutations. Conclusion This study categorizes the range, frequency, coexisting driver mutations and clinical characteristics of the three classes of BRAF mutations in a large cohort of melanomas in a clinical diagnostic setting. Further prospective studies are warranted to elucidate the clinical outcomes and benefits of newly developed targeted therapy in melanoma patients carrying each class of BRAF mutation. Electronic supplementary material The online version of this article (10.1186/s12885-019-5864-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Parvez M Lokhandwala
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA.
| | - Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA.,Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Erika Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA
| | - Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA
| | - Aparna Pallavajjalla
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA
| | - Christopher D Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University School of Medicine, 1812 Ashland Ave, Suite 200, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
El‐Deiry WS, Goldberg RM, Lenz H, Shields AF, Gibney GT, Tan AR, Brown J, Eisenberg B, Heath EI, Phuphanich S, Kim E, Brenner AJ, Marshall JL. The current state of molecular testing in the treatment of patients with solid tumors, 2019. CA Cancer J Clin 2019; 69:305-343. [PMID: 31116423 PMCID: PMC6767457 DOI: 10.3322/caac.21560] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The world of molecular profiling has undergone revolutionary changes over the last few years as knowledge, technology, and even standard clinical practice have evolved. Broad molecular profiling is now nearly essential for all patients with metastatic solid tumors. New agents have been approved based on molecular testing instead of tumor site of origin. Molecular profiling methodologies have likewise changed such that tests that were performed on patients a few years ago are no longer complete and possibly inaccurate today. As with all rapid change, medical providers can quickly fall behind or struggle to find up-to-date sources to ensure he or she provides optimum care. In this review, the authors provide the current state of the art for molecular profiling/precision medicine, practice standards, and a view into the future ahead.
Collapse
Affiliation(s)
- Wafik S. El‐Deiry
- Associate Dean for Oncologic Sciences, Warren Alpert Medical School; Director, Joint Program in Cancer Biology, Brown University and the Lifespan Cancer Institute; Professor of Pathology & Laboratory Medicine and Professor of Medical ScienceBrown UniversityProvidenceRI
| | - Richard M. Goldberg
- Professor of Medicine and DirectorWest Virginia University Cancer InstituteMorgantownWV
| | - Heinz‐Josef Lenz
- Professor of Medicine, Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCA
| | | | - Geoffrey T. Gibney
- Associate Professor of Medicine, Co‐Leader of the Melanoma Disease GroupLombardi Comprehensive Cancer Institute, MedStar Georgetown Cancer InstituteWashingtonDC
| | - Antoinette R. Tan
- Co‐Director of Phase I Program, Department of Solid Tumor Oncology and Investigational TherapeuticsLevine Cancer Institute, Atrium HealthCharlotteNC
| | - Jubilee Brown
- Professor and Associate Director of Gynecologic OncologyLevine Cancer Institute, Atrium HealthCharlotteNC
| | - Burton Eisenberg
- Professor of Clinical SurgeryUniversity of Southern CaliforniaLos AngelesCA
- Executive Medical DirectorHoag Family Cancer InstituteNewport BeachCA
| | | | - Surasak Phuphanich
- Professor of Neurology, Director, Division of Neuro‐OncologyBarrow Neurological InstitutePhoenixAZ
| | - Edward Kim
- Chair, Solid Tumor Oncology and Investigational TherapeuticsLevine Cancer Institute, Atrium HealthCharlotteNC
| | - Andrew J. Brenner
- Associate Professor of Medicine, Mays Cancer Center at University of Texas Health San Antonio Cancer CenterSan AntonioTX
| | - John L. Marshall
- Professor of Medicine and Oncology, Director, Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer InstituteMedStar Georgetown Cancer InstituteWashingtonDC
| |
Collapse
|
19
|
Smith KN, Llosa NJ, Cottrell TR, Siegel N, Fan H, Suri P, Chan HY, Guo H, Oke T, Awan AH, Verde F, Danilova L, Anagnostou V, Tam AJ, Luber BS, Bartlett BR, Aulakh LK, Sidhom JW, Zhu Q, Sears CL, Cope L, Sharfman WH, Thompson ED, Riemer J, Marrone KA, Naidoo J, Velculescu VE, Forde PM, Vogelstein B, Kinzler KW, Papadopoulos N, Durham JN, Wang H, Le DT, Justesen S, Taube JM, Diaz LA, Brahmer JR, Pardoll DM, Anders RA, Housseau F. Persistent mutant oncogene specific T cells in two patients benefitting from anti-PD-1. J Immunother Cancer 2019; 7:40. [PMID: 30744692 PMCID: PMC6371497 DOI: 10.1186/s40425-018-0492-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
Background Several predictive biomarkers are currently approved or are under investigation for the selection of patients for checkpoint blockade. Tumor PD-L1 expression is used for stratification of non-small cell lung (NSCLC) patients, with tumor mutational burden (TMB) also being explored with promising results, and mismatch-repair deficiency is approved for tumor site-agnostic disease. While tumors with high PD-L1 expression, high TMB, or mismatch repair deficiency respond well to checkpoint blockade, tumors with lower PD-L1 expression, lower mutational burdens, or mismatch repair proficiency respond much less frequently. Case presentation We studied two patients with unexpected responses to checkpoint blockade monotherapy: a patient with PD-L1-negative and low mutational burden NSCLC and one with mismatch repair proficient colorectal cancer (CRC), both of whom lack the biomarkers associated with response to checkpoint blockade, yet achieved durable clinical benefit. Both maintained T-cell responses in peripheral blood to oncogenic driver mutations – BRAF-N581I in the NSCLC and AKT1-E17K in the CRC – years after treatment initiation. Mutation-specific T cells were also found in the primary tumor and underwent dynamic perturbations in the periphery upon treatment. Conclusions These findings suggest that T cell responses to oncogenic driver mutations may be more prevalent than previously appreciated and could be harnessed in immunotherapeutic treatment, particularly for patients who lack the traditional biomarkers associated with response. Comprehensive studies are warranted to further delineate additional predictive biomarkers and populations of patients who may benefit from checkpoint blockade. Electronic supplementary material The online version of this article (10.1186/s40425-018-0492-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kellie N Smith
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Nicolas J Llosa
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Tricia R Cottrell
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas Siegel
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Hongni Fan
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Prerna Suri
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Hok Yee Chan
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Haidan Guo
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Teniola Oke
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Anas H Awan
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Franco Verde
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Ludmila Danilova
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Division of Biostatistics and Bioinformatics, Johns Hopkins University, Baltimore, MD, USA
| | - Valsamo Anagnostou
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Ada J Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Brandon S Luber
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Division of Biostatistics and Bioinformatics, Johns Hopkins University, Baltimore, MD, USA
| | - Bjarne R Bartlett
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,The Swim Across America Laboratory, John Hopkins University, Baltimore, MD, USA.,Ludwig Center and Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA.,Present address: B.R.B.,Bioinformatics Core, Department of Complementary & Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA
| | - Laveet K Aulakh
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,The Swim Across America Laboratory, John Hopkins University, Baltimore, MD, USA.,Ludwig Center and Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA
| | - John-William Sidhom
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Qingfeng Zhu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Cynthia L Sears
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Leslie Cope
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Division of Biostatistics and Bioinformatics, Johns Hopkins University, Baltimore, MD, USA
| | - William H Sharfman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth D Thompson
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,The Swim Across America Laboratory, John Hopkins University, Baltimore, MD, USA
| | - Joanne Riemer
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen A Marrone
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Jarushka Naidoo
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Victor E Velculescu
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Patrick M Forde
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Bert Vogelstein
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Ludwig Center and Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kenneth W Kinzler
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Ludwig Center and Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Nickolas Papadopoulos
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Ludwig Center and Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer N Durham
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Hao Wang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Division of Biostatistics and Bioinformatics, Johns Hopkins University, Baltimore, MD, USA
| | - Dung T Le
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | | | - Janis M Taube
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Luis A Diaz
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,The Swim Across America Laboratory, John Hopkins University, Baltimore, MD, USA.,Ludwig Center and Howard Hughes Medical Institute, Johns Hopkins University, Baltimore, MD, USA.,Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julie R Brahmer
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Drew M Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Robert A Anders
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Franck Housseau
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
20
|
Chae YK, Tamragouri KB, Chung J, Lin X, Miller V, Ali SM, Giles FJ. Large-Cell Neuroendocrine Carcinoma of the Lung: A Focused Analysis of BRAF Alterations and Case Report of a BRAF Non-V600-Mutated Tumor Responding to Targeted Therapy. JCO Precis Oncol 2018; 2:1-12. [PMID: 35135105 DOI: 10.1200/po.17.00150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE In advanced stages, large-cell neuroendocrine carcinoma of the lung (L-LCNEC) mimics small-cell lung cancer despite its traditional classification as a non-small-cell lung cancer. Here we present a focused analysis of BRAF mutations in this population. PATIENTS AND METHODS Comprehensive genomic profiling of tumor tissues was performed from a cohort of 300 patients with biopsy-proven L-LCNEC. Specimens were either from a primary lung lesion or metastatic site. RESULTS In 13 patients, 14 unique BRAF alterations (amplifications, mutations) were identified. The importance of biomarker-driven therapy is subsequently highlighted with our case of a 69-year-old man diagnosed with metastatic L-LCNEC who did not respond to cisplatin plus etoposide. A significant durable response was then demonstrated with therapy targeted toward a BRAF non-V600E activating mutation (G469R) associated with biomarker response identified through circulating cell-free tumor DNA analysis. A change in clonal allele frequency from nearly 40% to nondetectable was observed. CONCLUSION Although uncommon, L-LCNEC does seem to contain activating and therefore actionable alterations. We thus highlight the value of pursuing next-generation sequencing for patients with this disease.
Collapse
Affiliation(s)
- Young Kwang Chae
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Keerthi B Tamragouri
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Jon Chung
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Xiaoqi Lin
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Vincent Miller
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Siraj M Ali
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| | - Francis J Giles
- Young Kwang Chae, Keerthi B. Tamragouri, and Francis J. Giles, Northwestern University Feinberg School of Medicine; Young Kwang Chae and Francis J. Giles, Robert H. Lurie Comprehensive Cancer Center, Northwestern University; Xiaoqi Lin, Northwestern Memorial Hospital, Northwestern University, Chicago, IL; and Jon Chung, Vincent Miller, and Siraj M. Ali, Foundation Medicine, Cambridge, MA
| |
Collapse
|
21
|
De Marchi F, Haley L, Fryer H, Ibrahim J, Beierl K, Zheng G, Gocke CD, Eshleman JR, Belchis D, Illei P, Lin MT. Clinical Validation of Coexisting Activating Mutations Within EGFR, Mitogen-Activated Protein Kinase, and Phosphatidylinositol 3-Kinase Pathways in Lung Cancers. Arch Pathol Lab Med 2018; 143:174-182. [PMID: 30485130 DOI: 10.5858/arpa.2017-0495-oa] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Mutations within the same signature transduction pathway are redundant and, therefore, most are mutually exclusive. Laboratory errors, however, may introduce unexpected coexisting mutations. OBJECTIVE.— To validate coexisting mutations within epidermal growth factor receptor (EGFR), mitogen-activated protein kinase, and phosphatidylinositol 3-kinase pathways. DESIGN.— In this retrospective study for quality assessment of next-generation sequencing in a clinical diagnostics setting, coexisting mutations within EGFR, KRAS, NRAS, BRAF, AKT1, and PIK3CA genes were examined in 1208 non-small cell lung cancers. RESULTS.— EGFR mutations did not coexist with BRAF mutations, neither kinase-activated nor kinase-impaired mutations. There was a low but similar incidence (3.3%-5.1%) of PIK3CA mutations in BRAF-, EGFR-, and KRAS-mutated lung cancers and a rare incidence of coexisting KRAS and EGFR mutations detected in 1 of 1208 lung cancers (0.08%) or 1 of 226 EGFR-mutated lung cancers (0.4%). Coexisting BRAF p.V600E mutation was observed in 3 of 4 AKT1 p.E17K-mutated lung cancers. Mutational profiling of DNA reisolated from subareas with the same or different histomorphology, using an alternative assay, confirmed that coexisting mutations might present within the same (whole or subclonal) population or different populations and clarified that the so-called coexisting activating KRAS and BRAF mutations originally reported in a specimen were indeed present in separate lung nodules submitted in the same block. CONCLUSIONS.— The results supported that EGFR and BRAF mutations are early driver mutations in lung cancers. Guidelines from official organizations to establish standard operating procedures are warranted to validate unexpected coexisting mutations and, if clinically indicated, to determine their presence in the same or different tumor populations.
Collapse
Affiliation(s)
- Federico De Marchi
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Lisa Haley
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Henderson Fryer
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Junaid Ibrahim
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Katie Beierl
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Gang Zheng
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Christopher D Gocke
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - James R Eshleman
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Deborah Belchis
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Peter Illei
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Ming-Tseh Lin
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| |
Collapse
|
22
|
Chat-Uthai N, Vejvisithsakul P, Udommethaporn S, Meesiri P, Danthanawanit C, Wongchai Y, Teerapakpinyo C, Shuangshoti S, Poungvarin N. Development of ultra-short PCR assay to reveal BRAF V600 mutation status in Thai colorectal cancer tissues. PLoS One 2018; 13:e0198795. [PMID: 29879227 PMCID: PMC5991739 DOI: 10.1371/journal.pone.0198795] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/25/2018] [Indexed: 02/06/2023] Open
Abstract
The protein kinase BRAF is one of the key players in regulating cellular responses to extracellular signals. Somatic mutations of the BRAF gene, causing constitutive activation of BRAF, have been found in various types of human cancers such as malignant melanoma, and colorectal cancer. BRAF V600E and V600K, most commonly observed mutations in these cancers, may predict response to targeted therapies. Many techniques suffer from a lack of diagnostic sensitivity in mutation analysis in clinical samples with a low cancer cell percentage or poor-quality fragmented DNA. Here we present allele-specific real-time PCR assay for amplifying 35- to 45-base target sequences in BRAF gene. Forward primer designed for BRAF V600E detection is capable of recognizing both types of BRAF V600E mutation, i.e. V600E1 (c.1799T>A) and V600E2 (c.1799_1800delTGinsAA), as well as complex tandem mutation caused by nucleotide changes in codons 600 and 601. We utilized this assay to analyze Thai formalin-fixed paraffin-embedded tissues. Forty-eight percent of 178 Thai colorectal cancer tissues has KRAS mutation detected by highly sensitive commercial assays. Although these DNA samples contain low overall yield of amplifiable DNA, our newly-developed assay successfully revealed BRAF V600 mutations in 6 of 93 formalin-fixed paraffin-embedded colorectal cancer tissues which KRAS mutation was not detected. Ultra-short PCR assay with forward mutation-specific primers is potentially useful to detect BRAF V600 mutations in highly fragmented DNA specimens from cancer patients.
Collapse
Affiliation(s)
- Nunthawut Chat-Uthai
- Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Sutthirat Udommethaporn
- Clinical Molecular Pathology Laboratory, Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Puttarakun Meesiri
- Clinical Molecular Pathology Laboratory, Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chetiya Danthanawanit
- Clinical Molecular Pathology Laboratory, Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yannawan Wongchai
- Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chinachote Teerapakpinyo
- Chulalongkorn GenePRO Center, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Shanop Shuangshoti
- Chulalongkorn GenePRO Center, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Naravat Poungvarin
- Clinical Molecular Pathology Laboratory, Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
23
|
Molnár E, Rittler D, Baranyi M, Grusch M, Berger W, Döme B, Tóvári J, Aigner C, Tímár J, Garay T, Hegedűs B. Pan-RAF and MEK vertical inhibition enhances therapeutic response in non-V600 BRAF mutant cells. BMC Cancer 2018; 18:542. [PMID: 29739364 PMCID: PMC5941622 DOI: 10.1186/s12885-018-4455-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Currently, there are no available targeted therapy options for non-V600 BRAF mutated tumors. The aim of this study was to investigate the effects of RAF and MEK concurrent inhibition on tumor growth, migration, signaling and apoptosis induction in preclinical models of non-V600 BRAF mutant tumor cell lines. METHODS Six BRAF mutated human tumor cell lines CRL5885 (G466 V), WM3629 (D594G), WM3670 (G469E), MDAMB231 (G464 V), CRL5922 (L597 V) and A375 (V600E as control) were investigated. Pan-RAF inhibitor (sorafenib or AZ628) and MEK inhibitor (selumetinib) or their combination were used in in vitro viability, video microscopy, immunoblot, cell cycle and TUNEL assays. The in vivo effects of the drugs were assessed in an orthotopic NSG mouse breast cancer model. RESULTS All cell lines showed a significant growth inhibition with synergism in the sorafenib/AZ628 and selumetinib combination. Combination treatment resulted in higher Erk1/2 inhibition and in increased induction of apoptosis when compared to single agent treatments. However, single selumetinib treatment could cause adverse therapeutic effects, like increased cell migration in certain cells, selumetinib and sorafenib combination treatment lowered migratory capacity in all the cell lines. Importantly, combination resulted in significantly increased tumor growth inhibition in orthotropic xenografts of MDAMB231 cells when compared to sorafenib - but not to selumetinib - treatment. CONCLUSIONS Our data suggests that combined blocking of RAF and MEK may achieve increased therapeutic response in non-V600 BRAF mutant tumors.
Collapse
Affiliation(s)
- Eszter Molnár
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary
| | - Dominika Rittler
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary
| | - Marcell Baranyi
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary
| | - Michael Grusch
- Institute of Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Balázs Döme
- Department of Thoracic Surgery, Medical University of Vienna, 1090, Vienna, Austria.,National Korányi Institute of TB and Pulmonology, Budapest, 1085, Hungary.,Department of Thoracic Surgery, Semmelweis University-National Institute of Oncology, Budapest, 1122, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, 1122, Hungary
| | - Clemens Aigner
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, 45239, Essen, Germany
| | - József Tímár
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary.,HAS-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary
| | - Tamás Garay
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary.,HAS-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary.,HAS Postdoctoral Fellowship Program Hungarian Academy of Sciences, Budapest, 1051, Hungary
| | - Balázs Hegedűs
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary. .,Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, 45239, Essen, Germany. .,HAS-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary.
| |
Collapse
|
24
|
Mufti M, Ching S, Farjami S, Shahangian S, Sobnosky S. A Case Series of Two Patients Presenting With Pericardial Effusion as First Manifestation of Non-Small Cell Lung Cancer With BRAF Mutation and Expression Of PD-L1. World J Oncol 2018; 9:56-61. [PMID: 29760834 PMCID: PMC5942209 DOI: 10.14740/wjon1092w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the number one cause of cancer-related deaths in the United States. Involvement of pericardium occurs once cancer has progressed to stage IV which can cause massive effusion in the pericardial sac. This can lead to cardiac tamponade which can be fatal very quickly if untreated. The following is a two patient case series in which both patients presented with large pericardial effusion. The first patient sought medical attention due to new onset palpitations and was found to have hemorrhagic pericardial effusion and pulmonary embolism (PE). The second patient presented with shortness of breath. Investigations revealed that she had pericardial and pleural effusions along with multiple metastases throughout the body. Both patients ended up with a diagnosis of non-small cell lung cancer (NSCLC) with BRAF mutation. One patient had V600E mutation; other patient had a variant p.D594N mutation. Both patients also had expression of PD-L1.
Collapse
Affiliation(s)
- Muhammad Mufti
- Department of Medicine, St. Mary Medical Center, Long Beach, CA, USA
| | - Steven Ching
- Department of Medicine, St. Mary Medical Center, Long Beach, CA, USA
| | - Sassan Farjami
- Department of Hematology/Oncology, St. Mary Medical Center, Long Beach, CA, USA.,Pacific Shores Medical Group, Long Beach, CA, USA
| | | | - Serap Sobnosky
- Department of Cardiology, St. Mary Medical Center, Long Beach, CA, USA
| |
Collapse
|
25
|
Inamura K. Clinicopathological Characteristics and Mutations Driving Development of Early Lung Adenocarcinoma: Tumor Initiation and Progression. Int J Mol Sci 2018; 19:ijms19041259. [PMID: 29690599 PMCID: PMC5979290 DOI: 10.3390/ijms19041259] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with lung adenocarcinoma representing the most common lung cancer subtype. Among all lung adenocarcinomas, the most prevalent subset develops via tumorigenesis and progression from atypical adenomatous hyperplasia (AAH) to adenocarcinoma in situ (AIS), to minimally invasive adenocarcinoma (MIA), to overt invasive adenocarcinoma with a lepidic pattern. This stepwise development is supported by the clinicopathological and molecular characteristics of these tumors. In the 2015 World Health Organization classification, AAH and AIS are both defined as preinvasive lesions, whereas MIA is identified as an early invasive adenocarcinoma that is not expected to recur if removed completely. Recent studies have examined the molecular features of lung adenocarcinoma tumorigenesis and progression. EGFR-mutated adenocarcinoma frequently develops via the multistep progression. Oncogene-induced senescence appears to decrease the frequency of the multistep progression in KRAS- or BRAF-mutated adenocarcinoma, whose tumor evolution may be associated with epigenetic alterations and kinase-inactive mutations. This review summarizes the current knowledge of tumorigenesis and tumor progression in early lung adenocarcinoma, with special focus on its clinicopathological characteristics and their associations with driver mutations (EGFR, KRAS, and BRAF) as well as on its molecular pathogenesis and progression.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
26
|
Type II RAF inhibitor causes superior ERK pathway suppression compared to type I RAF inhibitor in cells expressing different BRAF mutant types recurrently found in lung cancer. Oncotarget 2018; 9:16110-16123. [PMID: 29662630 PMCID: PMC5882321 DOI: 10.18632/oncotarget.24576] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 02/20/2018] [Indexed: 12/19/2022] Open
Abstract
A large fraction of somatic driver BRAF mutations in lung cancer are non-V600 and impaired-kinase. Non-V600 BRAF mutations predict sensitivity to combination of a type I RAF inhibitor, Dabrafenib, and a MEK inhibitor, Trametinib. Singly, Dabrafenib only weakly suppresses mutant BRAF-induced ERK signaling and can induce ERK paradoxical activation in CRAF-overexpressing cells. The present study compared the effects of Dabrafenib and a type II RAF inhibitor, AZ628, on ERK activity in HEK293T cells expressing several tumor-derived BRAF mutants, and in a non-V600 and impaired-kinase BRAF-mutant lung cancer cell line (H1666). Unlike Dabrafenib, AZ628 did not induce paradoxical ERK activation in CRAF-overexpressing cells and BRAF-mutant cells overexpressing CRAF were more responsive to AZ628 compared to Dabrafenib in terms of ERK inhibition. AZ628 inhibited ERK more effectively than Dabrafenib in both H1666 cells and HEK293T cells co-expressing several different BRAF-mutants with CRAF. Similarly, AZ628 plus Trametinib had better MEK-inhibitory and pro-apoptotic effects in H1666 cells than Dabrafenib plus Trametinib. Moreover, prolonged treatment of H1666 cells with AZ628 plus Trametinib produced greater inhibition of cell growth than Dabrafenib plus Trametinib. These results indicate that AZ628 has greater potential than Dabrafenib, both as a single agent and combined with Trametinib, for the treatment of non-V600 BRAF mutant lung cancer.
Collapse
|
27
|
Heterogeneity of resistance mutations detectable by nextgeneration sequencing in TKI-treated lung adenocarcinoma. Oncotarget 2018; 7:45237-45248. [PMID: 27304188 PMCID: PMC5216719 DOI: 10.18632/oncotarget.9931] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022] Open
Abstract
EGFR-mutated lung adenocarcinomas routinely develop resistance to tyrosine kinase inhibitors (TKI). To better characterize the relative frequencies of the resistance mechanisms, we analyzed 48 EGFR-mutated TKI-resistant specimens from 41 patients. Next-generation sequencing of post-treatment specimens detected EGFR p.T790M in 31 (79%) of 39 patients, PIK3CA mutations in 10 (26%), EGFR p.S768_V769delinsIL in one, and KRAS p.G12C in one. Five PIK3CA mutations were outside of codons 542, 545, and 1047. Three of four pre-treatment specimens did not carry the PIK3CA mutation found in the post-treatment sample. Small cell carcinoma transformation was identified in four patients; none had p.T790M, including two where p.T790M was identified in the co-existing adenocarcinoma. In p.T790M-mutated specimens, the allele frequency was less than 5% in 24% of cases. p.T790M allele frequency was usually lower than that of the sensitizing mutation indicating that the resistance mutation was present either in a subset of cells or, if the sensitizing mutation was amplified, in a subset of the sensitizing alleles of a dominant clone. Eight patients had multiple resistance mutations, suggesting either multiple separate resistant clones or a single clone harboring multiple resistance mechanisms. PIK3CA mutations appear to be a more significant resistance mechanism than previously recognized.
Collapse
|
28
|
Kotani H, Adachi Y, Kitai H, Tomida S, Bando H, Faber AC, Yoshino T, Voon DC, Yano S, Ebi H. Distinct dependencies on receptor tyrosine kinases in the regulation of MAPK signaling between BRAF V600E and non-V600E mutant lung cancers. Oncogene 2018; 37:1775-1787. [DOI: 10.1038/s41388-017-0035-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/27/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
|
29
|
Yang Y, Meng Y, Zhang H, Shen X, Li R, Yu L, Liu B, Wang L. Detection of EGFR and BRAF mutations by competitive allele-specific TaqMan polymerase chain reaction in lung adenocarcinoma. Oncol Lett 2017; 15:3295-3304. [PMID: 29467863 DOI: 10.3892/ol.2017.7652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/24/2017] [Indexed: 11/05/2022] Open
Abstract
Epithelial growth factor receptor (EGFR)-tyrosine kinase inhibitors are the standard first-line treatment for patients with metastatic non-small cell lung cancer (NSCLC) expressing sensitive EGFR-mutants. Other drugs target different driver mutants, including the serine/threonine-protein kinase B-raf (BRAF) inhibitor dabrafenib, which has exhibited promising efficacy for treating patients with metastatic BRAF-mutated NSCLC. Therefore, identifying patients carrying mutations that may be treated using targeted therapies is important. However, the methods of molecular detection presently applied in clinical practice, particularly detection of BRAF in NSCLC patients, require further investigation. Therefore, more sensitive and economic methods are required. The present study applied the competitive allele-specific TaqMan polymerase chain reaction (CastPCR) technology to the molecular detection of EGFR (del2235-2249, del2236-2250, T790M, L858R) and BRAF (V600E, G469A, D594G) mutations in 144 treatment-naive patients with lung adenocarcinoma, and analyzed the association between the mutation rates and patients' clinicopathological features. 51.4% (74/144) cases were identified harboring EGFR mutations. A total of 40.3% (58/144) patients carried sensitizing mutations (exon 19 deletion or L858R) and 14.6% (21/144) carried T790M mutations. 6.9% (10/144) mutation-positive patients were double-mutated. Total EGFR mutation rate was significantly increased in female compared with that of males (60.9 vs. 43.8%, P<0.05), in non-smokers compared with that of smokers (62.8 vs. 34.5%, P<0.05). In total, 8.3% (12/144) patients were identified with BRAF mutations. 16.7% were V600E (2/12) and 83.3% (10/12) were non-V600E mutants. Among the 10 non-V600E mutations, D594G accounted for 90.0% (9/10) and G469A accounted for 10.0% (1/10). Statistical analysis demonstrated that the BRAF mutation rate was not associated with any of the following clinicopathological features: Sex, age, smoking history, clinical stages, distant metastasis, differentiation degree, tumor size and regional lymph node metastasis (P≥0.05). CastPCR technology is a robust method with high sensitivity for the molecular detection of EGFR and BRAF mutations in clinical formalin-fixed paraffin-embedded samples.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oncology, The Affiliated Taikang Xianlin Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Yi Meng
- Department of Oncology, The Affiliated Taikang Xianlin Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Hang Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaoyan Shen
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Rutian Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Lifeng Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
30
|
Nieto P, Ambrogio C, Esteban-Burgos L, Gómez-López G, Blasco MT, Yao Z, Marais R, Rosen N, Chiarle R, Pisano DG, Barbacid M, Santamaría D. A Braf kinase-inactive mutant induces lung adenocarcinoma. Nature 2017; 548:239-243. [PMID: 28783725 PMCID: PMC5648056 DOI: 10.1038/nature23297] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 06/16/2017] [Indexed: 12/31/2022]
Abstract
The initiating oncogenic event in almost half of human lung adenocarcinomas is still unknown, a fact that complicates the development of selective targeted therapies. Yet these tumours harbour a number of alterations without obvious oncogenic function including BRAF-inactivating mutations. Inactivating BRAF mutants in lung predominate over the activating V600E mutant that is frequently observed in other tumour types. Here we demonstrate that the expression of an endogenous Braf(D631A) kinase-inactive isoform in mice (corresponding to the human BRAF(D594A) mutation) triggers lung adenocarcinoma in vivo, indicating that BRAF-inactivating mutations are initiating events in lung oncogenesis. Moreover, inactivating BRAF mutations have also been identified in a subset of KRAS-driven human lung tumours. Co-expression of Kras(G12V) and Braf(D631A) in mouse lung cells markedly enhances tumour initiation, a phenomenon mediated by Craf kinase activity, and effectively accelerates tumour progression when activated in advanced lung adenocarcinomas. We also report a key role for the wild-type Braf kinase in sustaining Kras(G12V)/Braf(D631A)-driven tumours. Ablation of the wild-type Braf allele prevents the development of lung adenocarcinoma by inducing a further increase in MAPK signalling that results in oncogenic toxicity; this effect can be abolished by pharmacological inhibition of Mek to restore tumour growth. However, the loss of wild-type Braf also induces transdifferentiation of club cells, which leads to the rapid development of lethal intrabronchiolar lesions. These observations indicate that the signal intensity of the MAPK pathway is a critical determinant not only in tumour development, but also in dictating the nature of the cancer-initiating cell and ultimately the resulting tumour phenotype.
Collapse
Affiliation(s)
- Patricia Nieto
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Chiara Ambrogio
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Laura Esteban-Burgos
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - María Teresa Blasco
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Zhan Yao
- Program in Molecular Pharmacology, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, M20 4BX Manchester, UK
| | - Neal Rosen
- Program in Molecular Pharmacology, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - Roberto Chiarle
- Department of Pathology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David G Pisano
- Bioinformatics Unit, Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Mariano Barbacid
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - David Santamaría
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| |
Collapse
|
31
|
Yang Y, Shen X, Li R, Shen J, Zhang H, Yu L, Liu B, Wang L. The detection and significance of EGFR and BRAF in cell-free DNA of peripheral blood in NSCLC. Oncotarget 2017; 8:49773-49782. [PMID: 28572536 PMCID: PMC5564806 DOI: 10.18632/oncotarget.17937] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 05/04/2017] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE Although driver mutation status is crucial to targeted therapy decision-making in non-small cell lung cancer (NSCLC), due to unavailable or inadequate biopsies, there are still many patients with unknown mutation status. A promising way to solve this problem is liquid biopsy, such as cell-free DNA (cfDNA) in peripheral blood. Additionally, due to the little amount of cfDNA, detecting methods with high sensitivity, specificity and economy are required in clinical practice. Here, we explored the feasibility of Competitive Allele-Specific TaqMan® PCR (CastPCR) detecting driver mutations in cfDNA from plasma in lung adenocarcinoma patients. RESULTS Sensitivity, specificity, concordance, PPV and NPV of CastPCR detecting EGFR mutations in cfDNA was 56.4% (31/55), 94.2% (49/52), 74.8% (80/107), 91.2% (31/34) and 67.1% (49/73), respectively. Notably, specificity and PPV for p.T790M both reached 100.0%. For BRAF detection, it was 28.6% (2/7), 93.0% (93/100), 88.8% (95/107), 22.2% (2/9) and 94.9% (93/98), respectively. MATERIALS AND METHODS Plasma specimens of 107 lung adenocarcinoma patients and their matched tumor formalin fixed paraffin embedded (FFPE) samples were analyzed. CastPCR was used to detect EGFR (c.2235_2249del, c.2236_2250del, c.2369C>T p.T790M, c.2573T>G p.L858R) and BRAF (c.1406G>C p.G469A, c.1799T>A p.V600E, c.1781A>G p.D594G) mutations. Mutation results of tumor tissue was set as gold standard, and the sensitivity, specificity, concordance, positive predictive value (PPV) and negative predictive value (NPV) were calculated for each mutation. CONCLUSIONS For patients whose tumor tissue is unavailable or inadequate, EGFR mutation detection in cfDNA with CastPCR could be first choice. Mutation positive results may provide reference for further clinical medication. While negative results indicate that detection in tissue should be considered as the following step. In this way, tumor tissue could be economized to the maximum extent and the risk of repeated percutaneous transthoracic lung biopsy could also be lowered to the maximum extent. For BRAF detection in cfDNA, CastPCR is a specific method while the sensitivity needs further exploration.
Collapse
Affiliation(s)
- Yang Yang
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
- Nanjing Xianlin Drum Tower Hospital, Nanjing 210046, PR China
| | - Xiaoyan Shen
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Rutian Li
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Jie Shen
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Hang Zhang
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Lixia Yu
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| | - Lifeng Wang
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, PR China
| |
Collapse
|
32
|
Lehmann-Che J, Poirot B, Boyer JC, Evrard A. La génétique somatique des tumeurs solides, un incontournable à l’ère de la médecine de précision. Therapie 2017; 72:217-230. [DOI: 10.1016/j.therap.2016.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 10/20/2022]
|
33
|
Buchanan DD, Clendenning M, Rosty C, Eriksen SV, Walsh MD, Walters RJ, Thibodeau SN, Stewart J, Preston S, Win AK, Flander L, Ouakrim DA, Macrae FA, Boussioutas A, Winship IM, Giles GG, Hopper JL, Southey MC, English D, Jenkins MA. Tumor testing to identify lynch syndrome in two Australian colorectal cancer cohorts. J Gastroenterol Hepatol 2017; 32:427-438. [PMID: 27273229 PMCID: PMC5140773 DOI: 10.1111/jgh.13468] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Tumor testing of colorectal cancers (CRC) for mismatch repair (MMR) deficiency is an effective approach to identify carriers of germline MMR gene mutation (Lynch syndrome). The aim of this study was to identify MMR gene mutation carriers in two cohorts of population-based CRC utilizing a combination of tumor and germline testing approaches. METHODS Colorectal cancers from 813 patients diagnosed with CRC < 60 years of age from the Australasian Colorectal Cancer Family Registry (ACCFR) and from 826 patients from the Melbourne Collaborative Cohort Study (MCCS) were tested for MMR protein expression using immunohistochemistry, microsatellite instability (MSI), BRAFV600E somatic mutation, and for MLH1 methylation. MMR gene mutation testing (Sanger sequencing and Multiplex Ligation Dependent Probe Amplification) was performed on germline DNA of patients with MMR-deficient tumors and a subset of MMR-proficient CRCs. RESULTS Of the 813 ACCFR probands, 90 probands demonstrated tumor MMR deficiency (11.1%), and 42 had a MMR gene germline mutation (5.2%). For the MCCS, MMR deficiency was identified in the tumors of 103 probands (12.5%) and seven had a germline mutation (0.8%). All the mutation carriers were diagnosed prior to 70 years of age. Probands with a MMR-deficient CRC without MLH1 methylation and a gene mutation were considered Lynch-like and comprised 41.1% and 25.2% of the MMR-deficient CRCs for the ACCFR and MCCS, respectively. CONCLUSIONS Identification of MMR gene mutation carriers in Australian CRC-affected patients is optimized by immunohistochemistry screening of CRC diagnosed before 70 years of age. A significant proportion of MMR-deficient CRCs will have unknown etiology (Lynch-like) proving problematic for clinical management.
Collapse
Affiliation(s)
- Daniel D Buchanan
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark Clendenning
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
- Envoi Specialist Pathologists, Herston, Queensland, Australia
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Stine V Eriksen
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael D Walsh
- Department of Histopathology, Sullivan Nicolaides Pathology, Brisbane, Queensland, Australia
| | - Rhiannon J Walters
- Cancer and Population Studies Group, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | - Stephen N Thibodeau
- Molecular Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jenna Stewart
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan Preston
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Aung Ko Win
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Louisa Flander
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Driss Ait Ouakrim
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Finlay A Macrae
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Genetic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Australia
- Colorectal Medicine and Genetics, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Alex Boussioutas
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Cancer Genomics and Predictive Medicine, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ingrid M Winship
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
- Genetic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Australia
| | - Graham G Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
- Department of Epidemiology and Institute of Health and Environment, School of Public Health, Seoul National University, Seoul, Korea
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Dallas English
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
34
|
Lehmann-Che J, Poirot B, Boyer JC, Evrard A. Cancer genomics guide clinical practice in personalized medicine. Therapie 2017; 72:439-451. [PMID: 28258721 DOI: 10.1016/j.therap.2016.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/02/2016] [Indexed: 01/04/2023]
Abstract
Targeted therapies have revolutionized the treatment of many cancers. Widely developed over the last decade, this new concept of precision medicine relies on the use of genomic technologies to analyze tumor samples in order to identify actionable targets and biomarkers of resistance. The goal is to optimize treatment by identifying which therapeutic approach is best for each patient, i.e. the treatment that is effective, has minimal adverse effects, and avoids unnecessary intervention and cost. The purpose of this review is to highlight, using a few seminal examples of therapeutic targets, the important contribution of appropriate analysis of key oncogenes or driver genes in making clinical decisions. Cancer genomics is now an indispensable part of clinical management. Furthermore, the development of next generation sequencing (NGS) will enable exploration of more and more genes of interest, leading to new treatment options for personalized medicine.
Collapse
Affiliation(s)
- Jacqueline Lehmann-Che
- Laboratoire d'oncologie moléculaire, hôpital Saint-Louis, 1, avenue Claude-Vellefaux 75475 Paris cedex 10, France; Unité CNRS UMR7212/U944, équipe de recherche translationnelle en oncologie, bâtiment Jean-Bernard, 75475 Paris, France.
| | - Brigitte Poirot
- Laboratoire d'oncologie moléculaire, hôpital Saint-Louis, 1, avenue Claude-Vellefaux 75475 Paris cedex 10, France; Unité CNRS UMR7212/U944, équipe de recherche translationnelle en oncologie, bâtiment Jean-Bernard, 75475 Paris, France
| | - Jean-Christophe Boyer
- Laboratoire de biochimie, CHU de Nîmes Carémeau, 30029 Nîmes, France; EA 2415, « Aide à la décision médicale personnalisée : aspects méthodologiques » IURC, faculté de médecine de Montpellier, 34093 Montpellier, France
| | - Alexandre Evrard
- Laboratoire de biochimie, CHU de Nîmes Carémeau, 30029 Nîmes, France; Unité Inserm U1194, Institut de recherche en cancérologie de Montpellier (IRCM), 34298 Montpellier, France
| |
Collapse
|
35
|
Lin EI, Tseng LH, Gocke CD, Reil S, Le DT, Azad NS, Eshleman JR. Mutational profiling of colorectal cancers with microsatellite instability. Oncotarget 2016; 6:42334-44. [PMID: 26517354 PMCID: PMC4747229 DOI: 10.18632/oncotarget.5997] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/05/2015] [Indexed: 01/11/2023] Open
Abstract
Microsatellite instability (MSI) is caused by defective mismatch repair in 15–20% of colorectal cancers (CRCs). Higher mutation loads in tumors with mismatch repair deficiency can predict response to pembrolizumab, an anti-programmed death 1 (PD-1) immune checkpoint inhibitor. We analyzed the mutations in 113 CRCs without MSI (MSS) and 29 CRCs with MSI-High (MSI-H) using the 50-gene AmpliSeq cancer panel. Overall, MSI-H CRCs showed significantly higher mutations than MSS CRCs, including insertion/deletion mutations at repeat regions. MSI-H CRCs showed higher incidences of mutations in the BRAF, PIK3CA, and PTEN genes as well as mutations in the receptor tyrosine kinase families. While the increased mutations in BRAF and PTEN in MSI-H CRCs are well accepted, we also support findings of mutations in the mTOR pathway and receptor tyrosine kinase family genes. MSS CRCs showed higher incidences of mutations in the APC, KRAS and TP53 genes, confirming previous findings. NGS assays may be designed to detect driver mutations for targeted therapeutics and to identify tumors with high mutation loads for potential treatment with immune checkpoint blockade therapies. Further studies may be warranted to elucidate potential targeted therapeutics against mutations in the mTOR pathway and the receptor tyrosine kinase family in MSI-H CRCs as well as the benefit of anti-PD-1 immunotherapy in hypermutated MSS CRCs or other cancers.
Collapse
Affiliation(s)
- Elaine I Lin
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Li-Hui Tseng
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Christopher D Gocke
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stacy Reil
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dung T Le
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nilofer S Azad
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James R Eshleman
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Molecular Diagnostics for Precision Medicine in Colorectal Cancer: Current Status and Future Perspective. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9850690. [PMID: 27699178 PMCID: PMC5028795 DOI: 10.1155/2016/9850690] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/10/2016] [Indexed: 02/08/2023]
Abstract
Precision medicine, a concept that has recently emerged and has been widely discussed, emphasizes tailoring medical care to individuals largely based on information acquired from molecular diagnostic testing. As a vital aspect of precision cancer medicine, targeted therapy has been proven to be efficacious and less toxic for cancer treatment. Colorectal cancer (CRC) is one of the most common cancers and among the leading causes for cancer related deaths in the United States and worldwide. By far, CRC has been one of the most successful examples in the field of precision cancer medicine, applying molecular tests to guide targeted therapy. In this review, we summarize the current guidelines for anti-EGFR therapy, revisit the roles of pathologists in an era of precision cancer medicine, demonstrate the transition from traditional “one test-one drug” assays to multiplex assays, especially by using next-generation sequencing platforms in the clinical diagnostic laboratories, and discuss the future perspectives of tumor heterogeneity associated with anti-EGFR resistance and immune checkpoint blockage therapy in CRC.
Collapse
|
37
|
Rodriguez FJ, Vizcaino MA, Lin MT. Recent Advances on the Molecular Pathology of Glial Neoplasms in Children and Adults. J Mol Diagn 2016; 18:620-634. [PMID: 27444975 PMCID: PMC5397677 DOI: 10.1016/j.jmoldx.2016.05.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/06/2016] [Accepted: 05/11/2016] [Indexed: 02/02/2023] Open
Abstract
Gliomas represent the most common primary intraparenchymal tumors of the central nervous system in adults and children and are a genetic and phenotypic heterogeneous group. Large multi-institutional studies and The Cancer Genome Atlas have provided firm insights into the basic genetic drivers in gliomas. The main molecular biomarkers routinely applied to evaluate diffuse gliomas include MGMT promoter methylation, EGFR alterations (eg, EGFRvIII), IDH1 or IDH2 mutations, and 1p19q co-deletion. Many of these markers have become standard of care for molecular testing and prerequisites for clinical trial enrollment. Other recent biomarkers include TERT promoter and ATRX mutations, alterations that identify specific molecular subgroups of diffuse gliomas with biological and clinical relevance. It has also become apparent that distinctive patterns of molecular genetic evolution develop in the context of current therapeutic regimens. Important insights have also been uncovered in the field of pediatric glioma, including the identification of recurrent mutation, fusion, and/or duplication events of the BRAF, FGFR1, MYB, and MYBL1 genes in pediatric low-grade gliomas, mutations affecting histone components (H3F3A p.K27M or p.G34) in pediatric high-grade gliomas, and aggressive subsets developing in midline central nervous system structures. Here, we summarize current concepts in molecular testing for glial tumors, including recent findings by large-scale discovery efforts and technologic advances that are affecting routine diagnostic work.
Collapse
Affiliation(s)
- Fausto J Rodriguez
- Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - M Adelita Vizcaino
- Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Cellular and Tissue Biology, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Ming-Tseh Lin
- Division of Molecular Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
38
|
Tafe LJ, Pierce KJ, Peterson JD, de Abreu F, Memoli VA, Black CC, Pettus JR, Marotti JD, Gutmann EJ, Liu X, Shirai K, Dragnev KH, Amos CI, Tsongalis GJ. Clinical Genotyping of Non-Small Cell Lung Cancers Using Targeted Next-Generation Sequencing: Utility of Identifying Rare and Co-mutations in Oncogenic Driver Genes. Neoplasia 2016; 18:577-83. [PMID: 27659017 PMCID: PMC5031899 DOI: 10.1016/j.neo.2016.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 11/15/2022]
Abstract
Detection of somatic mutations in non-small cell lung cancers (NSCLCs), especially adenocarcinomas, is important for directing patient care when targeted therapy is available. Here, we present our experience with genotyping NSCLC using the Ion Torrent Personal Genome Machine (PGM) and the AmpliSeq Cancer Hotspot Panel v2. We tested 453 NSCLC samples from 407 individual patients using the 50 gene AmpliSeq Cancer Hotspot Panel v2 from May 2013 to July 2015. Using 10 ng of DNA, up to 11 samples were simultaneously sequenced on the Ion Torrent PGM (316 and 318 chips). We identified variants with the Ion Torrent Variant Caller Plugin, and Golden Helix's SVS software was used for annotation and prediction of the significance of the variants. Three hundred ninety-eight samples were successfully sequenced (12.1% failure rate). In all, 633 variants in 41 genes were detected with a median of 2 (range of 0 to 7) variants per sample. Mutations detected in BRAF, EGFR, ERBB2, KRAS, NRAS, and PIK3CA were considered potentially actionable and were identified in 237 samples, most commonly in KRAS (37.9%), EGFR (11.1%), BRAF (4.8%), and PIK3CA (4.3%). In our patient population, all mutations in EGFR, KRAS, and BRAF were mutually exclusive. The Ion Torrent Ampliseq technology can be utilized on small biopsy and cytology specimens, requires very little input DNA, and can be applied in clinical laboratories for genotyping of NSCLC. This targeted next-generation sequencing approach allows for detection of common and also rare mutations that are clinically actionable in multiple patients simultaneously.
Collapse
Affiliation(s)
- Laura J Tafe
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH.
| | - Kirsten J Pierce
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jason D Peterson
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Francine de Abreu
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Vincent A Memoli
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Candice C Black
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jason R Pettus
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Edward J Gutmann
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Xiaoying Liu
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Keisuke Shirai
- Department of Oncology, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Konstantin H Dragnev
- Department of Oncology, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Christopher I Amos
- Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH; Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH and Norris Cotton Cancer Center and Dartmouth-Hitchcock Medical Center, Lebanon, NH
| |
Collapse
|
39
|
Matos P, Gonçalves V, Jordan P. Targeting the serrated pathway of colorectal cancer with mutation in BRAF. Biochim Biophys Acta Rev Cancer 2016; 1866:51-63. [DOI: 10.1016/j.bbcan.2016.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/15/2016] [Accepted: 06/19/2016] [Indexed: 12/19/2022]
|
40
|
Zheng G, Tsai H, Tseng LH, Illei P, Gocke CD, Eshleman JR, Netto G, Lin MT. Test Feasibility of Next-Generation Sequencing Assays in Clinical Mutation Detection of Small Biopsy and Fine Needle Aspiration Specimens. Am J Clin Pathol 2016; 145:696-702. [PMID: 27247373 DOI: 10.1093/ajcp/aqw043] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES To evaluate preanalytic factors contributing to failure of next-generation sequencing (NGS) assays. METHODS AmpliSeq Cancer Hotspot Panel was conducted in 1,121 of 1,152 formalin-fixed paraffin-embedded tissues submitted to a clinical laboratory, including 493 small biopsy or fine needle aspiration (FNA) specimens (44%) and 25 metastatic bone specimens (2.2%). RESULTS Single nucleotide mutations and/or insertion/deletion mutations were detected in 702 specimens. Thirty-eight specimens (3.4%) were reported as "no results" due to NGS assay failure. Higher failure rates were observed in specimens submitted for lung cancer panel and melanoma panel (3.1% and 3.7% vs 1.0% colorectal cancer panel), metastatic bone specimens (36% vs 2.6% nonbone specimens), referred specimens (5.0% vs 1.8% in-house specimens), and small biopsy and FNA specimens (5.8% and 3.1% vs 0.7% resection/excision specimens). Test feasibility was higher in in-house specimens than referred specimens (99.1% vs 96.9% in resection specimens, 94.4% vs 87.3% in small biopsy specimens, and 94.3% vs 58.8% in FNA specimens). CONCLUSIONS NGS assays demonstrated clinical utility in solid tumor specimens, including those taken by biopsy or FNA. Preanalytic factors identified by this study that may contribute to NGS assay failure highlight the need for pathologists to revisit tissue processing protocols in order to better optimize cancer mutational profiling.
Collapse
Affiliation(s)
| | | | - Li-Hui Tseng
- From the Departments of Pathology and Department of Medical Genetics, National Taiwan University Hospital, Taipei
| | | | - Christopher D Gocke
- From the Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - James R Eshleman
- From the Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | |
Collapse
|
41
|
Clinical detection and categorization of uncommon and concomitant mutations involving BRAF. BMC Cancer 2015; 15:779. [PMID: 26498038 PMCID: PMC4619530 DOI: 10.1186/s12885-015-1811-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/16/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Selective BRAF inhibitors, vemurafenib and dabrafenib, and the MEK inhibitor, trametinib, have been approved for treatment of metastatic melanomas with a BRAF p.V600E mutation. The clinical significance of non-codon 600 mutations remains unclear, in part, due to variation of kinase activity for different mutants. METHODS In this study, we categorized BRAF mutations according to the reported mutant kinase activity. A total of 1027 lung cancer, colorectal cancer or melanoma specimens were submitted for clinical mutation detection by next generation sequencing. RESULTS Non-codon 600 mutations were observed in 37% of BRAF-mutated tumors. Of all BRAF mutants, 75% were kinase-activated, 15% kinase-impaired and 10% kinase-unknown. The most common kinase-impaired mutant involves codon 594, specifically, p.D594G (c.1781A > G) and p.D594N (c.1780G > A). Lung cancers showed significantly higher incidences of kinase-impaired or kinase-unknown mutants. Kinase-impaired BRAF mutants showed a significant association with concomitant activating KRAS or NRAS mutations, but not PIK3CA mutations, supporting the reported interaction of these mutations. CONCLUSIONS BRAF mutants with impaired or unknown kinase activity as well as concomitant kinase-impaired BRAF mutations and RAS mutations were detected in lung cancers, colorectal cancers and melanomas. Different therapeutic strategies based on the BRAF mutant kinase activity and the concomitant mutations may be worthwhile.
Collapse
|