Skip to main content
Journal of Managed Care & Specialty Pharmacy logoLink to Journal of Managed Care & Specialty Pharmacy
. 2016 Oct;22(10):10.18553/jmcp.2016.15404. doi: 10.18553/jmcp.2016.15404

Biopsy Procedures and Molecular Testing Utilization and Related Costs in Patients with Metastatic Lung Cancer

Reshma Shinde 1,*, Xiting Cao 1, Smita Kothari 1
PMCID: PMC10397943  PMID: 27668568

Abstract

BACKGROUND:

Epidermal growth factor receptor (EGFR) gene mutations and anaplastic lymphoma kinase (ALK) gene rearrangements are key therapeutic targets for biomarker-driven treatment with an EGFR or ALK tyrosine kinase inhibitor (TKI) in patients with metastatic non-small cell lung cancer (NSCLC). To appropriately guide treatment decisions, since 2011, the National Comprehensive Cancer Network and the American Society of Clinical Oncology therefore recommend EGFR and ALK analysis in tumor samples obtained at the time of diagnosis in patients with non-squamous NSCLC. Currently, there are limited data on utilization patterns and cost of biopsy procedures and biomarker tests in patients with metastatic NSCLC who receive an EGFR or ALK TKI.

OBJECTIVES:

To (a) describe utilization patterns and costs associated with biopsy procedures and biomarker testing in patients with NSCLC who received erlotinib or crizotinib between 2009 and 2012 and (b) investigate the timing of these procedures relative to the erlotinib or crizotinib index date.

METHODS:

Adult patients with metastatic lung cancer were identified by ICD-9-CM diagnostic codes within the Truven Health Analytic MarketScan database. Patients were included in the analysis if they had an index erlotinib or crizotinib claim between January 1, 2009, and September 30, 2012 (index period) and were continuously enrolled for ≥ 12 months before the index claim. Because there is no specific ICD-9-CM diagnostic code for NSCLC, patients with metastatic lung cancer who received erlotinib or crizotinib were considered to have metastatic NSCLC. Using CPT and ICD-9-CM codes, lung biopsy procedures performed during the 24 months before or 12 months after the index claim date were identified. For every patient, biomarker testing claims for EGFR and ALK were identified using the molecular pathology stacked CPT code during the 2 months before or 1 month after the index date. The frequency of claims for biopsy procedures and biomarker testing was analyzed descriptively. The overall summary measures for biomarker testing, especially frequency of EGFR testing in patients receiving erlotinib, was also described as before and after 2011, the year when biomarker testing became part of the guidelines. Per patient and overall costs for biopsy procedures and biomarker testing were calculated from payer and patient perspectives.

RESULTS:

Of the 4,926 identified patients, 4,801 (97.5%) received erlotinib, and 125 (2.5%) received crizotinib. Biopsy procedure claims were identified for 3,579 (72.7%) patients, including 3,503 (73.0%) erlotinib recipients and 76 (60.8%) crizotinib recipients. Biomarker testing claims were identified for 675 (13.7%) patients, including 634 (13.2%) erlotinib recipients and 41 (32.8%) crizotinib recipients. Overall, most biomarker testing procedures (476 of 741) were identified in 435 (of 675) patients after year 2011. Also, among erlotinib recipients, percentage of patients receiving EGFR testing was increased over the index period. Per patient mean (SD) numbers of biopsy procedures and biomarker tests were 1.2 (1.1) and 0.2 (0.4), respectively. In the outpatient setting, per patient mean (SD) cost per biopsy procedure was $1,223 ($1,899) from the payer perspective and $60 ($147) from the patient perspective, whereas in the inpatient setting, it was $8,163 ($18,712) and $180 ($691), respectively. Among patients receiving at least 1 biomarker test, the per patient mean (SD) cost for the overall population was $891 ($1,062) and $43 ($229); for erlotinib recipients, it was $906 ($1,084) and $42 ($228); and for crizotinib recipients, it was $664 ($576) and $55 ($243) in payer and patient perspectives, respectively.

CONCLUSIONS:

This study provides insight into the use and cost of biopsy and biomarker testing procedures in patients with metastatic NSCLC. The low frequency of biomarker testing highlights the need for more awareness of testing to guide treatment decisions in these patients. Costs associated with biopsy procedures and biomarker testing provide insight into the economic impact on metastatic NSCLC patients treated with targeted therapy.


What is already known about this subject

  • Epidermal growth factor receptor (EGFR) gene mutations and anaplastic lymphoma kinase (ALK) gene rearrangements have become key therapeutic targets for biomarker-driven treatment in patients with advanced/metastatic non-small cell lung cancer (NSCLC) of non-squamous etiology.

  • Guidelines recommend EGFR and ALK biomarker testing to identify patients eligible for erlotinib and crizotinib therapy.

  • There is a lack of information on the use and cost of biopsy procedures and biomarker testing.

What this study adds

  • This retrospective claims database analysis study provided information on the utilization patterns of and costs associated with biopsy procedures and biomarker testing in patients with advanced lung cancer who received erlotinib or crizotinib.

  • Despite guideline recommendations, the frequency of biomarker testing after year 2011 is low.

  • The costs for biopsy sample collection and biomarker testing varied considerably, providing insight into the economic impact of these procedures in the management of patients with metastatic NSCLC who receive biomarker-driven therapies.

Tremendous advances in lung cancer biology over the past decade have paved the way for targeted therapy in patients with advanced non-small cell lung cancer (NSCLC).1 Mutations in the epidermal growth factor receptor (EGFR) gene and rearrangements of the anaplastic lymphoma kinase (ALK) gene have become key therapeutic targets for biomarker-driven treatment in patients with non-squamous NSCLC.2

Nearly 90% of EGFR mutations manifest as either deletions in exon 19 or substitutions in exon 21 (L858R); both of these EGFR mutations confer increased sensitivity to tyrosine kinase inhibitors (TKIs) that target the intracellular tyrosine kinase domain.3 Fusion of the echinoderm microtubule-associated protein-like-4 (EML4) and ALK genes is associated with lung adenocarcinomas lacking EGFR mutations and is correlated with ALK expression.4 In the United States, prevalence of EGFR mutations is 10%-15%, and ALK rearrangements are 2%-7%, depending on the population studied and the detection method used.5-9

Randomized controlled trials comparing EGFR TKIs to chemotherapy in first-line EGFR mutation-positive NSCLC patients have demonstrated a significant progression-free survival (PFS; range = 9.2-13.1 months; hazard ratio range = 0.16-0.48) and response rates ranging from 61%-83%.10-12 Based on these results, where sensitizing EGFR mutations predicted better response and PFS in patients with NSCLC,10,12-16 in 2011, the National Comprehensive Cancer Network (NCCN) and American Society of Clinical Oncology (ASCO) recommended EGFR analysis in tumor samples collected at the time of non-squamous NSCLC diagnosis before administration of erlotinib.17,18 After further approval of crizotinib in August 2011, NCCN recommended ALK analysis as well.19 In 2013, the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology also recommended EGFR and ALK testing for selection of lung cancer patients for EGFR and ALK TKIs.20 Compared with chemotherapy, these therapies have shown better quality of life and improvement in symptoms in the clinical trial setting21,22 and in clinical practice.23 A recent analysis also suggested that biomarker testing for EGFR mutations and ALK overexpression and biomarker conditional treatment is a cost-effective strategy compared with treatment with chemotherapy and no testing in metastatic NSCLC.24

Currently, there is a lack of information on utilization patterns of biopsy procedures and biomarker testing in NSCLC. This information is necessary to gain insight into the treatment patterns of targeted therapies as more and more similar therapies are becoming available. Since NSCLC treatment decisions are now based on a complex algorithm in which histology and biomarkers are important factors, a multidisciplinary approach to NSCLC management is imperative.25,26 Such a multidisciplinary approach means that it is important to understand the type of provider associated with these procedures, the settings of care (inpatient and outpatient), and the types of visits.

It is also crucial to explore costs associated with biopsy and biomarker testing procedures, as expanding use of targeted therapies necessitates increased use of these procedures, which may affect health care costs.27 An analysis conducted using integrated medical and pharmaceutical data from 4 Blue Cross Blue Shield plans showed a wide variation observed in per patient cost for EGFR testing. From a payer perspective, the plan-paid cost for EGFR testing ranged from $5 to $732.28 However, details were lacking, particularly for patient out-of-pocket costs for biomarker testing and for biopsy procedures.

The purpose of this study was to describe the use and costs of biopsy and biomarker testing procedures in patients with NSCLC who received erlotinib or crizotinib and to investigate the timing of these procedures relative to the erlotinib or crizotinib index date.

Methods

This descriptive, retrospective cohort study was conducted using the Commercial Claims and Encounters and Medicare Supplemental databases within the Truven Health Analytic MarketScan Research Databases. The MarketScan databases represent the medical experience of insured employees and their dependents for active employees, early retirees, COBRA continuers, and Medicare-eligible retirees with employer-provided Medicare Supplemental plans and represents over 77 million patients since 1996. It captures person-specific clinical utilization, expenditures, and enrollment across inpatient, outpatient, prescription drug, and carve-out services from a selection of large employers, health plans, and government and public organizations. The Commercial Claims and Encounters Database captures individuals insured by employer-sponsored plans (i.e., non-Medicare-eligible), and Medicare Supplemental Database captures Medicare-eligible retirees with employer-sponsored Medicare Supplemental plans, which predominantly contains fee-for-service plan data.

Identification of Patients Receiving Erlotinib or Crizotinib

The study period spanned from January 1, 2007, to September 30, 2013. The index period was January 1, 2009, through September 30, 2012. Patients were included in the study if they were aged ≥ 18 years; had ≥ 1 International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) diagnosis code at the primary and/or secondary position for metastatic lung cancer (162.2-162.5, 162.8-162.9) during the study period; had an index pharmacy claim for erlotinib or crizotinib identified by National Drug Code numbers (54868-5290, 54868-5447, 54868-5474, 50242-062, 50242-063, or 50242-064 for erlotinib; 0069-8140 or 0069-8141 for crizotinib) during the index period with no other erlotinib or crizotinib pharmacy claim in the previous 180 days; and had a continuous enrollment for ≥ 12 months before the index date (i.e., the date of the first pharmacy claim for erlotinib or crizotinib identified during the index period). Since the ICD-9-CM classification system does not provide separate diagnosis codes for NSCLC, in this study, patients with metastatic lung cancer who received erlotinib or crizotinib were assumed to have NSCLC, since these therapies are indicated exclusively for treatment of NSCLC. Patients were further categorized as newly diagnosed or previously diagnosed. Patients were defined as newly diagnosed if they had no identifiable ICD-9-CM code for lung cancer in 1 year before the first ICD-9-CM lung cancer code identified in the index period; others were defined as previously diagnosed.

Biopsy Procedures and Biomarker Testing Identification

Lung biopsy procedures performed at least up to 12 months and until 24 months before and 12 months after the index date were identified by Current Procedural Terminology (CPT) and/or ICD-9-CM procedure codes (Appendix A, available in online article). All CPT codes of the same biopsy type on the same day were considered to be related to the same biopsy procedure. Nonbiopsy-specific CPT codes for guided needle placement were considered to be associated with biopsy procedure only when they were identified on the same day as the biopsy-related CPT code. For inpatient admission claims, all claims with the biopsy procedure code as the primary procedure under the same diagnosis-related group code were considered to be associated with the same biopsy procedure. CPT codes for ancillary biopsy procedures were also identified. For all lung biopsy procedures, information on the type of biopsy, provider, facility where the biopsy was performed, and the type of visit (i.e., inpatient, outpatient, or emergency room) was identified.

During a period of 2 months before and 1 month after the index date, biomarker testing for EGFR and ALK was identified by using CPT codes classified as molecular pathology stacking codes (83890-83914) or array codes (88384-88386; Appendices B and C, available in online article). These CPT codes are a series of codes representing individual steps or techniques used in performing a test. Although these codes were discontinued as of January 2013, they were considered to be appropriate for this study, since the biomarker testing identification period ranged from November 1, 2008, to October 30, 2012. During the biomarker testing identification period, there was no specific identification code for EGFR and ALK; therefore, it was assumed that testing conducted in patients receiving erlotinib was for EGFR, and testing conducted in patients receiving crizotinib was for ALK. All biomarker testing-related CPT codes with the same date were identified as related to the same test and were considered to be part of the same pathology code stack. For all biomarker testing, information on the location of testing was also identified.

Biopsy Procedure and Biomarker Testing Cost Identification

Costs associated with lung biopsy procedures and biomarker testing were estimated from payer and patient perspectives. Payer costs included the net payment made by the payer toward the biopsy procedure or biomarker test; patient costs included the amount paid by the patient, which collectively included the amount paid as copayment, coinsurance, and/or deductible. Costs were described by the type of facility (inpatient or outpatient). Costs of biopsy procedures and biomarker testing were calculated based on the associated CPT codes previously described. For biopsy procedures, costs associated with accompanying ancillary procedures were also taken into account and contributed to the final cost. For biomarker testing, costs for interpretation and reporting were also taken into account. All costs were adjusted to April 2016, based on the Consumer Price Index.

Statistical Analyses

All study variables, including baseline characteristics and frequency of biopsy procedures and biomarker testing, were analyzed using descriptive statistics. The overall summary measures for biomarker testing and for patients receiving erlotinib, especially frequency of EGFR testing, was described before and after 2011, the year when biomarker testing became part of the guidelines.

Costs of biopsy procedures were calculated separately for the inpatient and outpatient settings, with summary statistics of net payment by payer and copayment by patients, including coinsurance and/deductibles, presented for the overall cohort. Costs of biomarker testing were calculated similarly without differentiating the setting of the test.

Statistical analyses were conducted using SAS software, version 9.2 (SAS Institute, Cary, NC).

Results

After applying the eligibility criteria, 4,926 patients with metastatic lung cancer who received erlotinib or crizotinib during the index period were identified. Of these, 4,801 (97.5%) received erlotinib, and 125 (2.5%) received crizotinib (Figure 1). Among erlotinib and crizotinib recipients, biopsy procedure claims were identified in 3,503 (73.0%) and 76 (60.8%) patients, respectively. Mean (standard deviation [SD]) number of biopsy procedures was 1.2 (1.1) for the total population. Biomarker testing claims were identified in 634 (13.2%) erlotinib and 41 (32.8%) crizotinib recipients. Overall mean (SD) number of testing procedures was 0.2 (0.4), and it was 0.1 (0.3) after 2011 in the total population.

FIGURE 1.

FIGURE 1

Patient Selection Flow

Patient baseline characteristics are summarized in Table 1 for the total population and for erlotinib and crizotinib recipients. Overall, 2,600 (52.8%) patients were female, and mean (SD) age at index date was 67 (11.6) years. Study population was evenly distributed throughout the study period except in 2012 because of the early cut-off date. Preferred provider organization plans covered almost half of the study population (n = 2,251, 45.7%). The majority of patients (n = 3,740, 76%) were newly diagnosed. Crizotinib recipients, of whom 121 (96.8%) were newly diagnosed, were more likely to start therapy in 2012.

TABLE 1.

Patient Baseline Characteristics

Characteristic Total Population N = 4,926 Erlotinib Recipients n = 4,801 Crizotinib Recipients n = 125
All Patients Previously Diagnosed Patients (n = 1,182) Newly Diagnosed Patients (n = 3,619) All Patients Previously Diagnosed Patients (n = 4) Newly Diagnosed Patients (n = 121)
Mean age at index date (SD), years 67 (11.6) 67 (11.5) 67.5 (10.8) 67.1 (11.7) 55 (11.4) 58.5 (11.1) 55.3 (11.5)
Sex, n (%)
 Male 2,326 (47.2) 2,274 (47.4) 564 (47.7) 1,710 (47.3) 52 (41.6) 2 (50.0) 50 (41.3)
 Female 2,600 (52.8) 2,527 (52.6) 618 (52.3) 1,909 (52.8) 73 (58.4) 2 (50.0) 71 (58.7)
Age group at index date, n (%)
 18-34 years 20 (0.4) 13 (0.3) 3 (0.3) 10 (0.3) 7 (5.6) 0 (0.0) 7 (5.8)
 35-44 years 80 (1.6) 67 (1.4) 12 (1.0) 55 (1.5) 13 (10.4) 1 (25.0) 12 (10.0)
 45-54 years 599 (12.2) 565 (11.8) 111 (9.4) 454 (12.5) 34 (27.2) 0 (0.0) 34 (28.1)
 55-64 years 1,591 (32.3) 1,540 (32.1) 375 (31.8) 1,165 (32.2) 51 (40.8) 1 (25.0) 50 (41.3)
 ≥ 65 years 2,636 (53.5) 2,616 (54.5) 681 (57.0) 1,935 (53.5) 20 (16.0) 2 (50.0) 18 (14.9)
Year of index date, n (%)
 2009 1,300 (26.4) 1,300 (27.1) 817 (69.1) 483 (13.4) 0 (0.0) 0 (0.0) 0 (0.0)
 2010 1,237 (25.1) 1,237 (25.8) 230 (19.5) 1,007 (27.8) 0 (0.0) 0 (0.0) 0 (0.0)
 2011 1,405 (28.5) 1,357 (28.3) 92 (7.8) 1,265 (35.0) 48 (38.4) 1 (25.0) 47 (38.8)
 2012 984 (20.0) 907 (18.9) 43 (3.6) 864 (23.9) 77 (61.6) 3 (75.0) 74 (61.2)
Type of insurance, n (%)
 EPO 32 (0.7) 29 (0.6) 2 (0.2) 27 (0.8) 3 (2.4) 0 (0.0) 3 (2.5)
 HMO 657 (13.3) 637 (13.3) 133 (11.3) 504 (13.9) 20 (16.0) 2 (50.0) 18 (14.9)
 POS 272 (5.5) 264 (5.5) 69 (5.8) 195 (5.4) 8 (6.4) 0 (0.0) 8 (6.6)
 PPO 2,251 (45.7) 2,175 (45.3) 583 (49.3) 1,592 (44.0) 76 (60.8) 2 (50.0) 74 (61.2)
 Other 1,714 (34.8) 1,696 (35.3) 395 (33.4) 1,301 (36.0) 18 (14.4) 0 (0.0) 18 (14.9)

EPO = exclusive provider organization; HMO = health maintenance organization; POS = point of service; PPO = preferred provider organization; SD=standard deviation.

Table 2 describes the characteristics and frequency of biopsy procedures. A total of 5,730 biopsy procedures were identified in 3,579 (72.7%) patients. Of all procedures, 4,393 (76.7%) were performed in newly diagnosed patients. Among those patients who received ≥ 1 biopsy procedure, 2,143 (59.9%) had only 1 procedure. Samples were most commonly collected by bronchial biopsy (n = 3,183, 55.6%) followed by percutaneous biopsy (n = 1,750, 30.5%). Most biopsy procedures were performed at outpatient visits (n = 4,200, 73.3%). The facility where the biopsy was performed was unknown for 57.7% of biopsy procedures because of incomplete data. Of the 2,426 biopsies for which the type of facility was known, the majority (n = 2,189, 90.2%) were performed in an acute care hospital. Among the personnel performing the biopsy procedures, radiologists (n = 2,120, 36.8%) were the most common, followed by pulmonologists (n = 1,076, 18.8%), physicians (n = 923, 16.1 %), and pathologists (n = 372, 6.5%).

TABLE 2.

Summary of Biopsy Procedures

Total Population (N = 4,926) Previously Diagnosed Patients (n = 1,186) Newly Diagnosed Patients (n = 3,740)
Received a biopsy procedure, n (%) 3,579 (72.7) 800 (67.4) 2,779 (74.3)
   Erlotinib recipients 3,503 798 2,705
   Crizotinib recipients 76 2 74
Number of biopsy procedures per patient, mean (SD) 1.2 (1.1) 1.1 (1.6) 1.17 (1.0)
   Erlotinib recipients 1.17 (1.1) 1.1 (1.6) 1.18 (1.0)
   Crizotinib recipients 1.02 (1.1) 0.5 (0.6) 1.04 (1.1)
Description of biopsy procedures
Total biopsy procedures, n 5,730 1,337 4,393
   Erlotinib recipients 5,602 1,335 4,267
   Crizotinib recipients 128 2 126
Biopsy procedures per patient, n (%)
   0 1,347 (27.3) 386 (32.6) 961 (25.7)
   1 2,143 (43.5) 464 (39.1) 1,679 (44.9)
   2 932 (18.9) 205 (17.3) 727 (19.4)
   3 360 (7.3) 91 (7.7) 269 (7.2)
   ≥ 4 144 (2.9) 40 (3.4) 104 (2.8)
Type of biopsy procedure, n (%)
   Bronchial 3,183 (55.6) 742 (62.6) 2,441 (65.3)
   Percutaneous 1,750 (30.5) 403 (34.0) 1,347 (36.0)
   Fine needle 690 (12) 166 (14.0) 524 (14.0)
   Surgical 36 (0.6) 11 (1.0) 25 (0.7)
   Other 71 (1.2) 15 (1.3) 56 (1.5)
Type of visit for biopsy procedure, n (%)
   Outpatient 4,200 (73.3) 1,040 (87.7) 3,160 (84.5)
   Inpatient 1,299 (22.7) 252 (21.3) 1,047 (28.0)
   Emergency department 10 (0.2) 0 10 (0.3)
   Other 221 (3.9) 45 (3.8) 176 (4.7)
Location of biopsy procedure, n (%)
   Acute care hospital 2,189 (38.2) 467 (39.4) 1,722 (46.0)
   Ambulatory surgery center 50 (0.9) 14 (1.2) 36 (1.0)
   Urgent care facility 3 (0.1) 1 (0.1) 2 (0.1)
   Other 184 (3.2) 54 (4.6) 130 (3.5)
   Unknown 3,304 (57.7) 801 (67.5) 2,503 (66.9)
Type of provider associated with biopsy procedure, n (%)
   Radiologist 2,108 (36.8) 446 (37.6) 1,662 (44.4)
   Pulmonologist 1,071 (18.7) 243 (20.5) 828 (22.1)
   Physician (family practice/internal medicine) 786 (13.7) 195 (16.4) 591 (15.8)
   Pathologist 547 (9.6) 111 (9.4) 436 (11.7)
   Surgeon (thoracic, general, or cardiothoracic) 227 (4.0) 60 (5.1) 167 (4.5)
   Other 548 (9.6) 142 (12.0) 406 (10.9)
   Unknown 443 (7.7) 140 (11.8) 303 (8.1)

SD = standard deviation.

Table 3 describes the summary of biomarker testing procedures. A total of 741 biomarker testing procedures were identified in 675 (13.7%) patients, with 654 and 87 performed in newly and previously diagnosed patients, respectively. Among those patients who received biomarker testing, 614 (91%) had ≥ 1 test. Overall, most biomarker testing procedures (476 of 741) were identified in 435 (of 675) patients after 2011. Frequency of patients receiving EGFR testing increased over the index period with more than 60% of the procedures occurring after 2011 (except for 2012 because of limits on the study period). Most biomarker tests were performed at either an independent laboratory (n = 275, 37.1%) or an outpatient hospital (n = 266, 36%).

TABLE 3.

Summary of Biomarker Testing Procedures

All Patients (N = 4,926) Previously Diagnosed Patients (n = 1,186) Newly Diagnosed Patients (n = 3,740)
Overall population
Patients receiving biomarker testing, n 675 80 595
   Before 2011 240 62 178
   After 2011 435 18 417
Biomarker testing procedures, n 741 87 654
   Before 2011 265 67 198
   After 2011 476 20 456
Mean number of biomarker testing procedures per patient (SD) 0.2 (0.4) 0.1 (0.3) 0.2 (0.4)
   Before 2011 0.05 (0.3) 0.06 (0.3) 0.05 (0.3)
   After 2011 0.10 (0.3) 0.02 (0.1) 0.12 (0.4)
Total biomarker testing procedures per patient, n (%)
   0 4,251 (86.3) 1,150 (93.3) 3,101 (84)
   1 614 (12.5) 75 (6.1) 539 (14.6)
   2 56 (1.1) 7 (0.6) 49 (1.3)
   3 5 (0.1) 0 (0.0) 5 (0.1)
Place of biomarker testing procedures, n (%)
   Independent laboratory 275 (37.1) 26 (2.2) 249 (6.7)
   Outpatient 266 (36) 33 (2.8) 233 (6.2)
   Office/clinic 174 (23.5) 26 (2.2) 148 (4.0)
   Inpatient 17 (2.3) 2 (0.2) 15 (0.4)
   Emergency room 2 (0.3) 0 (0.0) 2 (0.1)
   Other 7 (1.0) 0 (0.0) 7 (0.2)
Erlotinib recipients
Patients receiving biomarker testing procedures, n 634 80 554
 By year, n (%)
   2009 71 (11.2) 34 (2.8) 38 (1.0)
   2010 168 (26.5) 27 (2.3) 141 (3.8)
   2011 226 (35.6) 12 (1.0) 214 (5.7)
   2012 169 (26.7) 6 (0.5) 163 (4.4)
Biomarker testing procedures, n 698 87 611
 By year, n (%)
   2009 83 (11.9) 36 (3.0) 46 (1.2)
   2010 182 (26.1) 30 (2.5) 152 (4.1)
   2011 245 (35.1) 13 (1.1) 232 (6.2)
   2012 188 (26.9) 7 (0.6) 181 (4.8)
Mean number of biomarker testing procedures per patient (SD) 0.2 (0.4) 0.1 (0.3) 0.2 (0.4)
   Before 2011 0.06 (0.3) 0.06 (0.3) 0.05 (0.3)
   After 2011 0.09 (0.3) 0.02 (0.1) 0.11 (0.6)
Total biomarker testing procedures per patient, n (%)
   0 4,167 (84.8) 1,102 (92.9) 3,065 (82.0)
   1 574 (12.0) 73 (6.2) 501 (13.4)
   2 56 (1.2) 7 (0.6) 49 (1.3)
   3 4 (0.1) 0 (0.0) 4 (0.1)
Crizotinib recipients
Patients receiving biomarker testing procedures, n 41 0 41
Biomarker testing procedure, n 43 0 43
Mean number of biomarker testing procedure per patient (SD) 0.3 (0.5) 0 (0.0) 0.4 (0.5)
Total biomarker testing procedures per patient, n (%)
   0 84 (67.2) 4 (100.0) 80 (66.1)
   1 40 (32.0) 0 (0.0) 40 (33.1)
   2 0 (0.0) 0 (0.0) 0 (0.0)
   3 1 (0.8) 0 (0.0) 1 (0.1)

SD = standard deviation.

Table 4 describes the cost of biopsy procedures among patients receiving at least 1 biopsy procedure, categorized by setting (inpatient vs. outpatient) and by perspective (payer vs. patient). Overall mean (SD) biopsy procedure cost was $1,223 ($1,899) from the payer and $60 ($147) from the patient perspective in the outpatient setting. Incremental increases in per patient cost were observed with an increasing number of biopsy procedures. Mean (SD) cost for a biopsy procedure in erlotinib recipients was $60 ($147) and $1,220 ($1,896) from the patient and payer perspectives, respectively. In crizotinib recipients, mean (SD) costs were $70 ($145) and $1,340 ($2,035), respectively. Costs of the biopsy procedures in the outpatient setting were higher in newly diagnosed patients compared with previously diagnosed patients regardless of whether they received erlotinib or crizotinib. In the inpatient setting, mean (SD) biopsy procedure cost was $8,163 ($18,712) from the payer and $179 ($693) from the patient perspective. Similar to the outpatient setting, per patient costs incrementally increased with the increase in the number of biopsy procedures.

TABLE 4.

Cost of Biopsy Procedures in Patients with ≥ 1 Biopsy Procedure by Outpatient and Inpatient Setting

Total Population (N = 3,579) Previously Diagnosed Patients (n = 800) Newly Diagnosed Patients (n = 2,779)
Outpatient Inpatient Outpatient Inpatient Outpatient Inpatient
Total cost per patient, $
Copaya
All patients, n 3,031 973 709 183 2,322 790
   Mean (SD) 60 (147) 180 (691) 50 (151) 216 (742) 63 (146) 172 (679)
   Median 4 0 2 0 5 0
Erlotinib recipients, n 2,967 950 707 183 2,260 767
   Mean (SD) 60 (147) 179 (693) 50 (151) 216 (742) 63 (146) 170 (681)
   Median 4 0 2 0 5 0
Crizotinib recipients, n 64 23 2 0 62 23
   Mean (SD) 70 (145) 245 (610) 4 (6) NA 72 (147) 245 (610)
   Median 8 0 4 NA 11 0
Net payb
All patients, n 3,031 973 709 183 2,322 790
   Mean (SD) 1,223 (1,899) 8,163 (18,712) 1,127 (1,749) 12,409 (23,523) 1,252 (1,942) 7,180 (17,279)
   Median 534 331 491 473 542 322
Erlotinib recipients, n 2,967 950 707 183 2,260 767
   Mean (SD) 1,220 (1,896) 8,248 (18,874) 1,130 (1,751) 12,409 (23,523) 1,248 (1,939) 7,256 (17,457)
   Median 534 327 492 473 540 314
Crizotinib recipients, n 64 23 2 0 62 23
   Mean (SD) 1,340 (2,035) 4,651 (9,540) 36 (50) NA 1,382 (2,054) 4,651 (9,540)
   Median 694 574 36 NA 739 574
Cost per patient by number of biopsy procedures, $
1 procedure, n 1,911 861 431 166 1,480 695
Copay
   Mean (SD) 41 (114) 180 (713) 29 (91) 227 (776) 45 (119) 168 (698)
   Median 1 0 0 0 2 0
Net pay
   Mean (SD) 778 (1,194) 7,339 (16,879) 693 (1,030) 11,217 (22,199) 803 (1,237) 6,413 (15,213)
   Median 342 286 317 428 350 280
2 procedures, n 777 99 181 14 596 85
Copay
   Mean (SD) 84 (181) 167 (457) 79 (215) 116 (205) 86 (169) 175 (487)
   Median 13 0 4 5 17 0
Net pay
   Mean (SD) 1,581 (2,070) 12,954 (26,350) 1,309 (1,517) 17,169 (29,068) 1,664 (2,205) 12,260 (25,996)
   Median 839 689 696 1,541 873 685
3 procedures, n 250 13 68 3 182 10
Copay
   Mean (SD) 108 (207) 317 (679) 90 (219) 71 (122) 114 (203) 391 (765)
   Median 20 0 17 0 24 0
Net pay
   Mean (SD) 2,530 (2,742) 26,246 (41,013) 2,386 (2,854) 56,184 (32,492) 2,584 (2,705) 17,265 (40,246)
   Median 1,552 2,038 1,337 59,956 1,675 1,338

Note: Cost and SD are rounded to the nearest whole number.

a Copay corresponds to the cost in patient perspective.

b Net pay corresponds to the cost in payer perspective.

NA = not applicable; SD = standard deviation.

Table 5 shows the costs of biomarker testing in patients who received at least 1 test. Overall per patient mean (SD) cost was $43 ($229) and $891 ($1,062) from the patient and payer perspectives, respectively. For erlotinib recipients, per patient mean (SD) cost was $42 ($228) from the patient and $906 ($1,084) from the payer perspective, whereas it was $55 ($243) and $664 ($576) in crizotinib recipients from patient and payer perspectives, respectively. For patients receiving 1 biomarker test, per patient mean (SD) cost was $43 ($236) from patient and $819 ($932) payer perspectives. For patients receiving 2 tests, per patient mean (SD) cost was $38 ($159) from patient and $1,640 ($1,854) payer perspectives.

TABLE 5.

Cost of Biomarker Testing in Patients with ≥1 Biomarker Testing Procedure

Total Population (N = 675) Previously Diagnosed Patients (n = 80) Newly Diagnosed Patients (n = 595)
Total cost per patient, $
Copaya
All patients, n 675 80 595
   Mean (SD) 43 (229) 34 (147) 44 (238)
   Median 0 0 0
Erlotinib recipients, n 634 80 554
   Mean (SD) 42 (228) 34 (147) 43 (238)
   Median 0 0 0
Crizotinib recipients, n 41 0 41
   Mean (SD) 55 (243) NA 55 (243)
   Median 0 NA 0
Net payb
All patients, n 675 80 595
   Mean (SD) 891 (1,062) 827 (932) 900 (1,079)
   Median 551 395 599
Erlotinib recipients, n 634 80 554
   Mean (SD) 906 (1,084) 827 (932) 917 (1,105)
   Median 549 395 600
Crizotinib recipients, n 41 0 41
   Mean (SD) 664 (576) NA 664 (576)
   Median 563 NA 563
Cost per patient by no. of biomarker testing procedures, $
1 procedure, n 614 73 541
Copay
   Mean (SD) 43 (236) 34 (153) 45 (245)
   Median 0 0 0
Net pay
   Mean (SD) 819 (932) 845 (946) 816 (931)
   Median 513 400 524
2 procedures, n 56 7 49
Copay
   Mean (SD) 38 (159) 27 (60) 40 (169)
   Median 0 1 0
Net pay
   Mean (SD) 1,640 (1,854) 635 (807) 1,784 (1,921)
   Median 1,317 241 1,384
3 procedures, n 5 0 5
Copay
   Mean (SD) 0 NA 0
   Median 0 NA 0
Net pay
   Mean (SD) 1,368 (764) NA 1,368 (764)
   Median 1,608 NA 1,608

Note: Cost and SD are rounded to the nearest whole number.

a Copay corresponds to the cost in patient perspective.

b Net pay corresponds to the cost in payer perspective.

NA = not applicable; SD = standard deviation.

Discussion

This study analyzed the utilization patterns of and costs associated with biopsy procedures and biomarker testing in patients with metastatic lung cancer receiving erlotinib or crizotinib using a U.S. claims database. A steady increase in the number of erlotinib or crizotinib recipients over the course of the index period was observed. Results in crizotinib recipients should be interpreted with caution because of the small sample size, which was a result of the limited time between crizotinib approval in August 2011 and the end of the index period in September 2012.

Biopsy procedures were identified for only 73.0% of patients. It is possible that an archival tissue sample may have been used in patients without an identifiable biopsy CPT code. Approximately one third of patients (29.1%) received more than 1 biopsy procedure over the course of their disease, especially around the index date. Inadequate tissue sample may have prompted another biopsy procedure in these patients. Some studies have highlighted this issue and reported that only about 50% of tissue samples are adequate for biomarker testing.13,29,30 Moreover, a study conducted by Ellis et al. (2013) that investigated challenges in implementing EGFR testing in Canada identified inadequate diagnostic tissue for molecular testing as one of the major challenges.31 An international survey conducted among oncologists showed that in addition to inadequate tissue, poor performance status and long turnaround time were the main reasons for not testing.32

Although guidelines recommend that EGFR and ALK testing be done before starting treatment, we found that only 13.2% of erlotinib and 32.8% of crizotinib recipients received biomarker testing at any time. In erlotinib recipients, most patients received biomarker testing after 2011 (n = 395, 62.3%) but still the mean (SD) number of biomarker testing/patient was low (0.09 [0.3]). EGFR assay has been commercially available since 2005, but it became part of the guidelines only in early 2011, which shows delayed uptake of EGFR testing.33,34 A study examining hospital use of the EGFR assay showed that in 2010 only 12% of U.S. acute care hospitals ordered the EGFR assay, suggesting most lung cancer patients did not have access to this test.35 We did observe an increase in the number of patients receiving EGFR testing from 2009 onward, indicating a steady adoption of the guideline recommendations. Erlotinib recipients without any identifiable biomarker testing claim may have been treated based on clinical characteristics associated with EGFR mutation (e.g., nonsmoker, female, and Asian) in the absence of biomarker testing, since guidelines were still evolving during the index period of this study. As anticipated, we found more newly diagnosed patients receiving biomarker testing than previously diagnosed patients (16% vs. 6.7%) as these drugs are approved for first-line treatment. Our results are consistent with another analysis conducted using integrated medical and pharmaceutical data from 4 Blue Cross and Blue Shield plans, where only 36 of 125 (28.8.%) erlotinib recipients received a biomarker test in 2011.28 A multidisciplinary approach may be warranted to overcome the challenges in implementing recommended biomarker tests.36

The cost of biopsy and biomarker testing procedures varied by the setting. The mean biopsy procedure cost in the inpatient setting ($8,163) was consistent with that observed in a retrospective cohort study using the 5% Medicare random national sample data during 2009-2011 ($8,869 without adverse events).37 Unlike our study, this study did not look at the cost exclusively in patients receiving targeted therapies. The variability in the cost for biomarker testing might be explained by the lack of uniformity in the use of CPT codes while submitting claims for reimbursement during the index period.

Limitations

The results of this study should be considered in the context of several limitations. Because there is no specific ICD-9-CM code for NSCLC, patients with metastatic lung cancer receiving erlotinib or crizotinib were assumed to have non-small cell histology. Since biomarker test results (positive or negative) were not available in the claims database, we were unable to evaluate if the treatment was related to the test outcome. It is also difficult to accurately identify the biomarker testing procedures, since they are not consistently captured in the claims database. Different CPT codes are used for billing purposes of relatively similar biomarker tests. Of note, the CPT code for EGFR testing has been available since 2013, although a code for ALK testing is still lacking. Considering the scope of the current database, the link between the timing of the biomarker testing and the corresponding biopsy procedure performed to obtain the tissue sample cannot be established. A biopsy may have been performed for relapses or treatment change or as a part of a diagnostic work-up, but these reasons cannot be assessed from the database. Also, the reimbursed procedures and tests that were not part of this database are not included in the analysis. Because our study was restricted to patients receiving erlotinib or crizotinib, the results of this study cannot be generalized to all patients with metastatic lung cancer because these patients have different clinical presentation. Finally, because the MarketScan database does not capture information on smoking status or histology, we were not able to clearly identify factors associated with EGFR testing.

Conclusions

This study provides insight into utilization patterns and the cost of biopsy procedures and biomarker testing in patients with metastatic lung cancer. Despite inclusion in the treatment guidelines, the low frequency of biomarker testing after 2011 suggests a need for increased awareness of the importance of biomarker testing for guiding treatment decisions in these patients. Biopsy procedures and biomarker testing incurred significant costs in patients with metastatic lung cancer receiving targeted therapies, which suggests that they have a tangible economic impact.

Acknowledgments

Medical writing and editorial assistance was provided by Melanie Leiby, PhD, of Merck & Co and was funded by Merck & Co.

APPENDIX A. Biopsy-Related CPT and ICD-9-CM Codes

Type of Biopsy CPT (or ICD-9-CM Procedure) Code
Fine needle biopsy
   Fine needle aspiration; without imaging guidance 10021
   Fine needle aspiration; with imaging guidance 10022
   Closed (percutaneous needle biopsy of lung (FNA of lung or transthoracic needle biopsy of lung) 33.26 (ICD-9-CM)
Percutaneous biopsy
   Biopsy, lung or mediastinum, percutaneous needle 32405
   Biopsy, pleura; percutaneous needle 32400
Surgical biopsy
   Thoracotomy, limited, for biopsy of lung or pleura 32905
   Thoracotomy, with diagnostic biopsy(ies) of lung infiltrate(s) (e.g., wedge, incisional), unilateral 32096
   Thoracotomy, with diagnostic biopsy(ies) of lung nodule(s) or mass(es) (e.g., wedge, incisional), unilateral 32097
   Thoracotomy, with biopsy(ies) of pleura 32098
   Open biopsy of lung 33.28 (ICD-9-CM)
Bronchial biopsy
   Bronchoscopy, rigid or flexible, including fluoroscopic guidance, when performed; with bronchial or endobronchial biopsy(s), single or multiple sites 31625
   Bronchoscopy, rigid or flexible, including fluoroscopic guidance, when performed; with brushing or protected brushings 31623
   Bronchoscopy, rigid or flexible, including fluoroscopic guidance, when performed; with bronchial alveolar lavage 31624
   Bronchoscopy with transbronchial lung biopsy, single lobe 31628
   Bronchoscopy with transbronchial needle aspiration biopsy(s) trachea, main stem and/or lobar bronchus 31629
   Bronchoscopy with transbronchial lung biopsy(s), each additional lobe (list separately in addition to code for primary procedure) use with 31628 31632
   Bronchoscopy with transbronchial needle aspiration biopsy(s) each additional lobe (list separately in addition to code for primary procedure) use with 31629 31633
   Closed endoscopic biopsy of lung (fiber-optic (flexible) bronchoscopy with fluoroscopic guidance with biopsy or transbronchial lung biopsy) 33.27 (ICD-9-CM)
Other
   Thoracoscopic lung biopsy 33.20
   Closed [endoscopic] biopsy of bronchus 33.24
   Open biopsy of bronchus 33.25
Non-biopsy-specific procedures
   Ultrasound guidance for needle placement 76942
   Fluoroscopic guidance for needle placement 77002
   CT guidance for needle placement 77012
   MR guidance for needle placement 77021

CPT = Current Procedural Terminology; CT = computed tomography; FNA = fine needle aspiration; ICD-9-CM = International Classification of Diseases, Ninth Revision Clinical Modification; MR = magnetic resonance.

APPENDIX B. Biomarker Testing CPT Codes

CPT Code Description CPT Code
Molecular diagnostics; molecular isolation or extraction, each nucleic acid type (i.e., DNA or RNA) 83890
Isolation or extraction of highly purified nucleic acid, each nucleic acid type (i.e., DNA or RNA) 83891
Enzymatic digestion, each enzyme treatment 83892
Dot/slot blot production, each nucleic acid preparation 83893
Separation by gel electrophoresis (e.g., agarose, polyacrylamide), each nucleic acid preparation 83894
Nucleic acid probe, each 20 83896
Nucleic acid transfer (e.g., Southern, Northern), each nucleic acid preparation 83897
Amplification, target, each nucleic acid sequence 83898
Amplification, target, multiplex, first 2 nucleic acid sequences 83900
Amplification, target, multiplex, each additional nucleic acid sequence beyond 2 (list separately in addition to code for primary procedure) 83901
Reverse transcription 83902
Mutation scanning, by physical properties (e.g., single strand conformational olymorphisms [SSCP], heteroduplex, denaturing gradient gel electrophoresis [DGGE], RNA’ase A), single segment, each 83903
Mutation identification by sequencing, single segment, each segment 83904
Mutation identification by allele specific transcription, single segment, each segment 83905
Mutation identification by allele specific translation, single segment, each segment 83906
Lysis of cells prior to nucleic acid extraction (e.g., stool specimens, paraffin embedded tissue), each specimen 83907
Amplification, signal, each nucleic acid sequence 83908
Separation and identification by high resolution technique (e.g., capillary electrophoresis), each nucleic acid preparation 83909
Interpretation and report 83912
RNA stabilization 83913
Mutation identification by enzymatic ligation or primer extension, single segment, each segment (e.g., oligonucleotide ligation assay [OLA], single base chain extension [SBCE], or allele-specific primer extension [ASPE]) 83914
Array-based evaluation of multiple molecular probes; 11 through 50 probes 88384
51 through 250 probes 88385
251 through 500 probes 88386

CPT = Current Procedural Terminology.

APPENDIX C. Molecular Pathology CPT Code Changes Starting January 2013

Latest Update Test ID Second ID Test Name Current CPT Code New CPT Code
1/1/2013 20450 20450 FLT3 Mutation Analysis 83892, 83900, 83909 81245
1/1/2013 20451 20451 MPL Exon 10 Mutation Detection, Bone Marrow 83898, 83904x2, 83909x2 81403
1/1/2013 20452 20452 JAK2 Exon 12 Mutation Detection, Bone Marrow 83891, 83898, 83902, 83904x2, 83909x2 81403
1/1/2013 20458 20458 BCR/ABL, p190, Quant, Monitor 83891, 83896x2, 83898x2, 83902 81207
1/1/2013 20459 20459 PML/RARA Quantitative, PCR 83891, 83896, 83898x2, 83902 81315
1/1/2013 20463 20463 KIT Asp816Val Mutation Analysis 83898x2, 83909x2 81402
1/1/2013 20464 20464 BCR/ABL, RNA-Qual, Diagnostic 83891, 83900x2, 83901x10, 83902, 83914x2 81206 81207 81208
1/1/2013 20465 20465 BCR/ABL, p210, Quant, Monitor 83891, 83896x2, 83898x2, 83902 81206
1/1/2013 20471 20471 JAK2 Exon 12 Mutation Detection, B 83891, 83898, 83902, 83904x2, 83909x2 81403
1/1/2013 20472 20472 Nucleophosmin Mutation Analysis 83898, 83909 81401
1/1/2013 20473 20473 BCR/ABL Mutation, ASPE 83891, 83894x2, 83896x8, 83898x2, 83902, 83909, 83914x8 81403
1/1/2013 20474 20474 MPL Exon 10 Mutation Detection, B 83898, 83904x2, 83909x2 81403
1/1/2013 60142 60142 Circulating Tumor Cells, Prostate 0279T, 0280T 86152, 86153
1/1/2013 60694 60694 FOXL2 Mutation (C402G) Analysis by PCR and Pyrosequencing 83891, 83892, 83896x2, 83898, 83904, 83907, 83912 81479
1/1/2013 61207 61207 IDH1/IDH2 Genes, Known Mutations 83896x4, 83898x2, 83904x2, 83912 81403, 81403
7/1/2013 83361 83361 Synovial Sarcoma, RT-PCR, Paraffin 81479 81401x2
1/1/2013 83363 83363 Ewings Sarcoma, RT-PCR, Paraffin 83894, 83896x5, 83898x2, 83902, 83912 81401x2
1/1/2013 83365 83365 83894, 83896x2, 83898, 83902, 83912 81401x2
1/1/2013 83367 83367 Alveolar Rhabdomyosarcoma, RT-PCR,PF 83894, 83896x3, 83898x2, 83902, 83912 81401
1/1/2013 88955 88955 KIT, Mutation Analysis, Ex8 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 88956 88956 KIT, Mutation Analysis, Ex13 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 88957 88957 KIT, Mutation Analysis, Ex17 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 88958 88958 PDGFRA, Mutation Analysis, Ex14 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 89045 89045 BRAF Mutation (T1799A) Analysis 83896x2, 83898, 83904, 83912 81210
1/1/2013 89089 89089 Circulating Tumor Cells, Breast, B 0279T, 0280T 86152, 86153
1/1/2013 89162 89162 Circulating Tumor Cells, Colorectal 0279T, 0280T 86152, 86153
1/1/2013 89669 89669 KIT, Mutation Analysis, Ex9 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 89670 89670 KIT, Mutation Analysis, Ex11 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 89671 89671 PDGFRA, Mutation Analysis, Ex12 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 89672 89672 PDGFRA, Mutation Analysis, Ex18 83896x2, 83898, 83904, 83912, 83892, 83907, 83891 81404
1/1/2013 1A2 89401 CYP1A2 Genotype 83892x2, 83900x2, 83901, 83912, 83914x13 81479
1/1/2013 1A2O 60346 CYP1A2 Genotype, Saliva 83892x2, 83900x2, 83901, 83912, 83914x13 81479
1/1/2013 1STT 87857 First Trimester Maternal Screen 84163, 84702 81508
1/1/2013 2C19O 60335 CYP2C19 Genotype, Saliva 83892, 83894, 83900, 83901x3, 83909x10, 83912 81225
1/1/2013 2C19S 60439 CYP2C19 Sequence Genotype 83892, 83894, 83900, 83901x3, 83909x10, 83912 81225
1/1/2013 2C9S 60528 CYP2C9 Sequence Genotype 83892, 83894, 83900, 83901, 83909x6, 83912 81227
1/1/2013 2C9SO 60337 CYP2C9 Genotype, Saliva 83892, 83894, 83900, 83901, 83909x6, 83912 81227
1/1/2013 2D6 83180 CYP2D6 Genotype 83892, 83900, 83901x2, 83912, 83914x17 81226
1/1/2013 2D6O 60334 CYP2D6 Genotype, Saliva 83892, 83900, 83901x2, 83912, 83914x17 81226
1/1/2013 2D6T 87966 CYP2D6 Genotype - Tamoxifen Therapy 83892, 83900, 83901x2, 83912, 83914x17 81226
1/1/2013 2D6TO 60340 CYP2D6 Tamoxifen Genotype, Saliva 83892, 83900, 83901x2, 83912, 83914x17 81226
1/1/2013 3A4B 61241 CYP3A4 Genotype, B 83896x2, 83898, 83912 81401
1/1/2013 3A4O 61242 CYP3A4 Genotype, Saliva 83896x2, 83898, 83912 81401
1/1/2013 A1AT 82993 Alpha-1-Antitrypsin Genotyping, Blood 83890, 83896x2, 83898x2, 83912 81332
1/1/2013 A1ATR 83050 A1AT Deficiency Profile 82103, 83890, 83896x2, 83898x2, 83912 81332 82103
1/1/2013 ACVK 89393 ACVRL1 Gene, Known Mutation 83892, 83894, 83898, 83909x2, 83912 81479
1/1/2013 ADHP 83375 FH/ADH Genetic Reflex Panel 83892, 83898, 83912, 83914 81479
1/1/2013 AGPB 9499 Alpha-Globin Gene Analysis 83891, 83894, 83900x2, 83909, 83912, 83914x15 81257
1/1/2013 AGPBT 31032 Alpha-Globin Gene Analysis 83891, 83894, 83900x2, 83909, 83912, 83914x15 81404
1/1/2013 AGXKM 89916 AGXT Gene, Known Mutation 83891, 83912 81479
1/1/2013 AGXMS 89915 AGXT Gene, Full Gene Analysis 83891, 83898x12, 83909x24, 83912 81479
1/1/2013 AGXT 83643 AGXT Mutation Analysis (G170R) 83890, 83892, 83894, 83898, 83912 81479
1/1/2013 AJPWO 88887 Ashkenazi Jewish Mutation Analysis Panel Without Cystic Fibrosis (CF) 83890, 83892, 83896x31, 83900, 83909, 83912, 83914x31 81200, 81330, 81290, 81260, 81255, 81251, 81242, 81209
1/1/2013 AMYKM 83705 Amyloidosis, Transthyretin-Associated Familial, Known Mutation 83890, 83898, 83909x2, 83912 81403
1/1/2013 AMYL 83667 Familial Amyloidosis, DNA Sequence 83890, 83898x4, 83909x8, 83912 81404
1/1/2013 APO1K 60724 APOA1 Gene, Known Mutation 83890, 83898, 83909x2, 83912 81479
1/1/2013 APO1S 60723 APOA1 Gene, Full Gene Analysis 83890, 83898x5, 83909x10, 83912 81479
1/1/2013 APO2K 60726 APOA2 Gene, Known Mutation 83890, 83898, 83909x2, 83912 81479
1/1/2013 APO2S 60725 APOA2 Gene, Full Gene Analysis 83890, 83898x4, 83909x8, 83912 81479
1/24/2013 APOB 89097 APOB Genotype 83892, 83898, 83912, 83914 81401
1/1/2013 APOE 80905 Apolipoprotein E Genotyping, B 83890, 83892, 83894, 83898, 83912 81401
7/1/2013 ARPKD 88911 ARPKD Mutation Screen 81479 81408
7/1/2013 ARPKM 88912 ARPKD Known Mutation 81479 81403
7/1/2013 ARSAK 61260 ARSA Gene, Known Mutation 81479 81403
7/1/2013 ARSAS 61259 ARSA Gene, Full Gene Analysis 81479 81405
1/1/2013 BA190 83336 BCR/ABL, p190, Quant, Monitor 83891, 83896x2, 83898x2, 83902 81207
1/1/2013 BADX 89006 BCR/ABL, RNA-Qual, Diagnostic 83891, 83900x2, 83901x10, 83902, 83914x2 81206, 81207, 81208
1/1/2013 BAKDM 89609 BCR/ABL Mutation, ASPE 83891, 83894x2, 83896x8, 83898x2, 83902, 83909, 83914x8 81403
1/1/2013 BCGBM 31141 Immunoglobulin Gene Rearrange, BM 83912, 83891, 83900, 83901x4, 83909x4 81262 81264
1/1/2013 BCGR 83123 Immunoglobulin Gene Rearrange, B Varies 81262 81264
1/1/2013 BCGRV 31142 Immunoglobulin Gene Rearrange, V 83912, 83891, 83900, 83901x4, 83909x4 81262 81264
1/1/2013 BCLL 89008 IGH Somatic Hypermutation in B-CLL 83891, 83898x7, 83902, 83904x2, 83909x8 81263
1/1/2013 BCRAB 89007 BCR/ABL, p210, Quant, Monitor 83891, 83896x2, 83898x2, 83902 81206
1/1/2013 BGDD 89575 Beta Globin Gene Del/Dup 83900, 83909, 83914x8 81403
1/1/2013 BGLOB 83287 Beta Globin Gene, Large Del/Dup 83900, 83909, 83914x8 81403
7/1/2013 BIL28 61702 IL28B Polymorphism Genotype, B 81479 81400
1/1/2013 BLM 85316 Bloom Syndrome, Mutation Analysis, 2281del6/ins7 83890, 83892, 83896, 83900, 83909, 83912, 83914 81209
1/1/2013 BRAF 87980 BRAF Mutation Analysis (V600E), Tumor 83898, 83909x2, 83912 81210
1/1/2013 BRAFM 83837 BRAF Mutation Analysis (V600), Melanoma 83890, 83896x2, 83898, 83912, 88387 81210
1/1/2013 BTDKM 89013 BTD Gene, Known Mutation 83890, 83892x2, 83894x2, 83898x2, 83909x4, 83912 81403
1/1/2013 BTDMS 89012 BTD Gene, Full Gene Analysis 83891, 83898x7, 83909x14, 83912 81404
1/1/2013 BTKFP 89742 BTK Full-Gene Panel, B 83892x14, 83894x7, 83898x8, 83900x6, 83909x42, 83912, 83890, 88184 81479, 88184
7/1/2013 BTKK 89306 BTK Gene, Known Mutation 81479 81403
7/1/2013 BTKKM 29304 BTK Gene, Known Mutation 81479 81403
7/1/2013 BTKS 89307 BTK, Full Gene Sequence 81479 81406
7/1/2013 BTKSP 29305 BTK, Full Gene Sequence 81479 81406
1/1/2013 BWSRS 61010 BWS/RSS Molecular Analysis 83891, 83892, 83900, 83909x2, 83912, 83914x26 81401
7/1/2013 CACTK 61195 SLC25A20 Gene, Known Mutation 81479 81403
7/1/2013 CACTS 61194 SLC25A20 Gene, Full Gene Analysis 81479 81405
1/1/2013 CANW 81780 Canavan Disease, Mutation Analysis, ASPA 83890, 83892, 83896x5, 83900, 83909, 83912, 83914x5 81200
1/1/2013 CDKKM 60229 CDKN1C Gene, Known Mutation 83891, 83898, 83909x2, 83912 81479
1/1/2013 CDKMS 60228 CDKN1C Gene, Full Gene Analysis 83891, 83898x6, 83909x12, 83912 81479
1/1/2013 CELI 88906 Celiac Associated HLA-DQ Typing 86816 81376x2
1/1/2013 CFPB 9497 Cystic Fibrosis Mutation Analysis, 106-Mutation Panel 83788, 83890, 83900x8, 83901x89, 83912, 83914x106 81220
1/1/2013 CFTRK 88880 CFTR Gene, Known Mutation 83891, 83892x2, 83894x2, 83898x2, 83909x4, 83912 81221
1/1/2013 CFTRM 88876 CFTR Gene, Full Gene Analysis 83891, 83898x30, 83909x60, 83912 81223
1/1/2013 CGH 88898 aCGH, Whole Genome, Constitutional 88399 81228
1/1/2013 CHEP 84427 Chimerism-Recipient Engraft Varies 81267
1/1/2013 CHIDB 83182 Chimerism Donor Varies 81265
1/1/2013 CHRGB 83186 Chimerism Pre Recip Varies 81265
1/1/2013 COMT 83301 Catechol Methyltransferase Genotype 83892, 83898, 83912, 83914x2 81479
1/1/2013 COMTO 60336 COMT Genotype, Saliva 83892, 83898, 83912, 83914x2 81479
1/1/2013 CPOXK 61264 CPOX Gene, Known Mutation 83891, 83898, 83909x2, 83912 81479
1/1/2013 CPOXS 61263 CPOX Gene, Full Gene Analysis 83891, 83898x8, 83909x16, 83912 81479
1/1/2013 CPTKM 61121 CPT2 Gene, Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 CPTMS 61120 CPT2 Gene, Full Gene Analysis 83891, 83898x8, 83909x16, 83912 81404
7/1/2013 CSRKM 83704 CASR Known Mutation 81479 81403
7/1/2013 CSRMS 83703 CASR Mutation Screen 81479 81405
1/1/2013 CYPKP 89082 CYP21A2 Known Mutation 83892x2, 83894, 83900, 83901x2, 83909x2, 83912 81403
1/1/2013 CYPSP 89081 CYP21A2 Full Gene Analysis 83892x2, 83894, 83900, 83901x2, 83909x16, 83912 81405
1/1/2013 DISI 89185 HLA Class I Mol Typing DiseaseAssoc 83890, 83894, 83898, 83912 81372
1/1/2013 DISII 32864 HLA ClassII Mol Typing DiseaseAssoc 83890, 83894, 83898, 83912 81375
1/1/2013 DRD3 81776 DRD3 Genotype 83892, 83900, 83912, 83914 81479
1/1/2013 DRD3O 60342 DRD3 Genotype, Saliva 83892, 83900, 83912, 83914 81479
1/1/2013 DRD4 89096 Dopamine Receptor D4 Genotype, B 83894, 83898, 83912 81479
1/1/2013 DRD4O 60344 DRD4 Genotype, Saliva 83894, 83898, 83912 81479
1/1/2013 DRPLA 81801 DRPLA Gene Analysis 83890, 83898, 83909, 83912 81401
7/1/2013 EALDD 31719 ENG and ACVRL1, Large Del/Dup 81479 81405, 81479
1/1/2013 EGFR 61247 EGFR Gene, Mutation Analysis, 29 Mutation Panel, Tumor 83890, 83896x7, 83898x7, 83912 81235
7/1/2013 ENGK 89391 ENG Gene, Known Mutation 81479 81403
7/1/2013 ENGKM 31047 ENG Gene, Known Mutation 81479 81403
1/1/2013 EPOR 61679 EPOR Gene, Mutation Analysis, B 83900, 83904x4, 83909x4 81479
1/1/2013 F5DNA 81419 Factor V Leiden (R506Q) Mutation, B 83891, 83892, 83896x5, 83903, 83908x2, 83912 81241
1/1/2013 FABKM 88266 Fabry Disease Known Mutation 83890, 83892, 83894, 83898, 83909x2, 83912 81403
1/1/2013 FABMS 88264 Fabry Disease Full Gene Analysis 83891, 83898x8, 83909x16, 83912 81405
1/1/2013 FANCA 85318 Fanconi Anemia C Mutation Analysis, IVS4(+4) A->T and 322delG 83890, 83892, 83896x2, 83900, 83909, 83912, 83914x2 81242
1/1/2013 FAPG 91347 ADmark ApoE Genotype Analysis & Interpretation (Symptomatic) 83891, 83892x4, 83896x10, 83908x4, 83912 81401
1/1/2013 FAPKM 83001 Familial Adenomatous Polyposis (FAP) Known Mutation 83891, 83912 81202
5/1/2013 FAPMS 82582 Familial Adenomatous Polyposis (FAP) Mutation Screen 83891, 83894x4, 83898x21, 83905x4, 83906x4, 83909x34, 83912 81201, 81203
1/1/2013 FBKM 89311 FBN1 Gene, Known Mutation 83892, 83898, 83909x2, 83912 81403
1/1/2013 FBN1 89308 FBN1, Full Gene Sequence 83892x62, 83898x62, 83909x122, 83912 81408
1/1/2013 FBNN 89314 Neonatal Marfan Syndrome, FBN1 Gene 83892x9, 83898x9, 83909x18, 83912 81479
1/1/2013 FBNNP 29301 Neonatal Marfan Syndrome, FBN1 Gene 83892x7, 83898x7, 83912 81479
1/1/2013 FCCEV 57461 Complete CADASIL Evaluation 83891, 83898x23, 83904x23, 83909x23, 83912 81406
1/11/2013 FCMD 91452 Complete Myotonic Dystrophy Evaluation 83891, 83898x2, 83909x2, 83912 81404, 81479
1/1/2013 FD 85319 Familial Dysautonomia, Mutation Analysis, IVS20(+6T->C) and R696P 83890, 83892, 83896x2, 83900, 83909, 83912, 83914x3 81260
1/1/2013 FECHK 60372 FECH Gene, Known Mutation 83891, 83898, 83909x2, 83912 81479
1/1/2013 FECHS 60371 FECH Gene, Full Gene Analysis 83891, 83898x11, 83909x22, 83912 81479
1/1/2013 FFRED 91819 Friedreich Ataxia Repeat Expansion Analysis -Unknown Mutation 83891, 83892x2, 83894x3, 83896, 83897, 83898x2, 83912 81479
1/1/2013 FGAKM 60722 FGA Gene, Known Mutation 83890, 83898, 83909x2, 83912 81479
1/1/2013 FGAMS 60721 FGA Gene, Full Gene Analysis 83890, 83898x10, 83909x20, 83912 81479
1/1/2013 FIXKM 84320 Factor IX Gene Known Mutation 83891, 83892, 83894, 83898, 83912 81403
1/1/2013 FIXMS 84209 Factor IX Gene Mutation Screening 83891, 83892x3, 83894x8, 83900x3, 83901x2, 83912 81405
1/1/2013 FLIMB 91635 Limb Girdle Muscular Dystrophy Evaluation 83891, 83898x133, 83900x3, 83901x62, 83904x133, 83909x136, 83912, 83914x68 81404
1/1/2013 FLT 19739 FLT3 Mutation Analysis 83892, 83900, 83909 81245
1/1/2013 FPMP 91590 PMP22 Duplication/Deletion DNA 83891, 83900, 83901x31, 83909, 83912, 83914x33 81324
1/1/2013 FPRTG 91565 Prometheus TPMT Genetics 83896x6, 83898x3, 83907, 83912 81401
1/1/2013 FXFU 61118 Fragile X, Follow Up Analysis 83892x2, 83893x2, 83894x2, 83896, 83897x2 81244
1/1/2013 FXPB 9569 FragilexSyndrome, Molecular Analysis 83890, 83900, 83909, 83912 81243
1/1/2013 GAAKM 89897 Pompe Disease, Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 GAAMS 89898 Pompe Disease, Full Gene Sequencing 83891, 83898x19, 83909x38, 83912 81406
1/1/2013 GAL3 86202 Galectin-3, S 83520 82777
1/1/2013 GAL6 84366 Galactosemia Gene Analysis 83890, 83896x6, 83898x6, 83912 81401
7/1/2013 GALCK 60695 Krabbe Disease, Known Mutation 81479 81403
7/1/2013 GALCS 60696 Krabbe Disease, Full Gene Analysis 81479 81406
1/1/2013 GALTK 84367 Galactosemia Known Mutation 83890, 83894x2, 83898x2, 83909x4, 83912 81403
1/1/2013 GALTM 88877 GALT Gene, Full Gene Analysis 83891, 83898x9, 83909x18, 83912 81406
1/1/2013 GAUW 81235 Gaucher Disease, Mutation Analysis, GBA 83890, 83892, 83896x8, 83900, 83909, 83912, 83914x8 81251
1/1/2013 GBAKM 60712 Gaucher Disease, Known Mutation 83891, 83892, 83894, 83898, 83909x4, 83912 81403
1/1/2013 GBAMS 60711 Gaucher Disease, Full Gene Analysis 83891, 83892, 83894, 83898, 83909x22, 83912 81479
1/1/2013 GFDKM 89899 FTCD Gene, Known Mutation 83891, 83898x2, 83909x4, 83912 81479
1/1/2013 GFDMS 89900 FTCD Gene, Full Gene Analysis 83891, 83898x14, 83909x28, 83912 81479
7/1/2013 GRNKM 89187 Progranulin Gene, Known Mutation 81479 81403
7/1/2013 GRNMS 89188 Progranulin Gene, Full Gene Analysis 81479 81406
1/1/2013 GSNKM 60718 GSN Gene, Known Mutation 83890, 83898, 83909x2, 83912 81479
1/1/2013 GSNMS 60717 GSN Gene, Full Gene Analysis 83890, 83898x17, 83909x34, 83912 81479
1/1/2013 HAPB 80297 Hemophilia A Mol Anal for Inversion 83891, 83892x2, 83893x2, 83894x2, 83896, 83897x2, 83912 81403
7/1/2013 HCCP 61703 Hereditary Colon Cancer Panel 81201, 81228, 81292, 81295, 81298, 81317, 81319, 81321, 81401, 81405, 81406, 81479 81405, 81406
1/1/2013 HD 61622 Huntington Disease Analysis 83891, 83898x2, 83909, 83912 81401
1/1/2013 HEMP 61337 Hereditary Erythrocytosis Mut, B 83890, 83900x3, 83901x3, 83904x20, 83909x20, 83912 81479
1/1/2013 HEXKM 89283 HEXA Gene, Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 HEXMS 89278 HEXA Gene, Full Gene Analysis 83891, 83898x14, 83909x28, 83912 81406
1/9/2013 HGBMO 29374 HGB Electrophoresis, Molecular 83891, 83898x4, 83900, 83904x12, 83909x13, 83914x8 81257 81403 81401
1/1/2013 HHEMO 81508 Hemochromatosis HFE Gene Analysis, Blood 83890, 83896x2, 83898x2, 83912 81256
7/1/2013 HHTM 89394 ENG and ACVRL1, Large Del/Dup 81479 81405, 81479
7/1/2013 HHTP 89394 ENG and ACVRL1, Full Gene Analysis 81479 81406, 81405, 81479
7/1/2013 HHTS 31056 ENG and ACVRL1, Full Gene Analysis 81479 81406, 81405, 81479
7/1/2013 HHTSQ 89442 HHT Gene Sequencing 81479
1/1/2013 HIF2A 61681 HIF2A Gene, Mutation Analysis, B 83900, 83904x4, 83909x4 81479
1/1/2013 HL15O 60348 HLAB 1502 Genotype, Saliva 83894x5, 83900, 83901x8, 83912 81381
1/1/2013 HL57O 60347 HLAB 5701 Abacavir Genotype, Saliva 83894, 83900, 83901, 83912 81381
1/1/2013 HLA15 89347 HLAB 1502 Genotype, Carbamazepine 83894x5, 83900, 83901x8, 83912 81381
1/1/2013 HLA57 89346 HLA-B 5701 Genotype, Abacavir 83894, 83900, 83901, 83912 81381
1/1/2013 HMBSK 61217 HMBS Gene, Known Mutation 83891, 83898, 83909x2, 83912 81479
1/1/2013 HMBSS 61216 HMBS Gene, Full Gene Sequence 83891, 83898x12, 83909x24, 83912 81479
1/1/2013 HNPCC 17073 HNPCC Screen 83890x2, 83900x2, 83901x6, 83909x2, 83912 81301
1/1/2013 HP 83019 Hereditary Pancreatitis, Mutation Screen 83890, 83898x2, 83909x4, 83912 81401
1/1/2013 HPKM 88691 Hered Pancreatitis, Known Mut 83890, 83898, 83909x2, 83912 81403
1/1/2013 HTR2 83303 Serotonin Receptor 2A/2C Genotype 83892, 83900, 83901x3, 83912, 83914x5 81479
1/1/2013 HTR2O 60338 Serotonin Receptor 2A/2C, Saliva 83892, 83900, 83901x3, 83912, 83914x5 81479
1/1/2013 HTT 83302 Serotonin Transporter Genotype 83894, 83898, 83912 81479
1/1/2013 HTTO 60339 Serotonin Transporter (LPR), Saliva 83894, 83898, 83912 81479
7/1/2013 HURLK 61482 Hurler Syndrome, Known Mutation 81479 81403
7/1/2013 HURLS 61481 Hurler Syndrome, Full Gene Analysis 81479 81406
1/1/2013 IVD 83644 IVD Mutation Analysis (A282V) 83890, 83892, 83894, 83898, 83912 81400
1/1/2013 JAK2B 88715 JAK2 V617F Mutation Detection, B 83891, 83896, 83898, 83912 81270
1/1/2013 JAK2M 31155 JAK2 V617F Mutation Detection, BM 83891, 83896, 83898, 83912 81270
1/1/2013 JAK2V 31156 JAK2 V617F Mutation Detection, V 83891, 83896, 83898, 83912 81270
1/1/2013 JAKXB 89189 JAK2 Exon 12 Mutation Detection, B 83891, 83898, 83902, 83904x2, 83909x2 81403
1/1/2013 JAKXM 60025 JAK2 Exon 12 Mutation Detection, BM 83891, 83898, 83902, 83904x2, 83909x2 81403
1/1/2013 KITAS 88802 KIT Asp816Val Mutation Analysis 83898x2, 83909x2 81402
1/1/2013 KRAS 89378 KRAS Gene, 7 Mutation Panel, Tumor 83890, 83896x7, 83898x7, 83912 81275
1/1/2013 KRASQ 89377 KRAS Gene, Exon 2 Gene Seq, Tumor 83890, 83892, 83894, 83898, 83909x2, 83912 81479
7/1/2013 LDLFQ 89124 LDLR Gene Sequencing 81470 81406
7/1/2013 LDLK_ 80209 LDLR Gene, Known Mutation 81479 81403
7/1/2013 LDLM 89073 LDLR Large De/Dup RUO 81479 81405
7/1/2013 LDLMP 29285 LDLR Large De/Dup RUO 81479 81405
7/1/2013 LDLRK 81183 LDLR Gene, Known Mutation 81479 81403
7/1/2013 LDLRQ 80098 LDLR, Full Gene Sequence 81479 81406
7/1/2013 LDLRS 81013 LDLR, Full Gene Sequence 81479 81406
7/1/2013 LDLSP 29284 LDLR, Full Gene Sequence 81479 81406
1/1/2013 LUMSC 84353 Luminex Class I/II Ab Scrn, B 86021 86828
1/1/2013 LYZKM 60720 LYZ Gene Known Mutation 83890, 83898, 83909x2, 83912 81479
1/1/2013 LYZMS 60719 LYZ Gene Full Gene Analysis 83890, 83898x4, 83909x8, 83912 81479
7/1/2013 MAHKM 89135 MMACHC Gene, Known Mutation 81479 81403
7/1/2013 MAHMS 89436 MMACHC Gene, Full Gene Analysis 81479 81404
7/1/2013 MAPTK 87925 MAPT Known Mutation 81479 81403
7/1/2013 MAPTM 87924 MAPT Screening Sequence Analysis 81479 81406
1/1/2013 MAS 89573 Alpha Globin Gene Sequence 83898x2, 83904x8, 83909x8 81257
1/1/2013 MBS 89405 Beta Globin Gene Sequence 83898x2, 83904x4, 83909x4 81404
1/1/2013 MCADK 83934 MCAD, Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 MCADS 60116 MCAD Mutation Screen 83890, 83898x12, 83909x22, 83912 81479
1/1/2013 MCC 88636 Maternal Cell Contamination, Molecular Analysis 83890, 83900x6, 83909x2, 83912 81265
1/1/2013 MCDKM 89831 MLYCD Gene, Known Mutation 83891, 83912 81479
1/1/2013 MCDMS 89830 MLYCD Gene, Full Gene Analysis 83891, 83898x7, 83909x14, 83912 81479
1/1/2013 MCIV 85321 Mucolipidosis IV, Mutation Analysis, IVS3(-2) A->G and del6.4kb 83890, 83892, 83896x2, 83900, 83909, 83912, 83914x2 81290
1/1/2013 ME2KM 89285 MECP2 Gene, Known Mutation 83891, 83912 81303
1/1/2013 ME2MS 89284 MECP2 Gene, Full Gene Analysis 83891, 83898x7, 83909x14, 83912 81302
1/1/2013 MENKM 81082 MEN2 (2A,2B,FMTC) Known Mutation 83891, 83898, 83909x2, 83912 81403
1/1/2013 MENMS 80573 MEN2 (2A,2B,FMTC) Mutation Screen 83891, 83898x6, 83909x12, 83912 81405
1/1/2013 MHDKM 61098 MMADHC Gene, Known Mutation 83891, 83898x2, 83909x4, 83912 81479
1/1/2013 MHDMS 61097 MMADHC Gene, Full Gene Analysis 83891, 83898x8, 83909x16, 83912 81479
1/1/2013 MHRS1 33092 High Resolution Class I Phenotype 83894, 83898, 83912 81379
1/1/2013 MHRS2 33093 High Resolution Class II Phenotype 83894, 83898, 83912 81382x2
1/1/2013 MLBRF 87931 MLH1 Hypermethylation/BRAF Mutation 83898x3, 83907, 83909x3, 83912 81210 81479
1/1/2013 MLH12 83191 MLH1/MSH2 Mutation Screen 83891, 83898x37, 83909x74, 83912 81292, 81295, 81294, 81297
1/1/2013 MLH1H 87978 MLH1 Hypermethylation (also part of a profile test 87931 with BRAF) 83898x2, 83907, 83909, 83912 81479
1/1/2013 MLHKM 83002 MLH1 Known Mutation 83891, 83912 81293
1/1/2013 MLHMS 83015 MLH1 Mutation Screen 83891, 83898x19, 83909x38, 83912 81292, 81294
1/1/2013 MMLDD 83192 hMLH1/hMSH2 Large Deletion/Duplication 83900, 83909, 83914x37 81294, 81297
1/1/2013 MPCR 80226 DNA Analysis B 83891, 83894x2, 83898x2, 83912 81479
1/1/2013 MPLB 89776 MPL Exon 10 Mutation Detection, B 83898, 83904x2, 83909x2 81403
1/1/2013 MPLM 60024 MPL Exon 10 Mutation Detection, BM 83898, 83904x2, 83909x2 81403
1/1/2013 MSH2K 83082 MSH2 Known Mutation 83891, 83912 81296
1/1/2013 MSH2M 83016 MSH2 Mutation Screen 83891, 83898x18, 83909x36, 83912 81295, 81297
1/1/2013 MSH6K 83706 MSH6 Known Mutation 83891, 83912 81299
1/1/2013 MSH6L 88893 hMSH6 Large Deletion/Duplication 83900, 83909, 83914x10 81300
1/1/2013 MSH6M 83723 MSH6 Mutation Screen, Blood 83891, 83898x19, 83909x38, 83912 81298, 81300
1/1/2013 MSHDB 89849 MSH6 Large Deletion/Duplication, MLPA 83900, 83909, 83914x10 81300
1/1/2013 MSIO 88566 Microsatellite Instability (MSI), Tumor 83890x2, 83900x2, 83901x6, 83909x2, 83912 81301
1/1/2013 MTHFR 81648 Methylenetetrahydrofol Reduc Mut, B 83891, 83892, 83896x5, 83903, 83908x2, 83912 81291
1/1/2013 MYH 84304 MYH Gene Analysis 83891, 83892x2, 83894x2, 83898x2, 83912 81401
1/1/2013 NAGR 82943 Hexosaminidase A and Tot, WBC/Mole 83080x2 83080x2
3/25/2013 NARC 82026 Narcolepsy Associated Ag, B 83890, 83894, 83898, 83912, 86816 81376
1/1/2013 NAT2 83389 NAT2, Full Gene Sequence 83892, 83894, 83898, 83909x8, 83912 81479
1/1/2013 NAT2F 29262 NAT2, Full Gene Sequence 83892, 83894, 83898, 83912 81479
1/1/2013 NAT2O 60345 NAT2 Full Gene Sequence, Saliva 83892, 83894, 83898, 83909x8, 83912 81479
7/1/2013 NPCKM 83118 Niemann-Pick Type C, Known Mutation 81479 81403
7/1/2013 NPCMS 89015 NPC Mutation Screen 81479 81404, 81406
1/1/2013 NPD 85322 Niemann-Pick Disease, Types A and B, Mutation Analysis 83890, 83892, 83896x4, 83900, 83909, 83912, 83914x4 81330
1/1/2013 NPDKM 61116 Niemann-Pick A-B, Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 NPDMS 61117 Niemann-Pick A-B Full Gene Analysis 83891, 83898x11, 83909x22, 83912 81479
1/1/2013 NPM1 89292 Nucleophosmin Mutation Analysis 83898, 83909 81401
1/1/2013 NT2FO 32808 NAT2, Full Gene Sequence 83892, 83894, 83898, 83912 81479
7/1/2013 OIL28 61701 IL28B Polymorphism Genotype, Saliva 81479 81400
1/1/2013 OPRM1 89612 OPRM1 Genotype, Naltrexone Efficacy 83896x2, 83898, 83912 81479
1/1/2013 OPRMO 60352 OPRM1 Genotype, Naltrexone, Saliva 83896x2, 83898, 83912 81479
1/1/2013 PHD2 61683 PHD2 Gene, Mutation Analysis, B 83900, 83901x3, 83904x12, 83909x12 81479
1/1/2013 PMLR 84114 PML/RARA Quantitative, PCR 83891, 83896, 83898x2, 83902 81315
1/1/2013 PMS2K 61174 PMS2 Gene, Known Mutation 83891, 83912 81318
1/1/2013 PMS2S 61173 PMS2 Gene, Full Gene Analysis 83891, 83894x3, 83898x7, 83900, 83909x33, 83912, 83914x34 81317
1/1/2013 PROD 60552 Influenza Virus A/B and RSV, PCR 87502, 87798 87631
1/1/2013 PT11 89463 PTPN11, Full Gene Sequence, B 83892x15, 83894, 83898x15, 83909x28, 83912 81406
1/1/2013 PT11S 33870 PTPN11, Full Gene Sequence 83892x15, 83894, 83898x15, 83912 81406
1/1/2013 PT1K 89464 PTPN11 Gene, Known Mutation, B 83892, 83894, 83898, 83909x2, 83912 81403
1/1/2013 PT1KS 33874 PTPN11 Gene, Known Mutation 83892, 83894, 83898, 83912 81403
1/1/2013 PTNT 81742 Prothrombin G20210A Mutation, B 83891, 83892, 83896x5, 83903, 83908x2, 83912 81240
1/1/2013 PTP22 89315 PTPN22 Genotype, 1858C>T 83896x2, 83898, 83912 81479
1/1/2013 PWDNA 81153 Prader-Willi/Angelman Syndrome, Molecular Analysis 83891, 83892x2, 83900, 83909x2, 83912, 83914x25 81331
1/1/2013 QUAD 81149 Quad Screen (Second Trimester) Maternal, Serum 82105, 82677, 84702, 86336 81511
1/1/2013 SABC1 89667 Class I Antibody Single Ag Bead 86021 86832
1/1/2013 SABC2 89668 Class II Antibody Single Ag Bead 86021 86833
1/1/2013 SBMA 81176 Spinobulbar Musc Atrophy, Kennedy>s 83890, 83898, 83909, 83912 81401
1/1/2013 SCADK 83947 SCAD Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 SCADM 83939 SCAD Mutation Screen 83891, 83898x9, 83909x18, 83912 81405
1/1/2013 SDHDD 89555 SDH Deletion Detection 83900, 83901x21, 83909, 83912, 83914 81403
1/1/2013 SDHKM 89554 SDH Known Mutation 83892, 83894, 83900, 83909x2, 83912 81403
1/1/2013 SDHSB 89551 SDH Subunit B Gene Analysis 83892x3, 83894x3, 83898, 83900x2, 83901x3, 83909x16, 83912 81405
1/1/2013 SDHSC 89552 SDH Subunit C Gene Analysis 83892x3, 83894x3, 83898, 83900x2, 83901, 83909x12, 83912 81405
1/1/2013 SDHSD 89553 SDH Subunit D Gene Analysis 83892x2, 83894x2, 83900x2, 83909x8, 83912 81404
1/1/2013 SDHSP 89550 SDH Gene Analysis 83892x8, 83894x8, 83898x2, 83900x6, 83901x4, 83909x36, 83912 81404, 81405
1/1/2013 SEPTK 61101 SEPT9 Gene, Known Mutation 83891, 83912 81479
1/1/2013 SEPTS 61100 SEPT9 Gene, Mutation Screen 83891, 83898x3, 83900, 83909x7, 83912, 83914x16 81479
1/1/2013 SEQF 60700 Sequential Maternal Screening, Part 2, Serum 82105, 82677, 84702, 86336 81599
1/1/2013 SHDD 31846 Succinate Dehydrogenase (SDH) Deletion Detection 83900, 83901x21, 83909, 83912, 83914 81403
7/1/2013 SLC1B 61736 SLCO1B1 Genotype, Statin B 81479 81400
7/1/2013 SLC1O 61737 SLCO1B1 Genotype, Statin, Saliva 81479 81400
1/1/2013 SPCID 82971 Specimen Source Identification 83890, 83900x6, 83909x2, 83912 81265
1/1/2013 TACIF 84388 TACI, Full Gene Sequence 83894x5, 83898x5, 83909x12, 83912 81479
1/1/2013 TACIG 89122 TACI Gene, Known Mutation 83892, 83894, 83898, 83909x2, 83912 81479
1/1/2013 TACIM 28588 TACI Gene, Known Mutation 83892, 83894, 83898, 83912 81479
1/1/2013 TACIS 28582 TACI, Full Gene Sequence 83894x5, 83898x5, 83912 81479
1/1/2013 TCGBM 31139 T Cell Receptor Gene Rearrange, BM 83912, 83891, 83900, 83901x4, 83909x6 81340, 81342
1/1/2013 TCGR 83122 T Cell Receptor Gene Rearrange, B Varies 81340, 81342
1/1/2013 TCGRV 31140 T Cell Receptor Gene Rearrange, V 83912, 83891, 83900, 83901x4, 83909x6 81340, 81342
1/1/2013 TG1F 32501 TGFBR1 Full Gene Sequence 83891, 83892x11, 83894, 83898x11, 83912 81405
1/1/2013 TG1K 32504 TGFBR1 Gene, Known Mutation 83891, 83892, 83894, 83898, 83912 81403
1/1/2013 TG2F 31751 TGFBR2 Full Gene Sequence 83892x12, 83894, 83898x12, 83912 81405
1/1/2013 TG2K 31754 TGFBR2 Gene, Known Mutation 83892, 83894, 83898, 83912 81403
1/1/2013 TGF1 89459 TGFBR1, Full Gene Sequence 83891, 83892x11, 83894, 83898x11, 83909x18, 83912 81405
1/1/2013 TGF2 89461 TGFBR2 Full Gene Sequence 83892x12, 83894, 83898x12, 83909x22, 83912 81405
1/1/2013 TGFK1 89460 TGFBR1 Gene, Known Mutation 83891, 83892, 83894, 83898, 83909x2, 83912 81403
1/1/2013 TGFK2 89462 TGFBR2 Gene, Known Mutation 83892, 83894, 83898, 83909x2, 83912 81403
2/12/2013 TREC 87959 Thymopoiesis Assessment Profile 83896x4, 83898x2, 83907, 83912 81479
1/1/2013 TSD 82588 Tay-Sachs Disease, Mutation Analysis, HEXA 83890, 83892, 83896x7, 83900, 83909, 83912, 83914x7 81255
1/1/2013 TUMSI 82500 Microsatellite Instability (MSI), Tumor 83890x2, 83900x2, 83901x6, 83909x2, 83912 81301
1/1/2013 U1A1 83949 UGT1A1 TA Repeat Genotype 83898, 83909, 83912 81350
1/1/2013 U1A1O 60343 UGT1A1 TA Repeat Genotype, Saliva 83898, 83909, 83912 81350
1/1/2013 UBEKM 89920 UBE3A Gene, Known Mutation 83891, 83898, 83909x2, 83912 81403
1/1/2013 UBEMS 89919 UBE3A Gene, Full Gene Analysis 83891, 83898x12, 83909x24, 83912 81406
1/1/2013 UGKMO 32811 UGT1A1 Gene, Known Mutation 83892, 83894, 83898, 83912 81403
1/1/2013 UGT1 29338 UGT1A1 Gene Sequence, Irinotecan 83892, 83894, 83900, 83901x3, 83912 81350
1/1/2013 UGT1O 32809 UGT1A1 Gene Sequence, Irinotecan 83892, 83894, 83900, 83901x3, 83912 81350
1/1/2013 UGT2 89611 UGT1A1 Sequence, Hyperbilirubinemia 83892, 83894, 83900, 83901x3, 83909x19, 83912 81350
1/1/2013 UGT2O 60350 UGT1A1 Sequence, Hyperbili, Saliva 83892, 83894, 83900, 83901x3, 83909x19, 83912 81350
1/1/2013 UGTH 30984 UGT1A1 Sequence, Hyperbilirubinemia 83892, 83894, 83900, 83901x3, 83912 81350
1/1/2013 UGTHO 32810 UGT1A1 Sequence, Hyperbilirubinemia 83892, 83894, 83900, 83901x3, 83912 81350
1/1/2013 UGTI 89397 UGT1A1 Gene Sequence, Irinotecan 83892, 83894, 83900, 83901x3, 83909x19, 83912 81350
1/1/2013 UGTIO 60349 UGT1A1 Sequence, Irinotecan, Saliva 83892, 83894, 83900, 83901x3, 83909x19, 83912 81350
1/1/2013 UGTK 89396 UGT1A1 Gene, Known Mutation 83892, 83894, 83898, 83909x2, 83912 81403
1/1/2013 UGTKM 30987 UGT1A1 Gene, Known Mutation 83892, 83894, 83898, 83912 81403
1/1/2013 UGTKO 60351 UGT1A1 Gene, Known Mutation, Saliva 83892, 83894, 83898, 83909x2, 83912 81403
1/1/2013 UPD 82970 Uniparental Disomy 83891, 83900x3, 83909, 83912 81402
1/1/2013 VHDD 31762 VHL Deletion Detection 83900, 83901x14, 83909, 83914 81403
1/1/2013 VHLD 89211 VHL Deletion Detection 83900, 83901x14, 83909, 83912, 83914 81403
1/1/2013 VHLKP 89084 VHL Known Mutation 83892, 83894, 83900, 83909x2, 83912 81403
1/1/2013 VHLSP 89083 VHL Full Gene Analysis 83892x2, 83894, 83900, 83901x4, 83909x6, 83912 81404
1/1/2013 VLCKM 60037 VLCAD Deficiency, Known Mutation 83891, 83898, 83909x2, 83912 81403
1/1/2013 VLCMS 60036 VLCAD Deficiency, Full Gene Analysis 83891, 83898x13, 83909x26, 83912 81406
1/1/2013 VWD2N 81662 von Willebrand Disease 2N (Normandy) 83890, 83892x3, 83894x3, 83898x3, 83912 81401
1/1/2013 WARFO 60341 Warfarin Sensitive Genotype, Saliva 83892, 83894, 83900, 83901x2, 83909x8, 83912 81227, 81355
1/1/2013 WARFP 60529 Warfarin Sensitivity Genotype 83892, 83894, 83900, 83901x2, 83909x8, 83912 81227, 81355
1/1/2013 WARFQ 87844 Warfarin Sensitivity Genotyping 83909x8 81223
1/1/2013 WARFS 89033 Warfarin Sensitivity, Genotype 83892, 83894, 83900, 83901x2, 83912 81227, 81355
1/1/2013 WARSO 32807 Warfarin Sensitivity Genotype, Saliva 83892, 83894, 83900, 83901x2, 83912 81227, 81355
1/1/2013 WDKM 83698 Wilson Disease Known Mutation 83891, 83898x2, 83909x4, 83912 81403
1/1/2013 WDMS 83697 Wilson Disease Mutation Screen 83891, 83898x22, 83909x44, 83912 81406
1/1/2013 YMICR 82992 Y Microdeletion 83890, 83894x2, 83900, 83912 81403
1/1/2013 ZYG 81252 Zygosity Testing (Multiple Births) 83891, 83900x6, 83909x2, 83912 81265

CPT = Current Procedural Terminology.

References

  • 1.Ettinger DS. Ten years of progress in non-small cell lung cancer. J Natl Compr Canc Netw. 2012;10(3):292-95. [DOI] [PubMed] [Google Scholar]
  • 2.Berge EM, Doebele RC. Targeted therapies in non-small cell lung cancer: emerging oncogene targets following the success of epidermal growth factor receptor. Semin Oncol. 2014;41(1):110-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Miller VA, Riely GJ, Zakowski MF, et al. Molecular characteristics of bronchioloalveolar carcinoma and adenocarcinoma, bronchioloalveolar carcinoma subtype, predict response to erlotinib. J Clin Oncol. 2008;26(9):1472-78. [DOI] [PubMed] [Google Scholar]
  • 4.Zhang X, Zhang S, Yang X, et al. Fusion of EML4 and ALK is associated with development of lung adenocarcinomas lacking EGFR and KRAS mutations and is correlated with ALK expression. Mol Cancer. 2010;9:188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Camidge DR, Kono SA, Flacco A, et al. Optimizing the detection of lung cancer patients harboring anaplastic lymphoma kinase (ALK) gene rearrangements potentially suitable for ALK inhibitor treatment. Clin Cancer Res. 2010;16(22):5581-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hirsch FR, Bunn PA, Jr.. EGFR testing in lung cancer is ready for prime time. Lancet Oncol. 2009;10(5):432-33. [DOI] [PubMed] [Google Scholar]
  • 7.Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2011;363(18):1693-703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shaw AT, Yeap BY, Mino-Kenudson M, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 2009;27(26):4247-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Solomon B, Varella-Garcia M, Camidge DR.. ALK gene rearrangements: a new therapeutic target in a molecularly defined subset of non-small cell lung cancer. J Thorac Oncol. 2009;4(12):1450-54. [DOI] [PubMed] [Google Scholar]
  • 10.Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361(10):947-57. [DOI] [PubMed] [Google Scholar]
  • 11.Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31(27):3327-34. [DOI] [PubMed] [Google Scholar]
  • 12.Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, openlabel, randomised, phase 3 study. Lancet Oncol. 2011;12(8):735-42. [DOI] [PubMed] [Google Scholar]
  • 13.Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol. 2009;29(21):2866-74. [DOI] [PubMed] [Google Scholar]
  • 14.Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362(25):2380-88. [DOI] [PubMed] [Google Scholar]
  • 15.Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11(2):121-28. [DOI] [PubMed] [Google Scholar]
  • 16.Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13(3):239-46. [DOI] [PubMed] [Google Scholar]
  • 17.Ettinger DS, Akerley W, Bepler G, et al. Non-small cell lung cancer. J Natl Compr Canc Netw. 2010;8(7):740-801. [DOI] [PubMed] [Google Scholar]
  • 18.Keedy VL, Temin S, Somerfield MR, et al. American Society of Clinical Oncology provisional clinical opinion: epidermal growth factor receptor (EGFR) mutation testing for patients with advanced non-small-cell lung cancer considering first-line EGFR tyrosine kinase inhibitor therapy. J Clin Oncol. 2011;29(15):2121-27. [DOI] [PubMed] [Google Scholar]
  • 19.Riely GJ, Chaft JE, Ladanyi M, et al. Incorporation of crizotinib into the NCCN guidelines. J Natl Compr Canc Netw. 2011;9(12):1328-30. [DOI] [PubMed] [Google Scholar]
  • 20.Lindeman NI, Cagle PT, Beasley MB, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol. 2013;8(7):823-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cao Y, Xiao G, Qiu X, et al. Efficacy and safety of crizotinib among Chinese EML4-ALK-positive, advanced-stage non-small cell lung cancer patients. PLoS One. 2014;9(12):e114008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chen G, Feng J, Zhou C, et al. Quality of life (QoL) analyses from OPTIMAL (CTONG-0802), a phase III, randomised, open-label study of first-line erlotinib versus chemotherapy in patients with advanced EGFR mutation-positive non-small-cell lung cancer (NSCLC). Ann Oncol. 2013;24(6):1615-22. [DOI] [PubMed] [Google Scholar]
  • 23.Yang SC, Lai WW, Hsiue TR, et al. Health-related quality of life after first-line anti-cancer treatments for advanced non-small cell lung cancer in clinical practice. Qual Life Res. 2016;25(6):1441-49. [DOI] [PubMed] [Google Scholar]
  • 24.Romanus D, Cardarella S, Cutler D, et al. Cost-effectiveness of multiplexed predictive biomarker screening in non-small-cell lung cancer. J Thorac Oncol. 2015;10(4):586-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Blackhall F, Thatcher N, Booton R, et al. The impact on the multidisciplinary team of molecular profiling for personalized therapy in non-small cell lung cancer. Lung Cancer. 2013;79(2):101-03. [DOI] [PubMed] [Google Scholar]
  • 26.Ellis PM. The importance of multidisciplinary team management of patients with non-small-cell lung cancer. Curr Oncol. 2012;19(Suppl 1):S7-S15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Graham DM, Leighl NB. Economic impact of tissue testing and treatments of metastatic NSCLC in the era of personalized medicine. Front Oncol. 2014;4:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.De Souza JA, Gleason PP, Starner CI, Qiu YP. Erlotinib and patterns of pharmacogenomic (PGx) testing. J Clin Oncol. 2012;30 (Suppl 34):abstract 182. [Google Scholar]
  • 29.Reck M, Hermes A, Tan EH, et al. Tissue sampling in lung cancer: a review in light of the MERIT experience. Lung Cancer. 2011;74(1):1-6. [DOI] [PubMed] [Google Scholar]
  • 30.Stevenson M, Christensen J, Shoemaker D, et al. Tumor acquisition for biomarker research in lung cancer. Cancer Invest. 2014;32(6):291-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ellis PM, Verma S, Sehdev S, et al. Challenges to implementation of an epidermal growth factor receptor testing strategy for non-smallcell lung cancer in a publicly funded health care system. J Thorac Oncol. 2013;8(9):1136-41. [DOI] [PubMed] [Google Scholar]
  • 32.Spicer J, Tischer B, Peters M. EGFR mutation testing and oncologist treatment choice in advanced NSCLC: global trends and differences. Ann Oncology. 2015;26(Suppl 1):i60. [Abstract]. [Google Scholar]
  • 33.Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350(21):2129-39. [DOI] [PubMed] [Google Scholar]
  • 34.Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304(5676):1497-500. [DOI] [PubMed] [Google Scholar]
  • 35.Lynch JA, Khoury MJ, Borzecki A, et al. Utilization of epidermal growth factor receptor (EGFR) testing in the United States: a case study of T3 translational research. Genet Med. 2013;15(8):630-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Levy BP, Chioda MD, Herndon D, et al. Molecular testing for treatment of metastatic non-small cell lung cancer: how to implement evidence-based recommendations. Oncologist. 2015;20(10):1175-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lokhandwala T, Dann R, Johnson M, et al. Costs of the diagnostic workup for lung cancer: a Medicare claims analysis: diagnosis/staging. Int J Radiat Oncol Biol Phys. 2014;90(5 Suppl):S9-S10. [Google Scholar]

Articles from Journal of Managed Care & Specialty Pharmacy are provided here courtesy of Academy of Managed Care Pharmacy

RESOURCES