1
|
Khan OM, Mallardo D, Decock J. Editorial: Cancer genomics in the era of precision medicine. Front Genet 2024; 15:1378917. [PMID: 38435061 PMCID: PMC10904538 DOI: 10.3389/fgene.2024.1378917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/08/2024] [Indexed: 03/05/2024] Open
Affiliation(s)
- Omar M. Khan
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Domenico Mallardo
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics INT IRCCS Fondazione “G Pascale”, Napoli, Italy
| | - Julie Decock
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
2
|
Cooper L, Chen J. Changes in Companion Diagnostic Labelling: Implementation of FDA's April 2020 Guidance for Industry for In Vitro CDx Labeling for Specific Oncology Therapeutic Groups. Ther Innov Regul Sci 2022; 56:689-697. [PMID: 35689144 PMCID: PMC9356945 DOI: 10.1007/s43441-022-00422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/19/2022] [Indexed: 11/26/2022]
Abstract
Advanced understanding of the molecular pathways of oncologic diseases has shifted therapeutic treatment development to focus on mechanism of actions targeting specific genomic alterations. These precision medicines are indicated for patient subsets defined by these specific mutations as determined by diagnostic devices approved by the Food and Drug Administration (FDA). The Intended Use section within the companion diagnostic (CDx) labeling has historically specified the therapeutic products for which they have been clinically validated. In April 2020, the FDA reiterated their position that therapeutic class labeling may be used, if appropriate, instead of named products. Labels for FDA approved in vitro CDxs were reviewed to evaluate the implementation of therapeutic class labeling. A total of 47 devices have been approved as of 2 January 2022, of which 3 labels were found to contain therapeutic class labeling: two devices targeting EGFR mutations for the treatment of non-small cell lung cancer (NSCLC), and one targeting BRAF V600E and BRAF/MEK inhibitor combinations for melanoma. Two devices received therapeutic class labeling upon initial approval, while the third implemented the language though a label revision. A total of 25 different indications were identified across the 47 CDx devices, of which 9 (34.6%) were associated with more than 1 CDx device. Implementation of therapeutic class labeling has been slow following the release of the FDA’s April 2020 guidance; however, the potential to incorporate such language into existing and newly approved CDx labels exists. Precedence and manufacturer experience are expected to drive an increase in therapeutic class labeling.
Collapse
Affiliation(s)
- Lisa Cooper
- Department of Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Stanley S. Bergen Building, Suite 136, 65 Bergen Street, Newark, NJ, 07101-1709, USA.
| | - Joyce Chen
- Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
3
|
Ouellette SB. Landscape of granted US patents in personalized diagnostics for oncology from 2014 to 2018. Expert Opin Ther Pat 2019; 29:191-198. [PMID: 30712415 DOI: 10.1080/13543776.2019.1575809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Personalized diagnostic testing (PDx) is a key component of the precision medicine toolkit and has shown the most development in cancer applications. Recent changes in the regulatory and legal landscapes regarding PDx development and commercialization have brought uncertainties to both intellectual property strategies and business model development. While the regulatory and legal uncertainties have been well-documented, there has been little reported analysis of the recent patent landscape and movement of IP into the PDx market. Areas covered: This article provides a snapshot landscape analysis of cancer-associated PDx US granted patents from 2014 to 2018, with a focus on claim types, biomarkers, and associated detection strategies, and assignee-specific IP portfolio analyses. Expert opinion: Patent-driven research is commonplace in the legal world for performing patentability, clearance, and validity analyses. The results from this review show that patent-driven analysis is also insightful for understanding strategies to build IP portfolios around biomarker and detection platforms, identifying partners and competitors, and driving PDx technologies into the market. This information is an important source of business intelligence and can provide companies or investors with valuable information for making strategic decisions in developing and commercializing PDx technologies.
Collapse
Affiliation(s)
- Steven B Ouellette
- a Biotechnology & Pharmaceuticals Group , Global Prior Art, Inc , Boston , MA , USA
| |
Collapse
|
4
|
Pierobon M, Wulfkuhle J, Liotta LA, Petricoin Iii EF. Utilization of Proteomic Technologies for Precision Oncology Applications. Cancer Treat Res 2019; 178:171-187. [PMID: 31209845 DOI: 10.1007/978-3-030-16391-4_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomic analysis of tumor specimens has revealed that cancer is fundamentally a proteomic disease at the functional level: driven by genomically defined derangements, but selected for in the proteins that are encoded and the aberrant activation of signaling and biochemical networks. This activation is measured by posttranslational modifications such as phosphorylation and other modifications that modulate cellular signaling, and these events cannot be effectively measured by genomic analysis alone. Moreover, these signaling networks by and large represent the targets for many FDA-approved and experimental molecularly targeted therapeutics. Consequently, it is important that we consider new classification schemas for oncology based not on tumor site of origin or histology under the microscope but on the functional protein signaling architecture. There are numerous proteomic technologies that could be discussed from a purely technological standpoint, but this chapter will concentrate on an overview of the main proteomic technologies available for conducting protein pathway activation analysis of clinical specimens such as multiplex immunoassays, phospho-specific flow cytometry, reverse phase protein microarrays, quantitative immunohistochemistry, and mass spectrometry. This chapter will focus on the application of these technologies to cancer-based clinical studies evaluating prognostic/predictive markers or for stratifying patients to personalized treatments.
Collapse
Affiliation(s)
- Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 20110, Manassas, VA, USA
| | - Julie Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 20110, Manassas, VA, USA
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 20110, Manassas, VA, USA
| | - Emanuel F Petricoin Iii
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 20110, Manassas, VA, USA.
| |
Collapse
|
5
|
Sherkow JS, Abbott R. Fortune and hindsight: gene patents' muted effect on medical practice. Br Med Bull 2018; 126:37-45. [PMID: 29608656 DOI: 10.1093/bmb/ldy009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 03/08/2018] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Physicians have long worried about gene patents' potential to restrict their medical practices. Fortune and hindsight have proven these worries exaggerated both in the UK and elsewhere. Neither current nor future medical practices appear to be impinged by gene patents, although they may be subject to future intellectual property disputes. SOURCES OF DATA Qualitative and quantitative (survey) studies of gene patents' effects on medical practice; recent developments in patent law. AREAS OF AGREEMENT Traditional gene patents do not appear to have restricted medical practice in the UK, although their effect elsewhere has been more nuanced. AREAS OF CONTROVERSY Whether patents will restrict the spread of newer medical technologies is unresolved. AREAS TIMELY FOR DEVELOPING RESEARCH Continuing survey data on practitioners' views concerning patents' role in the distribution of newer technologies would be beneficial.
Collapse
Affiliation(s)
- Jacob S Sherkow
- Innovation Center for Law and Technology, New York Law School, 185 West Broadway, New York, NY, USA.,Health Policy and Management, Columbia University Mailman School of Public Health, 722 W. 168th St., New York, NY, USA.,Center for Advanced Studies in Biomedical Innovation Law, University of Copenhagen Faculty of Law, Karen Blixens Plads 16, Copenhagen S, Denmark
| | - Ryan Abbott
- Law and Health Sciences, University of Surrey School of Law, Faculty of Law, Guildford, Surrey, UK.,David Geffen School of Medicine at University of California, 10833 Le Conte Ave, Los Angeles, CA, USA
| |
Collapse
|
6
|
Godoy-Gijón E, Yuste-Chaves M, Santos-Briz Á. BRAF Mutation Status Concordance Between Primary Cutaneous Melanomas and Corresponding Metastases: A Review of the Latest Evidence. ACTAS DERMO-SIFILIOGRAFICAS 2017; 108:894-901. [PMID: 28711165 DOI: 10.1016/j.ad.2016.12.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/06/2016] [Accepted: 12/31/2016] [Indexed: 11/22/2022] Open
Abstract
The identification of B-Raf proto-oncongene (BRAF) mutation and the emergence of targeted therapy marked a turning point in the treatment of melanoma. The study of mutation status concordance between primary tumors and metastases in this cancer has major treatment implications as it facilitates the selection of candidates for targeted therapy. This review analyzes the evidence on the level of mutation status concordance between primary tumors and different types of metastases in cutaneous melanoma and provides an overview of the advantages and disadvantages of the various methods used to detect BRAF mutations.
Collapse
Affiliation(s)
- E Godoy-Gijón
- Servicio de Dermatología, Complejo Asistencial de Zamora, Zamora, España.
| | - M Yuste-Chaves
- Servicio de Dermatología, Complejo Asistencial Universitario de Salamanca, Salamanca, España
| | - Á Santos-Briz
- Servicio de Anatomía Patológica, Complejo Asistencial Universitario de Salamanca, Salamanca, España
| |
Collapse
|
7
|
Godoy-Gijón E, Yuste-Chaves M, Santos-Briz Á. BRAF Mutation Status Concordance Between Primary Cutaneous Melanomas and Corresponding Metastases: A Review of the Latest Evidence. ACTAS DERMO-SIFILIOGRAFICAS 2017. [DOI: 10.1016/j.adengl.2017.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
8
|
Caldwell C, Johnson CE, Balaji VN, Balaji GA, Hammer RD, Kannan R. Identification and Validation of a PD-L1 Binding Peptide for Determination of PDL1 Expression in Tumors. Sci Rep 2017; 7:13682. [PMID: 29057919 PMCID: PMC5651871 DOI: 10.1038/s41598-017-10946-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 08/09/2017] [Indexed: 11/25/2022] Open
Abstract
Blocking the interaction between Programmed Death Ligand 1 (PD-L1) and its receptor, PD-1, is an effective method of treating many types of cancers. Certain tumors overexpress PD-L1, causing host immune cells that express PD-1 to bind PD-L1 and cease killing the tumor. Inhibition of PD-L1 and PD-1 binding can restore host immunity towards tumor killing, and many new drugs have been developed to target this interaction. Current methods of PD-L1 diagnosis have shown to vary based on the antibody, detection kit brand, antigen retrieval method, and clinically defined methods by the FDA. To refine detection of PD-L1, we have identified a peptide, RK-10, and used it to detect PD-L1 expressing tumors with immunohistochemistry or flow cytometry. Flow cytometry was performed on cell lines and patient tissues using a fluorescent peptide (RK-10-Cy5). Immunohistochemistry using a biotin-modified peptide (RK-10-Biotin) was tested against the FDA-approved SP263 clone on biopsied patient tissues. For this study, we evaluated specificity of RK-10 using IHC in over 200 patient tissues, including NSCLC and Hodgkin's Lymphoma. RK-10 shows staining in the tumor regions of FFPE tissues where the SP263 kit does not. RK-10-Cy5 peptide also demonstrates PD-L1 detection in NSCLC, breast, squamous cell carcinoma, and melanoma.
Collapse
Affiliation(s)
- Charles Caldwell
- Departments of Radiology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
- Departments of Bioengineering, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Cory E Johnson
- Departments of Pathology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - V N Balaji
- Molark Enterprises Pvt. Ltd., R.M.V.Extension, Bangalore, 560094, India
| | | | - Richard D Hammer
- Departments of Pathology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Raghuraman Kannan
- Departments of Radiology, School of Medicine, University of Missouri, Columbia, MO, 65212, USA.
- Departments of Bioengineering, School of Medicine, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
9
|
The impact of melanoma genetics on treatment response and resistance in clinical and experimental studies. Cancer Metastasis Rev 2017; 36:53-75. [PMID: 28210865 DOI: 10.1007/s10555-017-9657-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Recent attempts to characterize the melanoma mutational landscape using high-throughput sequencing technologies have identified new genes and pathways involved in the molecular pathogenesis of melanoma. Apart from mutated BRAF, NRAS, and KIT, a series of new recurrently mutated candidate genes with impact on signaling pathways have been identified such as NF1, PTEN, IDH1, RAC1, ARID2, and TP53. Under targeted treatment using BRAF and MEK1/2 inhibitors either alone or in combination, a majority of patients experience recurrences, which are due to different genetic mechanisms such as gene amplifications of BRAF or NRAS, MEK1/2 and PI3K mutations. In principle, resistance mechanisms converge on two signaling pathways, MAPK and PI3K-AKT-mTOR pathways. Resistance may be due to small subsets of resistant cells within a heterogeneous tumor mass not identified by sequencing of the bulk tumor. Future sequencing studies addressing tumor heterogeneity, e.g., by using single-cell sequencing technology, will most likely improve this situation. Gene expression patterns of metastatic lesions were also shown to predict treatment response, e.g., a MITF-low/NF-κB-high melanoma phenotype is resistant against classical targeted therapies. Finally, more recent treatment approaches using checkpoint inhibitors directed against PD-1 and CTLA-4 are very effective in melanoma and other tumor entities. Here, the mutational and neoantigen load of melanoma lesions may help to predict treatment response. Taken together, the new sequencing, molecular, and bioinformatic technologies exploiting the melanoma genome for treatment decisions have significantly improved our understanding of melanoma pathogenesis, treatment response, and resistance for either targeted treatment or immune checkpoint blockade.
Collapse
|
10
|
Cesuroglu T, Syurina E, Feron F, Krumeich A. Other side of the coin for personalised medicine and healthcare: content analysis of 'personalised' practices in the literature. BMJ Open 2016; 6:e010243. [PMID: 27412099 PMCID: PMC4947721 DOI: 10.1136/bmjopen-2015-010243] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Various terms and definitions are used to describe personalised approaches to medicine and healthcare, but in ambiguous and inconsistent ways. They mostly have been defined in a top-down manner. However, actual practices might take different paths. Here, we aimed to provide a 'practice-based' perspective on the debate by analysing the content of 'personalised' practices published in the literature. METHODS The search in PubMed and EMBASE (April 2014) using the terms frequently used for personalised approaches resulted in 5333 records. 2 independent researchers used different strategies for screening, resulting in 157 articles describing 88 'personalised' practices that were implemented/presented on at least 1 individual/patient case. The content analysis was grounded on these data and did not have a priori analytical frameworks. RESULTS 'Personalised medicine/healthcare' can be a commodity in the healthcare market, a way how health services are provided, or a keyword for emerging applications. It can help individuals/patients to gain control of their health, health professionals to provide better services, healthcare organisations to increase effectiveness and efficiency, or national health systems to increase performance. Country examples indicated that for integration of practices into health services, attitude towards innovations and health system and policy context is important. Categorisation based on the terms or the technologies used, if any, was not possible. CONCLUSIONS This study is the first to provide a comprehensive content analysis of the 'personalised' practices in the literature. Unlike the top-down definitions, our findings highlighted not the technologies but real-life issues faced by the practices. 'Personalised medicine' and 'personalised healthcare' can be differentiated by using the former for specific tools available and the latter for health services with a holistic approach, implemented in certain contexts. To realise integration of 'personalised medicine/healthcare' into real life, science, technology, health policy and practice, and society domains must work together.
Collapse
Affiliation(s)
- Tomris Cesuroglu
- Faculty of Health, Medicine and Life Sciences, Department of Social Medicine, Maastricht University, Maastricht, The Netherlands
| | - Elena Syurina
- Faculty of Health, Medicine and Life Sciences, Department of Health, Ethics and Society, Maastricht University, Maastricht, The Netherlands Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Frans Feron
- Faculty of Health, Medicine and Life Sciences, Department of Social Medicine, Maastricht University, Maastricht, The Netherlands
| | - Anja Krumeich
- Faculty of Health, Medicine and Life Sciences, Department of Health, Ethics and Society, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
11
|
Luo D, Smith JA, Meadows NA, Schuh A, Manescu KE, Bure K, Davies B, Horne R, Kope M, DiGiusto DL, Brindley DA. A Quantitative Assessment of Factors Affecting the Technological Development and Adoption of Companion Diagnostics. Front Genet 2016; 6:357. [PMID: 26858745 PMCID: PMC4730156 DOI: 10.3389/fgene.2015.00357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/10/2015] [Indexed: 12/28/2022] Open
Abstract
Rapid innovation in (epi)genetics and biomarker sciences is driving a new drug development and product development pathway, with the personalized medicine era dominated by biologic therapeutics and companion diagnostics. Companion diagnostics (CDx) are tests and assays that detect biomarkers and specific mutations to elucidate disease pathways, stratify patient populations, and target drug therapies. CDx can substantially influence the development and regulatory approval for certain high-risk biologics. However, despite the increasingly important role of companion diagnostics in the realization of personalized medicine, in the USA, there are only 23 Food and Drug Administration (FDA) approved companion diagnostics on the market for 11 unique indications. Personalized medicines have great potential, yet their use is currently constrained. A major factor for this may lie in the increased complexity of the companion diagnostic and corresponding therapeutic development and adoption pathways. Understanding the market dynamics of companion diagnostic/therapeutic (CDx/Rx) pairs is important to further development and adoption of personalized medicine. Therefore, data collected on a variety of factors may highlight incentives or disincentives driving the development of companion diagnostics. Statistical analysis for 36 hypotheses resulted in two significant relationships and 34 non-significant relationships. The sensitivity of the companion diagnostic was the only factor that significantly correlated with the price of the companion diagnostic. This result indicates that while there is regulatory pressure for the diagnostic and pharmaceutical industry to collaborate and co-develop companion diagnostics for the approval of personalized therapeutics, there seems to be a lack of parallel economic collaboration to incentivize development of companion diagnostics.
Collapse
Affiliation(s)
- Dee Luo
- Department of Biological Basis of Behavior, University of Pennsylvania Phildephila, PA, USA
| | - James A Smith
- The Oxford - UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxford, UK; Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of OxfordOxford, UK
| | | | - A Schuh
- National Institute of Health Research, Biomedical Research Centre, Molecular Diagnostic Centre, Oxford University Hospitals Oxford, UK
| | - Katie E Manescu
- Department of Biochemical Engineering, University College London London, UK
| | - Kim Bure
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford Oxford, UK
| | | | - Rob Horne
- The UCL School of Pharmacy, University College London London, UK
| | - Mike Kope
- SENS Research Foundation Mountain View, CA, USA
| | - David L DiGiusto
- Stem Cell and Cellular Therapeutics Operations at Stanford University Hospital and Clinic Stanford, CA, USA
| | - David A Brindley
- Stem Cell and Cellular Therapeutics Operations at Stanford University Hospital and ClinicStanford, CA, USA; The Oxford - UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxford, UK; Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of OxfordOxford, UK; USCF-Stanford Center of Excellence in Regulatory Science and InnovationSan Francisco, CA, USA; Centre for Behavioural Medicine, UCL School of Pharmacy, University College LondonLondon, UK; Harvard Stem Cell InstituteCambridge, MA, USA
| |
Collapse
|
12
|
Zhao X, Modur V, Carayannopoulos LN, Laterza OF. Biomarkers in Pharmaceutical Research. Clin Chem 2015; 61:1343-53. [PMID: 26408531 DOI: 10.1373/clinchem.2014.231712] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 08/17/2015] [Indexed: 11/06/2022]
Abstract
BACKGROUND Biomarkers are important tools in drug development and are used throughout pharmaceutical research. CONTENT This review focuses on molecular biomarkers in drug development. It contains sections on how biomarkers are used to assess target engagement, pharmacodynamics, safety, and proof-of-concept. It also covers the use of biomarkers as surrogate end points and patient selection/companion diagnostics and provides insights into clinical biomarker discovery and biomarker development/validation with regulatory implications. To survey biomarkers used in drug development--acknowledging that many pharmaceutical development biomarkers are not published--we performed a focused PubMed search employing "biomarker" and the names of the largest pharmaceutical companies as keywords and filtering on clinical trials and publications in the last 10 years. This yielded almost 500 entries, the majority of which included disease-related (approximately 60%) or prognostic/predictive (approximately 20%) biomarkers. A notable portion (approximately 8%) included HER2 (human epidermal growth factor receptor 2) testing, highlighting the utility of biomarkers for patient selection. The remaining publications included target engagement, safety, and drug metabolism biomarkers. Oncology, cardiovascular disease, and osteoporosis were the areas with the most citations, followed by diabetes and Alzheimer disease. SUMMARY Judicious biomarker use can improve pharmaceutical development efficiency by helping to select patients most appropriate for treatment using a given mechanism, optimize dose selection, and provide earlier confidence in accelerating or discontinuing compounds in clinical development. Optimal application of biomarker technology requires understanding of candidate drug pharmacology, detailed modeling of biomarker readouts relative to pharmacokinetics, rigorous validation and qualification of biomarker assays, and creative application of these elements to drug development problems.
Collapse
Affiliation(s)
| | - Vijay Modur
- Translational Medicine, Genzyme Corporation, Cambridge, MA
| | | | | |
Collapse
|
13
|
|
14
|
Mourah S, Denis MG, Narducci FE, Solassol J, Merlin JL, Sabourin JC, Scoazec JY, Ouafik L, Emile JF, Heller R, Souvignet C, Bergougnoux L, Merlio JP. Detection of BRAF V600 mutations in melanoma: evaluation of concordance between the Cobas® 4800 BRAF V600 mutation test and the methods used in French National Cancer Institute (INCa) platforms in a real-life setting. PLoS One 2015; 10:e0120232. [PMID: 25789737 PMCID: PMC4366252 DOI: 10.1371/journal.pone.0120232] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/20/2015] [Indexed: 01/01/2023] Open
Abstract
Vemurafenib is approved for the treatment of metastatic melanoma in patients with BRAF V600 mutation. In pivotal clinical trials, BRAF testing has always been done with the approved cobas 4800 BRAF test. In routine practice, several methods are available and are used according to the laboratories usual procedures. A national, multicenter, non-interventional study was conducted with prospective and consecutive collection of tumor samples. A parallel evaluation was performed in routine practice between the cobas 4800 BRAF V600 mutation test and home brew methods (HBMs) of 12 national laboratories, labelled and funded by the French National Cancer Institute (INCa). For 420 melanoma samples tested, the cobas method versus HBM showed a high concordance (93.3%; kappa = 0.86) in BRAF V600 genotyping with similar mutation rates (34.0% versus 35.7%, respectively). Overall, 97.4% and 98.6% of samples gave valid results using the cobas and HBM, respectively. Of the 185 samples strictly fulfilling the cobas guidelines, the concordance rate was even higher (95.7%; kappa = 0.91; 95%CI [0.85; 0.97]). Out of the 420 samples tested, 28 (6.7%) showed discordance between HBM and cobas. This prospective study shows a high concordance rate between the cobas 4800 BRAF V600 test and home brew methods in the routine detection of BRAF V600E mutations.
Collapse
Affiliation(s)
- Samia Mourah
- Department of Pharmacology-Genetics, AP-HP, Saint-Louis Hospital, Paris, France
- INSERM UMRS 976 Paris, F-75010, France
- * E-mail:
| | - Marc G. Denis
- Department of Biochemistry, Nantes University Hospital, Nantes, France
| | | | - Jérôme Solassol
- Department of Biology—Pathology, Montpellier University Hospital, Montpellier, France
| | - Jean-Louis Merlin
- Department of Tumor Biology, Alexis Vautrin Center, Vandoeuvre les Nancy, France
| | | | - Jean-Yves Scoazec
- Laboratory of Pathology and Cytopathology, Edouard Herriot Hospital, Lyon, France
- Department of Biology and Pathology, Gustave Roussy Hospital, Villejuif, France
| | - L’Houcine Ouafik
- Department of Cancer Biology, Marseille Nord University Hospital, Marseille, France
| | - Jean-François Emile
- Department of Pathology, Ambroise Paré Hospital, Boulogne-Billancourt, France
| | - Remy Heller
- Department of Microbiology and Molecular Biology, Colmar hospital, Colmar, France
| | | | | | | |
Collapse
|
15
|
Meadows NA, Morrison A, Brindley DA, Schuh A, Barker RW. An evaluation of regulatory and commercial barriers to stratified medicine development and adoption. THE PHARMACOGENOMICS JOURNAL 2014; 15:6-12. [DOI: 10.1038/tpj.2014.51] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 08/01/2014] [Accepted: 08/13/2014] [Indexed: 11/09/2022]
|
16
|
Foletto MC, Haas SE. Cutaneous melanoma: new advances in treatment. An Bras Dermatol 2014; 89:301-10. [PMID: 24770508 PMCID: PMC4008062 DOI: 10.1590/abd1806-4841.20142540] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/15/2013] [Indexed: 11/22/2022] Open
Abstract
Cutaneous melanoma is a challenge to treat. Over the last 30 years, no drug or
combination of drugs demonstrated significant impact to improve patient survival.
From 1995 to 2000, the use of cytokines such as interferon and interleukin become
treatment options. In 2011, new drugs were approved by the U.S. Food and Drug
Administration, including peginterferon alfa-2b for patients with stage III disease,
vemurafenib for patients with metastatic melanoma with the BRAF V600E mutation, and
ipilimumab, a monoclonal antibody directed to the CTLA-4 T lymphocyte receptor, to
combat metastatic melanoma in patients who do not have the BRAF V600E mutation. Both
ipilimumab and vemurafenib showed results in terms of overall survival. Other trials
with inhibitors of other genes, such as the KIT gene and MEK, are underway in the
search for new discoveries. The discovery of new treatments for advanced or
metastatic disease aims to relieve symptoms and improve patient quality of life.
Collapse
|
17
|
Shipitsin M, Small C, Giladi E, Siddiqui S, Choudhury S, Hussain S, Huang YE, Chang H, Rimm DL, Berman DM, Nifong TP, Blume-Jensen P. Automated quantitative multiplex immunofluorescence in situ imaging identifies phospho-S6 and phospho-PRAS40 as predictive protein biomarkers for prostate cancer lethality. Proteome Sci 2014; 12:40. [PMID: 25075204 PMCID: PMC4114438 DOI: 10.1186/1477-5956-12-40] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 06/18/2014] [Indexed: 11/10/2022] Open
Abstract
Background We have witnessed significant progress in gene-based approaches to cancer prognostication, promising early intervention for high-risk patients and avoidance of overtreatment for low-risk patients. However, there has been less advancement in protein-based approaches, even though perturbed protein levels and post-translational modifications are more directly linked with phenotype. Most current, gene expression-based platforms require tissue lysis resulting in loss of structural and molecular information, and hence are blind to tumor heterogeneity and morphological features. Results Here we report an automated, integrated multiplex immunofluorescence in situ imaging approach that quantitatively measures protein biomarker levels and activity states in defined intact tissue regions where the biomarkers of interest exert their phenotype. Using this approach, we confirm that four previously reported prognostic markers, PTEN, SMAD4, CCND1 and SPP1, can predict lethal outcome of human prostate cancer. Furthermore, we show that two PI3K pathway-regulated protein activities, pS6 (RPS6-phosphoserines 235/236) and pPRAS40 (AKT1S1-phosphothreonine 246), correlate with prostate cancer lethal outcome as well (individual marker hazard ratios of 2.04 and 2.03, respectively). Finally, we incorporate these 2 markers into a novel 5-marker protein signature, SMAD4, CCND1, SPP1, pS6, and pPRAS40, which is highly predictive for prostate cancer-specific death. The ability to substitute PTEN with phospho-markers demonstrates the potential of quantitative protein activity state measurements on intact tissue. Conclusions In summary, our approach can reproducibly and simultaneously quantify and assess multiple protein levels and functional activities on intact tissue specimens. We believe it is broadly applicable to not only cancer but other diseases, and propose that it should be well suited for prognostication at early stages of pathogenesis where key signaling protein levels and activities are perturbed.
Collapse
Affiliation(s)
| | | | - Eldar Giladi
- Metamark Genetics Inc, Cambridge, MA, USA ; Current address: Atreca, San Carlos, CA, USA
| | - Summar Siddiqui
- Metamark Genetics Inc, Cambridge, MA, USA ; Current address: Moderna, Cambridge, MA, USA
| | | | | | - Yi E Huang
- Metamark Genetics Inc, Cambridge, MA, USA
| | - Hua Chang
- Metamark Genetics Inc, Cambridge, MA, USA
| | - David L Rimm
- Department of Pathology, Yale University Medical School, New Haven, CT, USA
| | - David M Berman
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | | | - Peter Blume-Jensen
- Metamark Genetics Inc, Cambridge, MA, USA ; Current address: XTuit Pharmaceuticals, Inc, Cambridge, MA, USA
| |
Collapse
|
18
|
Pant S, Weiner R, Marton MJ. Navigating the rapids: the development of regulated next-generation sequencing-based clinical trial assays and companion diagnostics. Front Oncol 2014; 4:78. [PMID: 24860780 PMCID: PMC4029014 DOI: 10.3389/fonc.2014.00078] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/28/2014] [Indexed: 12/11/2022] Open
Abstract
Over the past decade, next-generation sequencing (NGS) technology has experienced meteoric growth in the aspects of platform, technology, and supporting bioinformatics development allowing its widespread and rapid uptake in research settings. More recently, NGS-based genomic data have been exploited to better understand disease development and patient characteristics that influence response to a given therapeutic intervention. Cancer, as a disease characterized by and driven by the tumor genetic landscape, is particularly amenable to NGS-based diagnostic (Dx) approaches. NGS-based technologies are particularly well suited to studying cancer disease development, progression and emergence of resistance, all key factors in the development of next-generation cancer Dxs. Yet, to achieve the promise of NGS-based patient treatment, drug developers will need to overcome a number of operational, technical, regulatory, and strategic challenges. Here, we provide a succinct overview of the state of the clinical NGS field in terms of the available clinically targeted platforms and sequencing technologies. We discuss the various operational and practical aspects of clinical NGS testing that will facilitate or limit the uptake of such assays in routine clinical care. We examine the current strategies for analytical validation and Food and Drug Administration (FDA)-approval of NGS-based assays and ongoing efforts to standardize clinical NGS and build quality control standards for the same. The rapidly evolving companion diagnostic (CDx) landscape for NGS-based assays will be reviewed, highlighting the key areas of concern and suggesting strategies to mitigate risk. The review will conclude with a series of strategic questions that face drug developers and a discussion of the likely future course of NGS-based CDx development efforts.
Collapse
Affiliation(s)
- Saumya Pant
- Merck Research Laboratories, Molecular Biomarkers and Diagnostics , Rahway, NJ , USA
| | - Russell Weiner
- Merck Research Laboratories, Molecular Biomarkers and Diagnostics , Rahway, NJ , USA
| | - Matthew J Marton
- Merck Research Laboratories, Molecular Biomarkers and Diagnostics , Rahway, NJ , USA
| |
Collapse
|
19
|
Hanamura N, Aruga A. Global development strategy for companion diagnostics based on the usage and approval history for biomarkers in Japan, the USA and the EU. Per Med 2014; 11:27-40. [PMID: 29751388 DOI: 10.2217/pme.13.100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
AIM The aim of this study was to identify gaps between Japan and the West in biomarker usage and the development of companion diagnostics. We also elaborated potential scenarios for companion diagnostic development. METHODS Information on drug labels in Japan, the USA and the EU was obtained from each regulatory authority's web site, as well as label information on in vitro diagnostic testing in Japan and the USA. RESULTS It is necessary to consider two factors when developing companion diagnostics globally: ethnic differences in gene mutations, and the approach to patient selection in clinical trials. A flowchart covering four scenarios was developed. CONCLUSION Two factors that should be taken account when developing companion diagnostics globally was specified. This flowchart is expected to serve as a guide for streaming the development of companion diagnostics.
Collapse
Affiliation(s)
- Nobuyuki Hanamura
- Cooperative Major in Advanced Biomedical Sciences, Joint Graduate School of Tokyo Women's Medical University & Waseda University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-866, Japan
| | - Atsushi Aruga
- Cooperative Major in Advanced Biomedical Sciences, Joint Graduate School of Tokyo Women's Medical University & Waseda University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-866, Japan.
| |
Collapse
|
20
|
Lengauer T. Stellenwert der Bioinformatik für die personalisierte Medizin. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2013; 56:1489-94. [DOI: 10.1007/s00103-013-1819-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Abstract
The incidence of malignant melanoma is increasing annually. Early stages can be cured with surgical intervention but metastatic disease has generally had a dismal prognosis with few effective interventions. A half of all melanomas possess a BRAF mutation, which can be targeted by specific inhibitors. Vemurafenib is an orally active, purposely designed mutant BRAF inhibitor, which has recently been shown to have a survival benefit measured in months in metastatic patients. In this article, the authors discuss the scientific rationale, drug development process and clinical trials that have led to vemurafenib becoming the first BRAF inhibitor approved for the treatment of patients with mutant BRAF metastatic melanoma.
Collapse
Affiliation(s)
- Heather M Shaw
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood, Middlesex, HA6 2RN, UK
| | | |
Collapse
|
22
|
Lopez-Rios F, Angulo B, Gomez B, Mair D, Martinez R, Conde E, Shieh F, Vaks J, Langland R, Lawrence HJ, de Castro DG. Comparison of testing methods for the detection of BRAF V600E mutations in malignant melanoma: pre-approval validation study of the companion diagnostic test for vemurafenib. PLoS One 2013; 8:e53733. [PMID: 23326492 PMCID: PMC3542342 DOI: 10.1371/journal.pone.0053733] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 12/04/2012] [Indexed: 12/31/2022] Open
Abstract
Background The cobas 4800 BRAF V600 Mutation Test is a CE-marked and FDA-approved in vitro diagnostic assay used to select patients with metastatic melanoma for treatment with the selective BRAF inhibitor vemurafenib. We describe the pre-approval validation of this test in two external laboratories. Methods Melanoma specimens were tested for BRAF V600 mutations at two laboratories with the: cobas BRAF Mutation Test; ABI BRAF test; and bidirectional direct sequencing. Positive (PPA) and negative (NPA) percent agreements were determined between the cobas test and the other assays. Specimens with discordant results were tested with massively parallel pyrosequencing (454). DNA blends with 5% mutant alleles were tested to assess detection rates. Results Invalid results were observed in 8/116 specimens (6·9%) with Sanger, 10/116 (8·6%) with ABI BRAF, and 0/232 (0%) with the cobas BRAF test. PPA was 97·7% for V600E mutation for the cobas BRAF test and Sanger, and NPA was 95·3%. For the cobas BRAF test and ABI BRAF, PPA was 71·9% and NPA 83·7%. For 16 cobas BRAF test-negative/ABI BRAF-positive specimens, 454 sequencing detected no codon 600 mutations in 12 and variant codon 600 mutations in four. For eight cobas BRAF test-positive/ABI BRAF-negative specimens, four were V600E and four V600K by 454 sequencing. Detection rates for 5% mutation blends were 100% for the cobas BRAF test, 33% for Sanger, and 21% for the ABI BRAF. Reproducibility of the cobas BRAF test was 111/116 (96%) between the two sites. Conclusions It is feasible to evaluate potential companion diagnostic tests in external laboratories simultaneously to the pivotal clinical trial validation. The health authority approved assay had substantially better performance characteristics than the two other methods. The overall success of the cobas BRAF test is a proof of concept for future biomarker development.
Collapse
Affiliation(s)
- Fernando Lopez-Rios
- Laboratorio de Dianas Terapéuticas, Hospital Universitario Sanchinarro, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Souchelnytskyi S. INDIVIDUALIZATION OF CANCER TREATMENT: CONTRIBUTION OF OMICS TECHNOLOGIES TO CANCER DIAGNOSTIC. BIOTECHNOLOGIA ACTA 2013. [DOI: 10.15407/biotech6.04.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
24
|
Otsubo Y, Ishiguro A, Uyama Y. Regulatory perspective on remaining challenges for utilization of pharmacogenomics-guided drug developments. Pharmacogenomics 2013; 14:195-203. [DOI: 10.2217/pgs.12.194] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pharmacogenomics-guided drug development has been implemented in practice in the last decade, resulting in increased labeling of drugs with pharmacogenomic information. However, there are still many challenges remaining in utilizing this process. Here, we describe such remaining challenges from the regulatory perspective, specifically focusing on sample collection, biomarker qualification, ethnic factors, codevelopment of companion diagnostics and means to provide drugs for off-target patients. To improve the situation, it is important to strengthen international harmonization and collaboration among academia, industries and regulatory agencies, followed by the establishment of an international guideline on this topic. Communication with a regulatory agency from an early stage of drug development is also a key to success.
Collapse
Affiliation(s)
- Yasuto Otsubo
- Office of New Drug II, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Akihiro Ishiguro
- Office of New Drug V, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Yoshiaki Uyama
- Office of Review Management, Pharmaceuticals & Medical Devices Agency (PMDA), Tokyo 100-0013, Japan
- Department of Regulatory Science of Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
25
|
BRAF inhibitor activity in V600R metastatic melanoma. Eur J Cancer 2012; 49:1073-9. [PMID: 23237741 DOI: 10.1016/j.ejca.2012.11.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 11/02/2012] [Indexed: 12/14/2022]
Abstract
Activating mutations in the BRAF gene occur in approximately 50% of melanomas. More than 70% of BRAF mutations are V600E and 10-30% are V600K. Potent and selective BRAF inhibitors have demonstrated significant clinical benefits in patients with V600E and V600K BRAF-mutated melanoma. V600R mutations constitute approximately 3-7% of all BRAF mutations and the activity of BRAF inhibitors in patients with this mutation is unknown. We have treated 45 patients with V600 mutated melanoma including patients with V600R mutation between July 2011 and October 2012 with the selective BRAF inhibitor dabrafenib (n=43) or vemurafenib (n=2) via a compassionate access programme. The overall response rate was 50% for the whole population with a progression-free survival of 5.5 months. Five objective responses were seen in six assessable patients with V600R BRAF mutation (n=9). Our experience suggests that patients with V600R BRAF mutations can be treated successfully with oral BRAF inhibitors, and molecular diagnostic assays should include detection of this type of mutation.
Collapse
|
26
|
Aligning the economic value of companion diagnostics and stratified medicines. J Pers Med 2012; 2:257-66. [PMID: 25562363 PMCID: PMC4251370 DOI: 10.3390/jpm2040257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 11/09/2012] [Accepted: 11/12/2012] [Indexed: 12/21/2022] Open
Abstract
The twin forces of payors seeking fair pricing and the rising costs of developing new medicines has driven a closer relationship between pharmaceutical companies and diagnostics companies, because stratified medicines, guided by companion diagnostics, offer better commercial, as well as clinical, outcomes. Stratified medicines have created clinical success and provided rapid product approvals, particularly in oncology, and indeed have changed the dynamic between drug and diagnostic developers. The commercial payback for such partnerships offered by stratified medicines has been less well articulated, but this has shifted as the benefits in risk management, pricing and value creation for all stakeholders become clearer. In this larger healthcare setting, stratified medicine provides both physicians and patients with greater insight on the disease and provides rationale for providers to understand cost-effectiveness of treatment. This article considers how the economic value of stratified medicine relationships can be recognized and translated into better outcomes for all healthcare stakeholders.
Collapse
|
27
|
Ponti G, Tomasi A, Pellacani G. Overwhelming response to Dabrafenib in a patient with double BRAF mutation (V600E; V600M) metastatic malignant melanoma. J Hematol Oncol 2012; 5:60. [PMID: 23031422 PMCID: PMC3473234 DOI: 10.1186/1756-8722-5-60] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 09/24/2012] [Indexed: 01/08/2023] Open
Abstract
The recent findings brought the necessity of testing the mutational status of a series of genes which had been already identified as responsible for melanomas development and progression, such as BRAF, CKIT and PTEN: the consequent results are, in fact, essential to guide the assessment of the novel treatment protocols based on tailored targeted therapies. We present here the case of a 66 year-old male patient, diagnosed with an advanced melanoma in June 2011, and treated with Dabrafenib for double mutant metastatic disease. The patient was referred to our attention for a large exophytic malignant melanoma on the left shoulder. After complete surgical excision and elective lymph node dissection for presence of metastatic sentinel lymph node, the patient has started high-dose interferon alfa-2b injections as adjuvant therapy for a complete negative staging. The treatment was interrupted in August 2011 due to the appearance of metastatic lymph nodes. Tumor burden was rapidly growing reaching in few months the size of a tennis ball for the tumor mass located in the shoulder. Mutational study of the tumor revealed a double BRAF mutation on V-600E and V600M. This finding incited us to enroll the patient in compassionate Dabrafenib clinical trial. The therapy was started on may 2012 at 150 mg bid dosage. Almost surprisingly for the rapidity of the effect, one week later the lesion on the shoulder has reduced its size by 60% and one month later it has completely disappeared from sight. CT scan of June 2012 documented the astonishing clinical response.
Collapse
Affiliation(s)
- Giovanni Ponti
- Department of Clinical and Diagnostic Medicine and Public Health, University Hospital of Modena and Reggio Emilia, via del Pozzo 7, 41100, Modena, Italy.
| | | | | |
Collapse
|
28
|
Ascierto PA, Kirkwood JM, Grob JJ, Simeone E, Grimaldi AM, Maio M, Palmieri G, Testori A, Marincola FM, Mozzillo N. The role of BRAF V600 mutation in melanoma. J Transl Med 2012; 10:85. [PMID: 22554099 PMCID: PMC3391993 DOI: 10.1186/1479-5876-10-85] [Citation(s) in RCA: 501] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 05/03/2012] [Indexed: 12/19/2022] Open
Abstract
BRAF is a serine/threonine protein kinase activating the MAP kinase/ERK-signaling pathway. About 50 % of melanomas harbors activating BRAF mutations (over 90 % V600E). BRAFV600E has been implicated in different mechanisms underlying melanomagenesis, most of which due to the deregulated activation of the downstream MEK/ERK effectors. The first selective inhibitor of mutant BRAF, vemurafenib, after highly encouraging results of the phase I and II trial, was compared to dacarbazine in a phase III trial in treatment-naïve patients (BRIM-3). The study results showed a relative reduction of 63 % in risk of death and 74 % in risk of tumor progression. Considering all trials so far completed, median overall survival reached approximately 16 months for vemurafenib compared to less than 10 months for dacarbazine treatment. Vemurafenib has been extensively tested on melanoma patients expressing the BRAFV600E mutated form; it has been demonstrated to be also effective in inhibiting melanomas carrying the V600K mutation. In 2011, both FDA and EMA therefore approved vemurafenib for metastatic melanoma carrying BRAFV600 mutations. Some findings suggest that continuation of vemurafenib treatment is potentially beneficial after local therapy in a subset of patients with disease progression (PD). Among who continued vemurafenib >30 days after local therapy of PD lesion(s), a median overall survival was not reached, with a median follow-up of 15.5 months from initiation of BRAF inhibitor therapy. For patients who did not continue treatment, median overall survival from the time of disease progression was 1.4 months. A clinical phase I/II trial is evaluating the safety, tolerability and efficacy of vemurafenib in combination with the CTLA-4 inhibitor mAb ipilimumab. In the BRIM-7 trial vemurafenib is tested in association with GDC-0973, a potent and highly selective inhibitor of MEK1/2. Preliminary data seem to indicate that an additional inhibitor of mutated BRAF, GSK2118436, might be also active on a wider range of BRAF mutations (V600E-K-D-R); actually, treatment with such a compound is under evaluation in a phase III study among stage III-IV melanoma patients positive for BRAF mutations. Overall, BRAF inhibitors were well tolerated; common adverse events are arthralgia, rash, fatigue, alopecia, keratoacanthoma or cutaneous squamous-cell carcinoma, photosensitivity, nausea, and diarrhea, with some variants between different inhibitors.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Istituto Nazionale Tumori Fondazione “G. Pascale”, Naples, Italy
- Unit of Medical Oncology and Innovative Therapy, Istituto Nazionale per lo Studio e la Cura dei Tumori “Fondazione G. Pascale”, Via Mariano Semmola, 80131, Naples, Italy
| | - John M Kirkwood
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Jean-Jacques Grob
- Hopital de la Timone, and Aix-Marseille Univ, 264 Rue St Pierre, 13885, Marseille CEDEX 05, Marseille, France
| | - Ester Simeone
- Department of Melanoma, Istituto Nazionale Tumori Fondazione “G. Pascale”, Naples, Italy
| | - Antonio M Grimaldi
- Department of Melanoma, Istituto Nazionale Tumori Fondazione “G. Pascale”, Naples, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, Department. of Oncology, University Hospital of of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council (CNR), Sassari, Italy
| | - Alessandro Testori
- Melanoma and muscle-cutaneous sarcomas Division, Istituto Europeo di Oncologia, Milan, Italy
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | - Nicola Mozzillo
- Department of Melanoma, Istituto Nazionale Tumori Fondazione “G. Pascale”, Naples, Italy
| |
Collapse
|