1
|
Loo K, Smithy JW, Postow MA, Betof Warner A. Factors Determining Long-Term Antitumor Responses to Immune Checkpoint Blockade Therapy in Melanoma. Front Immunol 2022; 12:810388. [PMID: 35087529 PMCID: PMC8787112 DOI: 10.3389/fimmu.2021.810388] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
With the increasing promise of long-term survival with immune checkpoint blockade (ICB) therapies, particularly for patients with advanced melanoma, clinicians and investigators are driven to identify prognostic and predictive factors that may help to identify individuals who are likely to experience durable benefit. Several ICB combinations are being actively developed to expand the armamentarium of treatments for patients who may not achieve long-term responses to ICB single therapies alone. Thus, negative predictive markers are also of great interest. This review seeks to deepen our understanding of the mechanisms underlying the durability of ICB treatments. We will discuss the currently available long-term data from the ICB clinical trials and real-world studies describing the survivorship of ICB-treated melanoma patients. Additionally, we explore the current treatment outcomes in patients rechallenged with ICB and the patterns of ICB resistance based on sites of disease, namely, liver or CNS metastases. Lastly, we discuss the landscape in melanoma in the context of prognostic or predictive factors as markers of long-term response to ICB.
Collapse
Affiliation(s)
- Kimberly Loo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Internal Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY, United States
| | - James W. Smithy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Michael A. Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Allison Betof Warner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
2
|
Park JJH, Hsu G, Siden EG, Thorlund K, Mills EJ. An overview of precision oncology basket and umbrella trials for clinicians. CA Cancer J Clin 2020; 70:125-137. [PMID: 32031692 PMCID: PMC7187272 DOI: 10.3322/caac.21600] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
With advancements in biomarkers and momentum in precision medicine, biomarker-guided trials such as basket trials and umbrella trials have been developed under the master protocol framework. A master protocol refers to a single, overarching design developed to evaluate multiple hypotheses with the general goal of improving the efficiency of trial evaluation. One type of master protocol is the basket trial, in which a targeted therapy is evaluated for multiple diseases that share common molecular alterations or risk factors that may help predict whether the patients will respond to the given therapy. Another variant of a master protocol is the umbrella trial, in which multiple targeted therapies are evaluated for a single disease that is stratified into multiple subgroups based on different molecular or other predictive risk factors. Both designs follow the core principle of precision medicine-to tailor intervention strategies based on the patient's risk factor(s) that can help predict whether they will respond to a specific treatment. There have been increasing numbers of basket and umbrella trials, but they are still poorly understood. This article reviews common characteristics of basket and umbrella trials, key trials and recent US Food and Drug Administration approvals for precision oncology, and important considerations for clinical readers when critically evaluating future publications on basket trials and umbrella trials and for researchers when designing these clinical trials.
Collapse
Affiliation(s)
- Jay J. H. Park
- Experimental Medicine, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Grace Hsu
- Department of Health Research Methodology, Evidence, and ImpactMcMaster UniversityHamiltonOntarioCanada
| | - Ellie G. Siden
- Experimental Medicine, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Kristian Thorlund
- Department of Health Research Methodology, Evidence, and ImpactMcMaster UniversityHamiltonOntarioCanada
- Cytel IncVancouverBritish ColumbiaCanada
| | - Edward J. Mills
- Department of Health Research Methodology, Evidence, and ImpactMcMaster UniversityHamiltonOntarioCanada
- Cytel IncVancouverBritish ColumbiaCanada
| |
Collapse
|
3
|
Reitsma M, Fox J, Borre PV, Cavanaugh M, Chudnovsky Y, Erlich RL, Gribbin TE, Anhorn R. Effect of a Collaboration Between a Health Plan, Oncology Practice, and Comprehensive Genomic Profiling Company from the Payer Perspective. J Manag Care Spec Pharm 2019; 25:601-611. [PMID: 30632889 PMCID: PMC10398083 DOI: 10.18553/jmcp.2019.18309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Comprehensive genomic profiling (CGP) is a next-generation sequencing-based methodology that detects 4 classes of genomic alterations, as well as gene signature biomarkers such as microsatellite instability and tumor mutational burden. In the context of precision oncology, CGP can help to direct treatment to genomically matched therapies. OBJECTIVE To describe the results of a 3-year observational analysis of patients undergoing testing with CGP assays (either FoundationOne or FoundationOne Heme) at a community oncology practice after a regional health plan implemented a medical policy that enabled coverage of CGP. METHODS A retrospective analysis of medical records was completed at the oncology practice from November 2013 to January 2017; this date range was chosen to coincide with the regional health plan's medical policy implementation of CGP. The medical policy provided coverage of CGP for patients with advanced solid and hematologic cancers. A medical record review assessed all previous and current molecular test results, matched therapy or clinical trial enrollment, and clinical outcomes (clinical benefit or disease progression). The potential cost diversion, from payer to study sponsor, for patients who enrolled in clinical trials was explored. RESULTS There were 96 patients in the community oncology practice who received CGP over the 3-year period, 86 of whom had clinically relevant genomic alterations. Of the 86, 15 patients were treated with genomically matched therapy, and 6 patients enrolled in clinical trials based on CGP results. In a subset of 32 patients who previously underwent conventional testing, most (84%) had clinically relevant genomic alterations detected by CGP that conventional testing did not identify, and a portion of these patients subsequently received treatment based on the CGP results. In the separate cost diversion analysis of 20 patients who enrolled in phase 1 clinical trials, an estimated $25,000 per-patient cost-benefit may have been accrued to the payer. CONCLUSIONS This observational analysis characterized the use of CGP in a large community oncology practice among a group of patients insured by a regional health plan. Clinical trial enrollment was facilitated by CGP use in the community setting and may have contributed to cost diversion from the payer to study sponsors. DISCLOSURES No separate study-related funding was provided by or to Priority Health, Foundation Medicine, and Cancer and Hematology Centers of West Michigan. Data analysis by Reitsma was conducted as part of an internship funded by Priority Health. Reitsma and Fox are employed by Priority Health. Anhorn, Vanden Borre, Cavanaugh, Chudnovsky, and Erlich are employed by Foundation Medicine.
Collapse
Affiliation(s)
- Mitchell Reitsma
- Priority Health, Grand Rapids, Michigan, and Oakland University William Beaumont School of Medicine, Rochester, Michigan
| | - John Fox
- Priority Health, Grand Rapids, Michigan
| | | | | | | | | | | | | |
Collapse
|
4
|
Wang J, Li J, Li Y, Wong WK. A model-based multithreshold method for subgroup identification. Stat Med 2019; 38:2605-2631. [PMID: 30887552 DOI: 10.1002/sim.8136] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 01/29/2019] [Accepted: 02/11/2019] [Indexed: 11/07/2022]
Abstract
Thresholding variable plays a crucial role in subgroup identification for personalized medicine. Most existing partitioning methods split the sample based on one predictor variable. In this paper, we consider setting the splitting rule from a combination of multivariate predictors, such as the latent factors, principle components, and weighted sum of predictors. Such a subgrouping method may lead to more meaningful partitioning of the population than using a single variable. In addition, our method is based on a change point regression model and thus yields straight forward model-based prediction results. After choosing a particular thresholding variable form, we apply a two-stage multiple change point detection method to determine the subgroups and estimate the regression parameters. We show that our approach can produce two or more subgroups from the multiple change points and identify the true grouping with high probability. In addition, our estimation results enjoy oracle properties. We design a simulation study to compare performances of our proposed and existing methods and apply them to analyze data sets from a Scleroderma trial and a breast cancer study.
Collapse
Affiliation(s)
- Jingli Wang
- Department of Statistics and Applied Probability, National University of Singapore, Singapore
| | - Jialiang Li
- Department of Statistics and Applied Probability, National University of Singapore, Singapore.,Duke University-NUS Graduate Medical School, Singapore.,Singapore Eye Research Institute, Singapore
| | - Yaguang Li
- University of Science and Technology of China, Hefei, China
| | - Weng Kee Wong
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
5
|
Chiba Y. Inference on covariate effect types for treatment effectiveness in a randomized trial with a binary outcome. Clin Trials 2019; 16:237-245. [PMID: 30760066 DOI: 10.1177/1740774519828301] [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: 11/16/2022]
Abstract
BACKGROUND/AIMS Some randomized clinical trials seek to establish covariate effect types that indicate whether a covariate is predictive and/or prognostic, in addition to endpoint evaluation. Here, for a case with a binary outcome, we propose that the covariate effect type should be assessed in terms of four types of potential responses: activated- (always-), inert- (never-), causative-, and preventive-responder. METHODS We introduce a new concept of covariate effect types differing from the commonly used "prediction" and "prognosis." We summarize the covariate effect types by inspecting the proportions of subjects in each response type in two subgroups of a covariate, and indicate whether the fractions are augmented, depleted, or neutral as one changes the level of the covariate. Although these proportions cannot generally be identified, we can derive the posterior distributions of the proportions by applying a recently developed Bayesian method. On the basis of the distributions, we would say that the covariate is "augmented-causative" if the difference between the proportions of causative-responders (who would respond if they received the treatment but would not if they did not) in two subgroups is positive, rather than that it is predictive. Similarly, we would say that the covariate is "neutral-activated" if the difference in the proportion of activated-responders (who would respond regardless of their randomized treatment assignment) is close to zero, rather than saying that the covariate is not prognostic. We further describe the relationship between our approach and standard subgroup analysis. RESULTS We applied our approach to data from a randomized clinical trial comparing nivolumab and docetaxel for subjects with advanced nonsquamous non-small-cell lung cancer; we assessed the covariate effect type of PD-L1 status, where PD-L1 is a ligand of the programmed death 1 (PD-1) receptor expressed by activated T cells. When the endpoint was the overall response rate, the posterior distributions for the differences between the proportions of subjects in response types in the PD-L1-positive and negative subgroups yielded an expected-a-posteriori estimate of 0.243 (95% credible interval (CI): 0.094, 0.374) for causative-responders and 0.014 (95% CI: -0.087, 0.125) for activated-responders. Thus, PD-L1 status was augmented-causative for nivolumab effectiveness, to an extent of 24.3%, and was neutral-activated. CONCLUSION Our approach characterizes the covariate effect types in terms of the response types, and to what extent. In a randomized clinical trial with a binary outcome, our approach is a potentially valuable addition to standard subgroup or regression analysis.
Collapse
Affiliation(s)
- Yasutaka Chiba
- Clinical Research Center, Kinki University Hospital, Osakasayama, Japan
| |
Collapse
|
6
|
Inno A, Di Noia V, Martini M, D'Argento E, Di Salvatore M, Arena V, Schinzari G, Orlandi A, Larocca LM, Cassano A, Barone C. Erlotinib for Patients with EGFR Wild-Type Metastatic NSCLC: a Retrospective Biomarkers Analysis. Pathol Oncol Res 2018; 25:513-520. [PMID: 29557085 DOI: 10.1007/s12253-018-0404-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 03/07/2018] [Indexed: 12/14/2022]
Abstract
Erlotinib is approved for the treatment of patients with EGFR mutation positive, metastatic NSCLC. It is also approved as second/third line therapy for EGFR mutation negative patients, but in this setting the benefit of erlotinib is modest and there is no validated biomarker for selecting EGFR wild-type patients who may benefit the most from the treatment. We retrospectively assessed EGFR and K-RAS mutational status, and EGFR, c-MET and IGF1-R expression in tumor samples of 72 patients with metastatic NSCLC treated with erlotinib after at least one prior line of chemotherapy, from 2008 to 2012. We analyzed the association between biomarkers and outcome (RR, PFS, and OS). EGFR mutated patients achieved a better RR (56% vs 8%, p = .002), PFS (10 vs 3 months, HR 0.53, p = 0.48) and OS (20 vs 6 months, HR 0.55, p = .07), compared to EGFR wild-type patients. Among 63 EGFR wild-type patients, those with EGFR high-expression had a better outcome in terms of RR (40% vs 2%, p = .002), PFS (7.5 vs 2 months, HR 0.45, p = .007) and OS (30 vs 5 months, HR 0.34, p < .001) compared to patients with EGFR intermediate or low/negative-expression. IGF1-R expression, c-MET expression and K-RAS mutational status did not significantly affect the outcome; however, no patients with K-RAS mutation or c-MET high-expression achieved an objective response. In patients with metastatic, chemo-refractory EGFR wild-type NSCLC, EGFR high-expression may represent a positive predictor of activity for erlotinib, whereas K-RAS mutation and c-MET high-expression may predict lack of activity. These findings deserve further prospective evaluation.
Collapse
Affiliation(s)
- Alessandro Inno
- Medical Oncology Unit, Cancer Care Center, Ospedale Sacro Cuore Don Calabria, Via don A. Sempreboni 5, 37024 Negrar, Verona, Italy.
| | - Vincenzo Di Noia
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Martini
- Department of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ettore D'Argento
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Vincenzo Arena
- Department of Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Schinzari
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Armando Orlandi
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Alessandra Cassano
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Barone
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
7
|
Abstract
While both prognostic and predictive cancer biomarkers predict clinical outcome, the term 'predictive biomarker' is reserved for the association of a specific therapy with a specific clinical outcome. The advent of genomic signatures and next generation sequencing as candidate predictive biomarkers has led to lengthy and expensive processes for biomarker qualification. The urgency to bring novel predictive cancer biomarkers to practice faster and cheaper requires strategies to lower the bar to biomarker implementation. Three strategies are suggested: identify biomarkers closely coupled to biologic mechanism associated with the clinical endpoint and scalable from cells to humans; identify biomarkers that can be reliably detected and quantified; and assess biomarkers by capacity to reduce toxicity as well as to increase therapy efficacy. Biomarker selection directly and closely related to production of end points by biologic mechanism demonstrated by a ladder of evidence should require less burden of proof clinically than biomarkers that are merely associative.
Collapse
Affiliation(s)
- Kenneth P H Pritzker
- Departments of Laboratory Medicine and Pathobiology, Surgery University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Winther Larsen A, Nissen PH, Meldgaard P, Weber B, Sorensen BS. EGFR CA repeat polymorphism predict clinical outcome in EGFR mutation positive NSCLC patients treated with erlotinib. Lung Cancer 2014; 85:435-41. [PMID: 25017413 DOI: 10.1016/j.lungcan.2014.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/20/2014] [Accepted: 06/21/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Somatic mutations in the epidermal growth factor receptor (EGFR) are predictors of efficacy for treatment with the EGFR tyrosine kinase inhibitor erlotinib in non-small cell lung cancer (NSCLC). A CA repeat polymorphism in intron 1 of the EGFR gene influences the transcription of the EGFR gene. This study evaluates the association between the CA repeat polymorphism and outcome in NSCLC patients treated with erlotinib. MATERIALS AND METHODS Number of CA repeats in the EGFR gene was evaluated with PCR-fragment length analysis by capillary electrophoresis in 432 advanced NSCLC patients treated with erlotinib irrespective of EGFR mutation status. Patients were dichotomized into harboring short allele (CA≤16 in any allele) or long alleles (CA>16 in both alleles). Number of repeats was correlated with clinical characteristic and outcome. A subgroup analysis was performed based on the somatic EGFR mutation status. RESULTS In EGFR mutation positive patients (N=62) we demonstrate a significantly higher median progression free survival (HR=0.39 (0.22-0.70); p=0.002) and overall survival (HR=0.43 (0.23-0.78); p=0.006) in patients also harboring a short CA repeat length vs. a long (median follow-up time of 52.2 months). The result remained highly significant in a multivariate Cox proportional hazards model. This correlation was not seen in EGFR mutation negative patients. CONCLUSION Our study demonstrate that in EGFR mutation positive NSCLC patients treated with erlotinib a low number of CA repeats in intron 1 of the EGFR gene is a predictor for both longer progression free survival and overall survival.
Collapse
Affiliation(s)
- Anne Winther Larsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | - Peter Henrik Nissen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Britta Weber
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
9
|
Grob TJ, Hoenig T, Clauditz TS, Atanackovic D, Koenig AM, Vashist YK, Klose H, Simon R, Pantel K, Izbicki JR, Bokemeyer C, Sauter G, Wilczak W. Frequent intratumoral heterogeneity of EGFR gene copy gain in non-small cell lung cancer. Lung Cancer 2012; 79:221-7. [PMID: 23238037 DOI: 10.1016/j.lungcan.2012.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 11/08/2012] [Accepted: 11/11/2012] [Indexed: 11/27/2022]
Abstract
Next to EGFR mutation, EGFR gene copy number evaluated by fluorescence in situ hybridization (FISH) emerged as a potential predictive marker for sensitivity to EGFR tyrosine kinase inhibitors, although controversial data exist. As the diagnostic accuracy of predictive biomarkers can be substantially limited by regional differences within tumors, heterogeneity of EGFR gene copy gain in NSCLC was assessed in this study. For this purpose, a novel tissue microarray (TMA) based analysis platform was developed. TMAs were constructed containing 8 different tissue cylinders from 144 primary NSCLCs. From 62 of these patients additional nodal metastases were sampled. EGFR gene copy number and EGFR expression was analyzed by FISH and immunohistochemistry according to the suggested guidelines. 13 (9.0%) of the 144 evaluated tumors showed EGFR amplification and 37 (25.7%) tumors high polysomy in at least one tumor area. In 7 (53.8%) of 13 amplified cases the analysis of different tumor areas revealed subclones without EGFR gene copy gain next to subclones with amplification. All of the 36 evaluable tumors with high polysomy showed heterogeneity of EGFR gene copy number with areas negative for gene copy gain within the individual tumors. Heterogeneity of EGFR gene copy gain in lung cancer challenges the concept of using small biopsies for the analysis of EGFR FISH status. EGFR gene copy number is highly heterogeneous within individual NSCLCs and this finding might well be a reason for the controversial clinical data existing regarding responsiveness to anti-EGFR therapy.
Collapse
Affiliation(s)
- Tobias J Grob
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Cho SH, Jeon J, Kim SI. Personalized medicine in breast cancer: a systematic review. J Breast Cancer 2012; 15:265-272. [PMID: 23091538 PMCID: PMC3468779 DOI: 10.4048/jbc.2012.15.3.265] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/06/2012] [Indexed: 01/05/2023] Open
Abstract
The recent advent of "-omics" technologies have heralded a new era of personalized medicine. Personalized medicine is referred to as the ability to segment heterogeneous subsets of patients whose response to a therapeutic intervention within each subset is homogeneous. This new paradigm in healthcare is beginning to affect both research and clinical practice. The key to success in personalized medicine is to uncover molecular biomarkers that drive individual variability in clinical outcomes or drug responses. In this review, we begin with an overview of personalized medicine in breast cancer and illustrate the most encountered statistical approaches in the recent literature tailored for uncovering gene signatures.
Collapse
Affiliation(s)
- Sang-Hoon Cho
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Jongsu Jeon
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Seung Il Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
|
12
|
Jakobsen JN, Sørensen JB. Intratumor heterogeneity and chemotherapy-induced changes in EGFR status in non-small cell lung cancer. Cancer Chemother Pharmacol 2011; 69:289-99. [PMID: 22130585 DOI: 10.1007/s00280-011-1791-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/16/2011] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Biomarker expression is increasingly being used to customize treatment in non-small cell lung cancer (NSCLC). The choice of systemic treatment usually depends on biomarker expression in the initial diagnostic biopsy taken before initiation of first-line treatment. Chemotherapy induces DNA damages in the tumor cells, and thus, biomarker expression in the tumor after systemic treatment might not be identical to biomarker expression in the diagnostic biopsy. NSCLC is highly heterogeneous and biomarker expression may vary in different areas within the same tumor. This review explores the tumor heterogeneity and chemotherapy-induced changes in EGFR biomarker status in NSCLC. METHODS A literature search was performed in August 2011 using pubmed. RESULTS Fifteen trials explored EGFR status in primary tumor and subsequent resected primary tumor, lymph node metastases, or organ metastases. Four papers compared EGFR status in primary tumor or metastases before and after systemic treatment. All trials included relatively few patients and used different chemotherapy regimes, biopsy locations, or time intervals between biopsies. CONCLUSIONS Tumor heterogeneity and probably also previous systemic treatment may be an obstacle for correct interpretation of EGFR status in NSCLC. Heterogeneity regarding EGFR mutations is probably rare and previously reported intra and intertumor heterogeneity may be due to methodological issues. In the current and future clinical scenario with many different options for systemic treatment both as 2nd line and beyond, it is increasingly important to further elucidate the role extent of chemotherapy-induced changes in biomarker expression for proper use of biomarkers in order to customize treatment and thus improve prognosis.
Collapse
Affiliation(s)
- Jan Nyrop Jakobsen
- Department of Oncology, Finsencentre, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | | |
Collapse
|
13
|
Soria JC, Mok TS, Cappuzzo F, Jänne PA. EGFR-mutated oncogene-addicted non-small cell lung cancer: current trends and future prospects. Cancer Treat Rev 2011; 38:416-30. [PMID: 22119437 DOI: 10.1016/j.ctrv.2011.10.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 10/06/2011] [Accepted: 10/22/2011] [Indexed: 01/05/2023]
Abstract
Non-small cell lung cancer (NSCLC) tumours with certain mutations in the epidermal growth factor receptor (EGFR) tyrosine kinase have been termed 'oncogene addicted' to reflect their dependence on EGFR-mediated pro-survival signalling and their high susceptibility to apoptosis induced by EGFR tyrosine kinase inhibitors (EGFR-TKIs, e.g. gefitinib and erlotinib). The most common mutations (L858R and exon 19 deletions) predict an improved clinical response to first-line oral EGFR-TKIs compared with standard platinum-based chemotherapy in patients with advanced NSCLC. Moreover, these mutations are also prognostic of a relatively indolent course of disease, regardless of treatment, as compared with classical NSCLC. Treatment strategies for oncogene-addicted NSCLC are therefore distinct from those for non-oncogene addicted NSCLC, and will depend on the specific genetic mutation present.
Collapse
Affiliation(s)
- Jean-Charles Soria
- Service des Innovations Thérapeutiques Précoces, Institut Gustave Roussy, INSERM Unit 981 and Paris University XI, 39 rue Camille Desmoulins, 94805 Villejuif, France.
| | | | | | | |
Collapse
|
14
|
Clark GM, McShane LM. Biostatistical Considerations in Development of Biomarker-Based Tests to Guide Treatment Decisions. Stat Biopharm Res 2011. [DOI: 10.1198/sbr.2011.09038] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
15
|
Quantitative immunohistochemistry in lung cancer: clinical perspective. Folia Histochem Cytobiol 2010; 48:7-11. [DOI: 10.2478/v10042-010-0014-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
16
|
Abstract
An ongoing research and multiple clinical trials involve new targeted therapies and less aggressive treatment regimens that improve survival in patients with lung cancer. Targeted therapeutic agents are based on the concept of discovering genetic alterations and the signaling pathways altered in cancer and have added significantly to our armamentarium in order to prolong patient survival and minimizing drug toxicity. Among 34 molecularly targeted drugs approved by U.S. Food and Drug Administration (FDA) for treatment of various cancers since 1998 three targeted therapies have been approved for treatment of lung cancer (gefitinib in 2002, erlotinib in 2003, and bevacizumab in 2006).This review focuses on the targeted therapies in lung cancer, the molecular biomarkers that help identify patients that will benefit for these targeted therapies, describes the basic molecular biology principles and selected molecular diagnostic techniques and the pathological features correlated with molecular abnormalities in lung cancer. Lastly, new molecular abnormalities described in lung cancer that are predictive to novel promising targeted agents in various phases of clinical trials are discussed.
Collapse
Affiliation(s)
- Lucian R Chirieac
- Harvard Medical School, Staff Pathologist, Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02465,
| | | |
Collapse
|
17
|
Provencio M, García-Campelo R, Isla D, de Castro J. Clinical-molecular factors predicting response and survival for tyrosine-kinase inhibitors. Clin Transl Oncol 2009; 11:428-36. [DOI: 10.1007/s12094-009-0381-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
18
|
Carlson JJ, Garrison LP, Ramsey SD, Veenstra DL. The potential clinical and economic outcomes of pharmacogenomic approaches to EGFR-tyrosine kinase inhibitor therapy in non-small-cell lung cancer. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2009; 12:20-27. [PMID: 18647257 DOI: 10.1111/j.1524-4733.2008.00415.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
OBJECTIVES Pharmacogenomic applications in oncology offer significant promise, but the clinical and economic implications remain unclear. The objective of this study was to evaluate the potential cost-utility of implementing epidermal growth factor receptor (EGFR) testing before initiating second-line therapy for advanced refractory non-small-cell lung cancer (NSCLC). METHODS We developed a decision analytic model to evaluate the cost-utility of EGFR protein expression or gene copy number testing compared to standard care with erlotinib in refractory advanced NSCLC patients. Costs and utilities were obtained from publicly available sources. We performed sensitivity analyses to evaluate uncertainty in the results. RESULTS The quality-adjusted life expectancies for erlotinib, EGFR protein expression testing, and gene copy number testing were: 0.44, 0.48, and 0.50 quality-adjusted life years (QALYs); and the costs were: $57,238, $63,512, and $66,447, respectively. The most cost-effective testing option, EGFR gene copy number testing, produced an incremental cost-effectiveness ratio of $162,018/QALY compared to no testing (erlotinib). The results were most sensitive to the survival estimates, health state utilities, and cost of disease progression. In the probabilistic sensitivity analyses, erlotinib without testing was the optimal treatment strategy until the $150,000/QALY willingness-to-pay threshold, after which gene copy testing was optimal. The discounted expected value of perfect information at a $100,000/QALY threshold in the USA over 5 years was $31.4 million. CONCLUSIONS The study results suggest that EGFR pharmacogenomic testing has the potential to improve quality-adjusted life expectancy in the treatment of refractory NSCLC by a clinically meaningful margin at a value commensurate with the approved therapies in this setting. Additional research in this area is warranted.
Collapse
Affiliation(s)
- Josh J Carlson
- School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
19
|
Lin CC, Calvo E, Papadopoulos KP, Patnaik A, Sarantopoulos J, Mita AC, Preston GG, Mita MM, Rodon J, Mays T, Yeh IT, O'Rourke P, Takimoto CH, Dancey JE, Chen H, Tolcher AW. Phase I study of cetuximab, erlotinib, and bevacizumab in patients with advanced solid tumors. Cancer Chemother Pharmacol 2008; 63:1065-71. [PMID: 18795291 DOI: 10.1007/s00280-008-0811-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2008] [Accepted: 07/18/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Complex interrelationships exist between the epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) receptor pathways. EGFR activation elicits cell proliferation and increased VEGF expression. To maximally inhibit EGFR and then downstream VEGF activity, this phase I study was initiated to determine the maximum tolerated dose (MTD) of erlotinib with fixed-dose cetuximab, and then combine with bevacizumab in patients with advanced malignancies. PATIENTS AND METHODS Patients with advanced malignancies likely to express EGFR were treated with a full dose of cetuximab intravenous weekly, combined with various doses of oral erlotinib daily (Part 1). Once the MTD was determined in Part 1, escalating doses of bevacizumab were administered intravenously biweekly (Part 2). RESULTS Forty patients were enrolled and received 155 courses over four dose levels. In Part 1, dose-limiting grade 3 rash occurred in two patients administered with erlotinib at 100 mg daily, and the MTD of erlotinib for this combination was 50 mg daily with standard-dose cetuximab (11 patients treated). Other adverse events included rash, diarrhea, fatigue, and hypomagnesemia. In Part 2, bevacizumab at 10 mg/kg intravenous every 2 weeks was safely added, with additional nondose-limiting headache, proteinuria, and hypertension. There was one partial response in a patient with renal cell carcinoma. Durable stable disease was observed in five patients for 6-11 months. CONCLUSIONS The MTD for Part 1 was 50 mg daily of erlotinib combined with standard cetuximab. Bevacizumab at 10 mg/kg biweekly can be safely administered with the MTD for erlotinib and cetuximab combination.
Collapse
Affiliation(s)
- Chia-Chi Lin
- Institute for Drug Development, Cancer Therapy and Research Center, San Antonio, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Dziadziuszko R, Hirsch FR. Advances in genomic and proteomic studies of non-small-cell lung cancer: clinical and translational research perspective. Clin Lung Cancer 2008; 9:78-84. [PMID: 18501093 DOI: 10.3816/clc.2008.n.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recent years have brought tremendous progress in the development of genomic and proteomic platforms to study cancer biology. Tests based on these platforms are helpful in early diagnosis, prognosis, and prediction of treatment benefit. Molecular studies performed on minimally invasive material (plasma, sputum) from individuals participating in longitudinal or case-control studies have approximately 70%-90% sensitivity and specificity to detect lung cancer. In operable non-small-cell lung cancer, genomic and proteomic studies yield better prognostic information than pathologic staging. There are several examples of successful identification of predictive assays for benefit from chemotherapy (ERCC1, RRM1, p27Kip1, and p53 expression) or targeted therapies (epidermal growth factor receptor [EGFR] gene copy number, EGFR activating mutations, EGFR protein expression, serum proteomic profile). These markers should be prospectively tested in clinical studies before they can be routinely used in the clinic.
Collapse
Affiliation(s)
- Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Poland.
| | | |
Collapse
|
21
|
Abstract
After failure of first-line chemotherapy for advanced non-small cell lung cancer, many patients remain candidates to receive further antitumor treatment. To guide clinical management of these patients and to suggest priorities for clinical research, an International Panel of Experts met in Naples (Italy) in April 2007. Results and evidence-based conclusions are presented in this article. Single-agent chemotherapy with docetaxel or pemetrexed is the recommended option for unselected patients with performance status 0 to 2 who are candidates for second-line chemotherapy for advanced non-small cell lung cancer. Docetaxel has demonstrated superiority compared with best supportive care. Pemetrexed has been shown to be noninferior to docetaxel, with a more favorable toxicity profile. Erlotinib is effective in pretreated patients, and can be given second-line in patients not suitable or intolerant to chemotherapy, and in all patients as third-line treatment after failure of second-line chemotherapy. Gefitinib failed to show superiority to placebo as second- or third-line treatment, but it has been shown to be noninferior to docetaxel. In selected patients such as lifetime nonsmokers or those of East-Asian ethnicity, erlotinib, or gefitinib (where licensed) may be considered as second-line treatment even if they are fit for chemotherapy. Best supportive care in addition to active treatment remains important for all patients, but may be the exclusive option for patients unsuitable for more aggressive therapy. Further research is mandatory, to find better treatments, and to identify clinical and molecular predictive markers of efficacy, both for chemotherapy and for novel biologic agents.
Collapse
|
22
|
Hirsch FR, Dziadziuszko R, Thatcher N, Mann H, Watkins C, Parums DV, Speake G, Holloway B, Bunn PA, Franklin WA. Epidermal growth factor receptor immunohistochemistry: comparison of antibodies and cutoff points to predict benefit from gefitinib in a phase 3 placebo-controlled study in advanced nonsmall-cell lung cancer. Cancer 2008; 112:1114-21. [PMID: 18219661 DOI: 10.1002/cncr.23282] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The ISEL (Iressa Survival Evaluation in Lung Cancer) clinical trial evaluated the efficacy of gefitinib versus placebo in pretreated nonsmall-cell lung cancer patients. Two different antibodies, scoring systems, and cutoff points of epidermal growth factor receptor (EGFR) protein expression were compared to predict response and survival of enrolled patients. METHODS EGFR expression was assessed in tumor samples by immunohistochemistry using the Dako EGFR pharmDx kit (scoring percent of tumor cells with positive staining) and Zymed monoclonal antibody clone 31G7 (scoring staining index derived from proportion of positive cells times staining intensity). RESULTS Data for EGFR expression were available for 379 patients for Dako and 357 patients for Zymed antibody (22% and 21%, respectively, of trial population). Objective response rates in gefitinib-treated EGFR-positive patients defined with various cutpoints with Dako antibody varied between 8% and 12%, and with Zymed antibody between 10% and 13%. Lower cutoff points with Dako antibody provided the best discrimination between EGFR-positive and EGFR-negative patients for survival hazard ratios comparing gefitinib to placebo, with a significant treatment/cutoff point interaction for 10% cutoff point (P = .049). A similar but less apparent trend was noted for Zymed antibody, although the discrimination between hazard ratios was not significant for any cutoff point analyzed. CONCLUSIONS Assessment with the Dako PharmDx kit and percentage of cells with positive staining may provide more accurate prediction of differential effect on survival with gefitinib than assessment with Zymed antibody and staining index. Using higher cutpoints to define positivity does not improve test discrimination.
Collapse
Affiliation(s)
- Fred R Hirsch
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO 80045, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bianchini D, Jayanth A, Chua YJ, Cunningham D. Epidermal growth factor receptor inhibitor-related skin toxicity: mechanisms, treatment, and its potential role as a predictive marker. Clin Colorectal Cancer 2008; 7:33-43. [PMID: 18279575 DOI: 10.3816/ccc.2008.n.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human epidermal growth factor receptor (HER1/EGFR/ErbB1) signaling is aberrant and overexpressed in many solid malignancies making it an appealing target for biologic agents. Among the classes of drugs targeting EGFR are monoclonal antibodies and EGFR tyrosine kinase inhibitors, which have been shown effective and generally well tolerated in different clinical settings. The majority of patients treated with EGFR inhibitors (EGFRIs) develop specific dose-dependent skin toxicity. This side effect may lead to physical and psychosocial discomfort which can result in dose reduction or treatment interruption. The relationship between rash and clinical outcome has stimulated interest in this particular toxicity as a possible surrogate marker of efficacy in patients treated with targeted agents against EGFR. This review aims to summarize and update the current knowledge of the clinical presentation, predictive and prognostic value, and the management of EGFRI-related skin toxicity.
Collapse
Affiliation(s)
- Diletta Bianchini
- Department of Medicine, Royal Marsden Hospital, Down's Road, Sutton, Surrey, SM2 5PT UK
| | | | | | | |
Collapse
|
24
|
|
25
|
Dziadziuszko R, Szostakiewicz B, Hirsch FR. Epidermal Growth Factor Receptor Targeted Therapy—Markers of Sensitivity and Response. Lung Cancer 2007. [DOI: 10.3109/9781420020359.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|