Coverage Policy Manual
Policy #: 2014013
Category: Laboratory
Initiated: June 2014
Last Review: November 2023
  Genetic Test: Li-Fraumeni Syndrome

Description:
Li-Fraumeni syndrome (LFS) is a cancer predisposition syndrome associated with the development of several different types of tumors. The syndrome is caused by germline mutations in the TP53 gene.
 
Background
LFS is a cancer predisposition syndrome associated a high lifetime cumulative risk of cancer and a tendency for multiple cancers in affected individuals. The syndrome was originally described in 1969 by 2 physician-scientists, Frederick P. Li and Joseph F. Fraumeni, based on a retrospective analysis of families with aggressive soft tissue sarcomas in young siblings and their biologically related cousins (Sorrell, 2013).
 
The tumor types that are most closely associated with LFS include soft tissue sarcomas, osteosarcomas, premenopausal breast cancer, brain tumors, and adrenal cortical carcinoma (Schneider, 1993). Other malignancies associated with LFS include a wide variety of gastrointestinal tract, lung, skin, and thyroid cancers as well as leukemias and lymphomas.
 
Individuals with LFS are at increased risk of developing multiple primary tumors, with subsequent malignancies not all being clearly related to the treatment of the previous neoplasms. The risk of developing a second tumor has been estimated at 40 to 49% (Schneider, 1993). In 1 study of 322 pathogenic variant carriers from France, Bougeard et al reported that 43% of individuals had multiple malignancies (Bougeard, 2015).
 
Individuals with LFS are at increased risk of both bone and soft tissue sarcomas. Sarcomas of various histologies account for 25% of the cancers reported in people with LFS, with the most commonly reported sarcomas in an international database being rhabdomyosarcoma before age 5 years and osteosarcoma at any age (Ognjanovic, 2012). Women with LFS are at greatly increased risk of developing premenopausal breast cancer, with the median age of diagnosis being 33 years of age (Schneider, 1993). Male breast cancer has rarely been reported in LFS families (Schneider, 1993). Many different types of brain tumors have been described in LFS, including astrocytomas, glioblastomas, medulloblastomas and choroid plexus carcinomas (Schneider, 1993). The median age of onset of LFS-related brain tumors is 16 years of age. Individuals with LFS are at increased risk of developing adrenal cortical carcinoma (ACC). In adults, in 1 series, it was estimated that 6% of individuals diagnosed with ACC after age 18 years have a germline TP53 mutation (Raymond, 2013).
 
Data from M.D. Anderson Cancer Center’s long-term clinical studies of LFS showed that the risk of developing soft tissue sarcomas is greatest before the age of 10, brain cancer appears to occur early in childhood with a smaller peak in risk in the fourth to fifth decade of life, risk for osteosarcoma is highest during adolescence, and breast cancer risk among females with LFS starts to increase significantly around age 20 and continues into older adulthood (Hwang, 2003).
 
Clinical Diagnosis
The diagnosis of LFS is based on an evolving set of clinical classification criteria, established using salient aspects of family history and tumor-related characteristics (Sorrell, 2013). The first formal set of criteria, the classic LFS criteria, were developed in 1988, and are the most stringent criteria used to make a clinical diagnosis of LFS (Sorrell, 2013).  
 
Classic LFS is defined by the presence of all of the following criteria:
        • A proband with a sarcoma before 45 years of age
        • A first-degree relative with any cancer before 45 years of age
        • A first- or second-degree relative with any cancer before 45 years of age or a sarcoma at any age (Schneider, 1993).  
 
Chompret et al developed criteria which were shown to have the highest positive predictive value, and which, when combined with the classic LFS criteria, provide the highest sensitivity for identifying individuals with LFS (Chompret, 2001). The Chompret criteria were updated in 2009 to assist in identifying families with milder phenotypes (Gonzalez, 2009). The Chompret criteria will also identify individuals with de novo TP53 mutations, whereas the classic LFS criteria require a family history.
 
The Chompret Criteria, most recently updated in 2015, are defined as the following:
        • Proband with tumor belonging to LFS tumor spectrum (eg, soft tissue sarcoma, osteosarcoma, brain tumor, premenopausal breast cancer, adrenocortical carcinoma, leukemia, lung bronchoalveolar cancer) before age 46 years AND at least 1 first- or second-degree relative with LFS tumor (except breast cancer if proband has breast cancer) before age 56 years or with multiple tumors; OR
        • Proband with multiple tumors (except multiple breast tumors), 2 of which belong to LFS tumor spectrum and the first of which occurred before age 46 years; OR
        • Patient with adrenocortical carcinoma (ACC), rhabdomyosarcoma of embryonal anaplastic subtype, or choroid plexus tumor, irrespective of family history OR
        • Female proband with breast cancer before age 31 years.4,3,
 
National Comprehensive Cancer Network (NCCN) guidelines recommend TP53 analysis for individuals who meet classic LFS criteria, Chompret criteria, or who have been diagnosed with early-onset breast cancer (age of diagnosis 35 years).
 
Molecular Diagnosis
LFS is associated with germline mutations in the TP53 gene (chromosome 17p13.1), which encodes for a ubiquitous transcription factor that is responsible for a complex set of regulatory functions that promote DNA repair and tumor suppression. TP53 is the only gene in which mutations are known to cause LFS, and no other inherited phenotypes are associated specifically with germline mutations involving TP53 (Schneider, 1993). The presence of a TP53 variant is considered diagnostic.
 
LFS is a highly penetrant cancer syndrome, with the of cancer being about 80% by age 70 (Schneider, 1993). LFS is inherited in an autosomal dominant manner. De novo germline TP53 mutations (no mutation is identified in either biologic parent) are estimated to be 7% to 20%.
 
Approximately 95% of mutations detected in TP53 gene are sequence variants (small intragenic deletions/insertions and missense, nonsense, and splice site mutations). Large deletion/duplications not readily detected by sequence analysis accounts for approximately 1% of the mutations detected (Schneider, 1993).
 
Certain genotype-phenotype correlations have been reported in families with LFS and TP53 mutations. Genotype-phenotype correlations in LFS are predictive of the age of onset of tumor, level of risk of developing tumor, and outcome in patients with TP53 germline mutations (Sorrell, 2013; Schneider, 1993).
 
Management
 
Treatment
The evaluation for cancer in an individual diagnosed with LFS should be based on personal medical history and, to some degree, the specific pattern of cancer in the family. Women with LFS who develop breast cancer are encouraged to consider bilateral mastectomies to reduce the risk of developing a second primary breast cancer and to avoid exposure to radiation therapy. Preventive measures may include prophylactic mastectomy in women, and in all patients with a TP53 mutation, avoidance of radiation therapy, as there is some evidence to suggest that TP53 mutations confer an increased sensitivity to ionizing radiation and the possibility of radiation-induced malignancies.
 
Surveillance
LFS confers a high risk of multiple different types of cancer, which poses challenges for establishing a comprehensive screening regimen, and many of the cancers associated with LFS do not lend themselves to early detection.
 
There is no international consensus on the appropriate clinical surveillance strategy in individuals with LFS, (Mai, 2012) but, in general, the strategy includes physical examination, colonoscopy and breast imaging. Other protocols that are being evaluated include additional imaging techniques and biochemical assessment.
 
NCCN has consensus-based screening guidelines.
 
Regulatory Status
Clinical laboratories may develop and validate tests in-house and market them as a laboratory service; laboratory-developed tests must meet the general regulatory standards of the Clinical Laboratory Improvement Amendments. Laboratories that offer laboratory-developed tests must be licensed under the Clinical Laboratory Improvement Amendments for high-complexity testing. To date, the U.S. Food and Drug Administration has chosen not to require any regulatory review of this test.
 
Coding
CPT code 81405 includes the following testing for Li-Fraumeni syndrome:
 
TP53 (tumor protein 53) (eg, Li-Fraumeni syndrome, tumor samples), full gene sequence or targeted sequence analysis of >5 exons
 
Duplication/deletion analysis for TP53 would be reported with the unlisted molecular pathology procedure code (81479).
 
Effective 1/1/2021, new CPT codes 81351 [TP53 (tumor protein 53) (eg, Li Fraumeni syndrome) gene analysis; full gene sequence], 81352 [TP53 (tumor protein 53) (eg, Li Fraumeni syndrome) gene analysis; targeted sequence analysis (eg, 4 oncology)], and 81353 [TP53 (tumor protein 53) (eg, Li Fraumeni syndrome) gene analysis; known familial variant] were added.

Policy/
Coverage:
Effective August 2023
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing for TP53 mutations meets member benefit certificate primary coverage criteria to confirm a diagnosis of Li-Fraumeni syndrome under the following conditions:
 
    1. The individual has undergone genetic counseling (96040 or S0265); AND
    2. Meets either the classic or the Chompret clinical diagnostic criteria for Li-Fraumeni syndrome (See diagnostic criteria below); or
    3. Has early-onset breast cancer (age of diagnosis 35 years); or
    4. Is a first degree relative of a proband with a known TP53 pathogenic variant; or
    5. Pediatric hypodiploid acute lymphoblastic leukemia for individuals age 18 years, as well as adolescent and young adult (AYA) patients >18 years treated in a pediatric oncology setting; (the latter could include patients up to age 30 years.)
 
Diagnostic criteria for LFS:
 
Classic LFS
 
Classic LFS is defined by the presence of all of the following:
 
    1. A proband with a sarcoma before 45 years of age; and
    2. A first-degree relative with any cancer before 45 years of age; and
    3. A first- or second-degree relative with any cancer before 45 years of age or a sarcoma at any age.
 
Chompret criteria
 
Chompret Clinical Diagnostic Criteria is defined as:
 
    1. Proband with tumor belonging to LFS tumor spectrum (eg, soft tissue sarcoma, osteosarcoma, brain tumor, premenopausal breast cancer, adrenocortical carcinoma, leukemia, lung bronchoalveolar cancer) before age 46 years; AND
    2. At least 1 first- or second-degree relative with LFS tumor (except breast cancer if proband has breast cancer) before age 56 years or with multiple tumors; or
    3. Proband with multiple tumors (except multiple breast tumors), 2 of which belong to LFS tumor spectrum and first of which occurred before age 46 years; or
    4. Patient with adrenocortical carcinoma (ACC), rhabdomyosarcoma of embryonal anaplastic subtype or choroid plexus tumor irrespective of family history; or
    5. Female proband with breast cancer before age 31 years.
 
Note: The optimal strategy for confirming a TP53 mutation in a proband would be:
    1. Sequencing of the entire TP53 coding region (exons 2-11), which detects about 95% of TP53 mutations in patients with LFS (81405).
    2. If sequencing is negative, then deletion/duplication analysis, which detects large deletions/duplications (81479). These types of mutations account for less than 1 percent of mutations in individuals meeting classic LFS criteria. Deletion/duplication analysis (81479) will be covered only if sequencing of the entire TP53 coding region (81405) is negative.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for a germline or targeted TP53 mutation for any indication or circumstance not described above as covered does not meet member benefit certificate primary coverage criteria.
 
For members with contracts without primary coverage criteria, genetic testing for a germline or targeted TP53 mutation is considered not medically necessary for any indication or circumstance not described above as covered.
 
Effective November 2020 through July 2023
 
In general, genetic cancer susceptibility panels are not covered, however, when coverage criteria of this or another specific policy are met, limited genetic cancer susceptibility panels are covered, including only the gene variants for which a given member qualifies, as outlined in the policy.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing for TP53 mutations meets member benefit certificate primary coverage criteria to confirm a diagnosis of Li-Fraumeni syndrome under the following conditions:
 
    • The patient has undergone genetic counseling (96040 or S0265),
 
AND
 
    • Meets either the classic or the Chompret clinical diagnostic criteria for Li-Fraumeni syndrome (See diagnostic criteria below), or
      • Has early-onset breast cancer (age of diagnosis 35 years), or
      • Is a first degree relative of the proband positive for TP53 mutation.
 
Diagnostic criteria for LFS:
 
Classic LFS
 A proband with a sarcoma before 45 years of age AND   
      • A first-degree relative with any cancer before 45 years of age AND   
      • A first- or second-degree relative with any cancer before 45 years of age or a sarcoma at any age   
 
Chompret criteria
 
      • Proband with tumor belonging to LFS tumor spectrum (eg, soft tissue sarcoma, osteosarcoma, brain tumor, premenopausal breast cancer, adrenocortical carcinoma, leukemia, lung bronchoalveolar cancer) before age 46 years AND at least 1 first- or second-degree relative with LFS tumor (except breast cancer if proband has breast cancer) before age 56 years or with multiple tumors; OR   
      • Proband with multiple tumors (except multiple breast tumors), 2 of which belong to LFS tumor spectrum and first of which occurred before age 46 years; OR   
      • Patient with adrenocortical carcinoma (ACC) or choroid plexus tumor, irrespective of family history   
 
Note: The optimal strategy for confirming a TP53 mutation in a proband would be:
1) sequencing of the entire TP53 coding region (exons 2-11), which detects about 95% of TP53 mutations in patients with LFS (81405). If sequencing is negative, then:
2) deletion/duplication analysis, which detects large deletions/duplications (81479). These types of mutations account for less than 1 percent of mutations in individuals meeting classic LFS criteria.
Deletion/duplication analysis (81479) will be covered only if sequencing of the entire TP53 coding region (81405) is negative.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for a germline TP53 mutation for all other indications does not meet member benefit certificate primary coverage criteria.
 
For members with contracts without primary coverage criteria, genetic testing for a germline TP53 mutation is considered not medically necessary for all other indications.
 
Effective Prior to November 2020
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing for TP53 mutations meets member benefit certificate primary coverage criteria to confirm a diagnosis of Li-Fraumeni syndrome under the following conditions:
 
    • The patient has undergone genetic counseling (96040 or S0265),
 
AND
 
    • Meets either the classic or the Chompret clinical diagnostic criteria for Li-Fraumeni syndrome (See diagnostic criteria below), or
    • Has early-onset breast cancer (age of diagnosis 35 years), or
    • Is a first degree relative of the proband positive for TP53 mutation.
 
Diagnostic criteria for LFS:
 
Classic LFS
 
        • A proband with a sarcoma before 45 years of age AND  
        • A first-degree relative with any cancer before 45 years of age AND  
        • A first- or second-degree relative with any cancer before 45 years of age or a sarcoma at any age  
 
Chompret criteria
 
        • Proband with tumor belonging to LFS tumor spectrum (eg, soft tissue sarcoma, osteosarcoma, brain tumor, premenopausal breast cancer, adrenocortical carcinoma, leukemia, lung bronchoalveolar cancer) before age 46 years AND at least 1 first- or second-degree relative with LFS tumor (except breast cancer if proband has breast cancer) before age 56 years or with multiple tumors; OR  
        • Proband with multiple tumors (except multiple breast tumors), 2 of which belong to LFS tumor spectrum and first of which occurred before age 46 years; OR  
        • Patient with adrenocortical carcinoma (ACC) or choroid plexus tumor, irrespective of family history  
 
Note: The optimal strategy for confirming a TP53 mutation in a proband would be:
1) sequencing of the entire TP53 coding region (exons 2-11), which detects about 95% of TP53 mutations in patients with LFS (81405). If sequencing is negative, then:
2) deletion/duplication analysis, which detects large deletions/duplications (81479). These types of mutations account for less than 1 percent of mutations in individuals meeting classic LFS criteria.
Deletion/duplication analysis (81479) will be covered only if sequencing of the entire TP53 coding region (81405) is negative.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for a germline TP53 mutation for all other indications does not meet member benefit certificate primary coverage criteria.
 
For members with contracts without primary coverage criteria, genetic testing for a germline TP53 mutation is considered not medically necessary for all other indications.
 

Rationale:
Analytic validity (technical accuracy of the test in detecting a mutation that is present or in excluding a mutation that is absent)
 
According to a large reference laboratory, analytic sensitivity and specificity for polymerase chain reaction sequencing for LFS TP53 testing and deletions/duplications testing by multiplex ligation-dependent probe amplification is greater than 95% (ARUP, 2014).  
 
The order of testing to optimize yield would be:
1) sequencing of the entire TP53 coding region (exons 2-11), which detects about 95% of TP53 mutations in patients with LFS. Examples of types of mutations detected by sequence analysis include small deletions/duplications, and missense, nonsense and splice site mutations; most are missense mutations
2) deletion/duplication analysis, which detects large deletions/duplications involving the coding region, exon 1, or promoter; these types of deletions/duplications are not readily detectable by sequence analysis of the coding and flanking intronic regions of genomic DNA. These types of mutations account for less than 1 percent of mutations found in individuals with LFS.
 
Clinical validity (diagnostic performance of the test [sensitivity, specificity, positive and negative predictive values] in detecting clinical disease)
Approximately 80% of families with features of LFS will have an identifiable TP53 mutation (Schneider, 1993). Families that have no identifiable TP53 mutation but share clinical features of LFS are more likely to have a different hereditary cancer syndrome (eg hereditary breast-ovarian cancer syndrome) (Schneider, 1993).  
 
Clinical utility (how the results of the diagnostic test will be used to change management of the patient and whether these changes in management lead to clinically important improvements in health outcomes)
The clinical utility of genetic testing can be considered in the following clinical situations: 1) individuals with suspected LFS, and 2) family members of individuals with LFS. These situations will be discussed separately next.
 
Individuals with suspected LFS. The clinical utility for these patients depends on the ability of genetic testing to make a definitive diagnosis and for that diagnosis to lead to management changes that improve outcomes. Direct evidence for the clinical utility of genetic testing in these patients, describing how a molecular diagnosis of LFS changed patient management, is limited. However, for patients who are diagnosed with LFS by identifying a TP53 mutation, the medical management focuses on increased cancer surveillance to detect tumors at the earliest, most treatable stages.
 
Villani et al conducted a prospective, observational study of members of 8 LFS families who were asymptomatic TP53 carriers (Villani, 2011). The participants either chose to undergo or to not undergo surveillance. Surveillance included biochemical and imaging studies, which included ultrasounds, brain magnetic resonance imaging (MRI) scans and rapid total body MRI scans. The primary outcome measure was detection of new cancers and the secondary outcome measure was overall survival. Of 33 mutation carriers that were identified, 18 underwent surveillance. The surveillance protocol detected 10 asymptomatic tumors in 7 patients, which included premalignant or low grade tumors (3 low-grade gliomas, a benign thyroid tumor, 1 myelodysplastic syndrome), and small, high-grade tumors (2 choroid plexus carcinomas, 2 adrenocortical carcinomas, 1 sarcoma). The 9 solid tumors that were detected were completely resected, and the patients were in complete remission. After a median follow-up of 24 months, all of the patients who had undergone surveillance were alive. In the nonsurveillance group, 12 high-grade, high-stage tumors developed in 10 patients, of which 2 were alive at the end of follow-up (p=0·04 for comparison of survival in the surveillance group). Three-year overall survival in the surveillance group was 100% and 21% in the nonsurveillance group (p=0·0155).
 
Family members. When a TP53 mutation has been identified in a proband, testing of at-risk relatives can identify those who also have the mutation and have LFS. These individuals need initial evaluation and ongoing surveillance.
 
Conclusions. Direct evidence of the clinical utility of TP53 testing is limited. However, the clinical utility of genetic testing for TP53 mutations is that genetic testing can confirm the diagnosis in patients with clinical signs and symptoms of LFS and in at-risk family members. Management changes include increased surveillance and possible prophylactic measures for the cancers that are associated with this syndrome.
 
Summary
Li-Fraumeni syndrome (LFS) is a cancer predisposition syndrome associated with the development of several different types of tumors. The syndrome is caused by germline mutations in the TP53 gene.
 
Analytic validity of TP53 mutation testing is high, and almost all of these mutations can be identified by sequence analysis; a far smaller number of mutations can be detected by deletion/duplication analysis.
 
The clinical validity of TP53 mutation testing is high in that a mutation can be identified in up to 80% of patients who meet the clinical criteria for a diagnosis of LFS.
 
The clinical utility of genetic testing for a TP53 mutation is high in that confirming a diagnosis in a patient with clinical criteria of LFS will lead to changes in clinical management by increasing surveillance to detect cancers known to be associated with LFS at an early and treatable stage, or to address possible prophylactic measures. Most cases of LFS are inherited, and testing of at-risk relatives will identify those who should also undergo increased cancer surveillance.
 
Practice Guidelines and Position Statements
National Comprehensive Cancer Network guidelines (v.1.2014) recommend the following for LFS management:
 
Breast cancer risk, women:
 
    • Breast awareness starting at age 18 years.
    • Clinical breast exam every 6-12 months, starting at age 20-25 years or 5-10 years before the earliest known breast cancer in the family.
    • Breast screening:
        • Age 20-29 years, annual breast MRI screening (preferred) or mammogram if MRI is unavailable or individualized based on earliest age of onset in family.
        • Age >30-75 years, annual mammogram, and breast MRI screening.
        • Age >75 years, management considered on an individual basis.
        • Discuss risk-reducing mastectomy and counsel regarding degree of protection and cancer risk, and reconstruction options.
        • Address psychosocial, social, and quality-of-life aspects of risk-reducing mastectomy.
 
Other cancer risks:
    • Annual comprehensive physical exam with high index of suspicion for the cancers associated with LFS.
    • Consider colonoscopy every 2-5 years starting no later than 25 years of age.
    • Therapeutic radiation therapy for cancer treatment should be avoided when possible.
    • Discuss limitations of screening for many cancers associated with LFS, educate regarding signs and symptoms of cancer, and base additional surveillance on individual family history.
 
For relatives:
    • Advise about possible inherited cancer risk to relatives, options for risk assessment, and management.
    • Recommend genetic counseling and consideration of genetic testing for at-risk relatives.
 
 
U.S. Preventive Services Task Force (USPSTF) Guidelines
No USPSTF guidelines for Li-Fraumeni syndrome testing have been identified.
 
2015 Update
A literature search conducted through May 2015 did not reveal any new information that would prompt a change in the coverage statement.
 
2017 Update
A literature search conducted through May 2017 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
There is a lack of published evidence on analytic validity of testing for TP53 mutations. It is expected that analytic validity will be high when testing is performed according to optimal laboratory standards. The website of 1 large laboratory claims analytic validity of greater than 95% but empirical, peer-reviewed data is not available.
 
Cohorts of individuals with adrenocortical carcinoma, which is diagnostic of LFS by the Chompret criteria, have been published (Petitjean, 2007; Wagner, 1994; Wasserman, 2015). In 1 study, 88 consecutive patients with adrenocortical carcinoma were evaluated.14 Direct sequencing of exons 2 through 11 together with multiplex ligation-dependent probe amplification was used to identify mutations. For the entire population, 50% of individuals had a pathogenic mutation detected. The detection rate was dependent on age, with 58% of individuals younger than 12 years of age having a mutation compared with 25% of individuals between ages 12 and 20.
 
There is a small amount of evidence on the clinical validity of testing for TP53 mutations. In patients who meet clinical criteria for LFS, the clinical sensitivity has been reported to range between 50% and 80%. The largest amount of evidence is on patients with adrenocortical carcinoma, which represents a subset of all patients with LFS. No evidence was identified on the clinical specificity of testing.
 
Diagnostic Testing in Individuals With Suspected LFS
Direct evidence for the clinical utility of genetic testing to confirm a diagnosis of LFS is lacking. An indirect chain of evidence can provide evidence of clinical utility if all the links in the chain of evidence are intact.
 
The elements contributing to the indirect chain of evidence are derived from evidence review 2.04.91 (general approach to genetic testing). The following series of questions represent the indirect chain of evidence for diagnostic testing to confirm a diagnosis of LFS.
 
Are there some individuals in which the diagnosis of LFS is uncertain following standard clinical workup without genetic testing?
 
Yes. There are standardized diagnostic criteria based on personal, clinical and family history. However, there are limitations to these methods of diagnosis. A detailed family history may not be complete or may not be available in many instances. There are different diagnostic instruments that use different criteria, and they may be used alone or in combination with each other. The population identified as having LFS will differ depending on the way the instruments are used. In addition, the available instruments do not have high overall accuracy. Therefore, there may be considerable uncertainty about the diagnosis using clinical criteria alone.
 
Can genetic testing make the diagnosis of LFS with certainty in patients with an uncertain clinical diagnosis?
 
Yes, in some patients. A positive genetic test will confirm the diagnosis of LFS with high certainty in individuals who meet clinical criteria. As a result, patients with a positive genetic test will have a high certainty for the diagnosis of LFS, whereas the diagnosis by clinical criteria alone has a high false positive rate of up to 50%.
 
Does establishment of a definitive diagnosis of LFS lead to management changes?
 
Yes. In the majority of cases, treatment and management will be unaffected by genetic testing, as individuals with a negative genetic test are likely to be treated as presumed LFS. However, there are some situations in which genetic testing may impact management. A positive test will facilitate the work-up for cancer susceptibility syndromes when multiple conditions are considered. Knowledge of mutation status may also assist in decision-making for prophylactic mastectomy by providing more definitive risk estimates.
 
Do the management changes result in improved health outcomes?
 
Yes. Outcomes are improved when a definitive diagnosis is made by avoiding the need for further testing to determine whether a cancer susceptibility syndrome is present. Better estimation of risk for breast cancer improves the capacity for informed decision-making regarding prophylactic mastectomy.
 
Testing Asymptomatic Individuals to Determine Future Risk of LFS
There is limited direct evidence on the clinical utility of genetic testing in this population. An indirect chain of evidence can provide evidence of clinical utility if all the links in the chain of evidence are intact. The elements contributing to the indirect chain of evidence are derived from evidence review 2.04.91 (general approach to genetic testing). The following series of questions represent the indirect chain of evidence for testing asymptomatic individuals to determine future risk of disease.
 
In summary the evidence of clinical utility of TP53 testing is limited. One observational study reported improved survival for screened patients. However, this study is limited by the observational design that included self-selection into screening protocols, likely resulting in selection bias. An indirect chain of evidence can demonstrate clinical utility of genetic testing for TP53 mutations. For diagnosis, a positive genetic test will increase the certainty of LFS, facilitate the overall workup for cancer susceptibility syndromes, and assist in decision-making for prophylactic mastectomy. For asymptomatic family members who have a close relative with a pathogenic mutation, genetic testing can confirm or exclude the presence of a mutation, and direct future screening interventions that are likely to improve outcomes.
 
2018 Update
A literature search conducted using the MEDLINE database through May 2018 did not reveal any new information that would prompt a change in the coverage statement.
 
2019 Update
Annual policy review completed with a literature search using the MEDLINE database through May 2019. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Testing for Suspected li-fraumeni syndrome
 In a 2015 study, 88 consecutive patients with adrenocortical carcinoma were evaluated (Wasserman, 2015). Direct sequencing of exons 2 through 11 together with multiplex ligation-dependent probe amplification was used to identify pathogenic variants. For the entire population, 50% of individuals had a pathogenic variant detected. The detection rate varied by age, with 58% of individuals younger than 12 years of age having a pathogenic variant compared with 25% of individuals between ages 12 and 20.
 
O’Shea et al retrospectively analyzed 123 individuals (118 women, 5 men) in Ireland undergoing full TP53sequencing (O’Shea, 2018). Classic criteria for LFS or Li-Fraumeni like syndrome were met by 64 (52%) individuals, none of whom was TP53-positive. Of the 59 (48%) individuals who did not meet classic criteria, 2 had pathogenic TP53 variants (3% detection rate), showing that broadened testing criteria may be beneficial. It was noted that the detection rate of this study (1.6%) was lower than those of similar studies, but the authors suggested that this might be due to the predominance of patients in this cohort with breast cancer, which has an associated lower detection rate.
 
Rana et al published a retrospective, single-laboratory analysis of 38,938 individuals who had undergone TP53 testing to compare different phenotype manifestations found in TP53-positive individuals identified by single-gene testing and multigene panel testing (MGPT) (Rana, 2018). The differences included a significantly lower median age at first cancer for MGPT TP53-positive patients (n=126) than single-gene testing TP53-positive patients (n=96; women: median age, 36 vs 28 years; p<0.001; men: median age, 40 vs 15 years; p<0.004). For breast cancer specifically, median ages were 40 years and 33 years for MGTP TP53-positive and single-gene testing TP53-positive women, respectively (p<0.001). Also, fewer MGPT TP53-positive patients met LFS testing criteria. The study: (1) lacked complete family histories, (2) enrolled predominantly women with breast cancer in the MGPT cohort, (3) used improved technology permitting detection of lower levels of TP53 variants, possibly contributing to misclassification, and (4) assessed a sample too small to investigate other possible factors for phenotypic variation.
 
Practice Guidelines and Position Statements
American Association for Cancer Research
The American Association for Cancer Research (2017) published recommendations for cancer screening and surveillance for patients with LFS (Kratz, 2017). Genetic counseling and clinical TP53 testing should be strongly considered in the following clinical situations:
 
“(i) … proband with an LFS spectrum tumor … prior to age 46 and at least one first- or second-degree relative with an LFS tumor … before the age of 56 years or with multiple tumors, (ii) … proband with multiple malignancies (except two breast cancers), of which at least two belong to the LFS spectrum, before age 46; (iii) … patients with rare tumors such as ACC, choroid plexus carcinoma, or embryonal anaplastic subtype rhabdomyosarcoma independent of family history; and (iv) breast cancer before age 31 years.”
 
Cancer surveillance has been shown to improve overall survival for surveillance and non-surveillance groups and should be offered as soon as either clinical or molecular diagnosis of LFS is established. The following surveillance protocols were recommended for children (birth to age 18) and adults.
 
For children:
    • Complete physical examination every 3-4 months and full neurologic assessment
    • Prompt assessment with primary care physician for any medical concerns
    • Abdominal and pelvic ultrasound every 3-4 months
    • Annual brain MRI
    • Annual whole-body MRI (WBMRI).
 
For adults:
    • Complete physical examination every six months
    • Prompt assessment with primary care physician for any medical concerns
    • Breast awareness (age18 years onward)
    • Clinical breast examination twice per year (age 20 years onward)
    • Annual breast MRI screening (ages 20-75)
    • Consider risk-reducing bilateral mastectomy
    • Annual brain MRI (age 18 years onward)
    • Annual WBMRI
    • Abdominal and pelvic ultrasound every 12 months
    • Upper endoscopy and colonoscopy every 2 to 5 years (age 25 years onward)
    • Annual dermatologic examination.
 
2020 Update
A literature search was conducted through May 2020.  There was no new information identified that would prompt a change in the coverage statement.  
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2021. No new literature was identified that would prompt a change in the coverage statement.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2022. No new literature was identified that would prompt a change in the coverage statement.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Holmfeldt et al performed a genomic analysis of 124 cases of hypodiploid acute lymphoblastic leukemia (ALL) and identified 2 distinct subtypes of this malignancy: near haploid ALL and low hypodiploid ALL (Holmfeldt, 2013). Near haploid cases were defined as having 24–31 chromosomes, and low hypodiploid cases with 32–39 chromosomes. A high frequency of TP53 alterations was identified in both pediatric and adult cases of low hypodiploid ALL (91.2% and 90.9%, respectively). Almost half (43.3%) of the TP53 mutations in pediatric low hypodiploid ALL were present in non-tumor cells with many of the mutations previously reported as LFS associated mutations. All of the TP53 mutations observed in adult cases were somatic.
 
Qian et al investigated germline TP53 variants in childhood ALL (Qian, 2018). Targeted sequencing of TP53 coding regions was performed for 3801 children participating in 2 ALL clinical trials. Within this cohort, 77 patients (2.0%) were found to have 49 unique nonsilent rare TP53 coding variants, with 22 of these variants classified as pathogenic. Children with TP53 pathogenic variants were more likely to have hypodiploid ALL 17/26 (65.4% vs. 1.2%; p<.001). Among the 64 hypodiploid ALL, 17 (27%) were found to have a pathogenic germline TP53 alteration, versus 9/3737 (0.24%) of the non-hypodiploid ALL.
 
Qian et al, introduced above, investigated TP53 variants in childhood ALL (Qian, 2018). When comparing children with ALL to non-ALL controls, the investigators found a significantly higher prevalence of TP53 pathogenic variants in the ALL group (odds ratio, 5.2; p<.001). Furthermore, the presence of TP53 pathogenic variants in children with ALL was associated with several significant findings. These children were more likely to have hypodiploid ALL compared to those without pathogenic variants (65.4% vs. 1.2%; p<.001). Additionally, they exhibited inferior event-free survival and overall survival rates (hazard ratio, 4.2 and 3.9, respectively; both p<.001). Moreover, children with TP53 pathogenic variants had a higher risk of developing secondary cancers, with a 5-year cumulative incidence of 25.1% versus 0.7% in those without such variants (p<.001).

CPT/HCPCS:
0102UHereditary breast cancer related disorders (eg, hereditary breast cancer, hereditary ovarian cancer, hereditary endometrial cancer), genomic sequence analysis panel utilizing a combination of NGS, Sanger, MLPA, and array CGH, with MRNA analytics to resolve variants of unknown significance when indicated (17 genes [sequencing and deletion/duplication])
81351TP53 (tumor protein 53) (eg, Li Fraumeni syndrome) gene analysis; full gene sequence
81352TP53 (tumor protein 53) (eg, Li Fraumeni syndrome) gene analysis; targeted sequence analysis (eg, 4 oncology)
81353TP53 (tumor protein 53) (eg, Li Fraumeni syndrome) gene analysis; known familial variant
81405Molecular pathology procedure, Level 6 (eg, analysis of 6-10 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 11-25 exons, regionally targeted cytogenomic array analysis) Cytogenomic constitutional targeted microarray analysis of chromosome 22q13 by interrogation of genomic regions for copy number and single nucleotide polymorphism (SNP) variants for chromosomal abnormalities (When performing cytogenomic [genome-wide] analysis, for constitutional chromosomal abnormalities. See 81228, 81229, 81349)
81479Unlisted molecular pathology procedure
96040Medical genetics and genetic counseling services, each 30 minutes face to face with patient/family
S0265Genetic counseling, under physician supervision, each 15 minutes

References: Schneider K, Zelley K, Nichols KE et al.(1993) Li-Fraumeni Syndrome. In: Pagon RA, Adam MP, Bird TD, et al., eds. GeneReviews. Seattle (WA)1993.

ARUP Laboratories. Website available online at: http://ltd.aruplab.com/Tests/Pub/2009302. Last accessed April 2014.

Bouaoun L, Sonkin D, Ardin M, et al.(2016) Variations in human cancers: new lessons from the IARC TP53 database and genomics data. Hum Mutat. Sep 2016;37(9):865-876. PMID 27328919

Bougeard G, Renaux-Petel M, Flaman JM, et al.(2015) Revisiting Li-Fraumeni syndrome from TP53 mutation carriers. J Clin Oncol. Jul 20 2015;33(21):2345-2352. PMID 26014290

Chompret A, Abel A, Stoppa-Lyonnet D et al.(2001) Sensitivity and predictive value of criteria for p53 germline mutation screening. J Med Genet 2001; 38(1):43-7.

Gonzalez KD, Noltner KA, Buzin CH et al.(2009) Beyond Li Fraumeni Syndrome: clinical characteristics of families with p53 germline mutations. J Clin Oncol 2009; 27(8):1250-6.

Holmfeldt L, Wei L, Diaz-Flores E, et al.(2013) The genomic landscape of hypodiploid acute lymphoblastic leukemia. Nat Genet. Mar 2013; 45(3): 242-52. PMID 23334668

Hwang SJ, Lozano G, Amos CI et al.(2003) Germline p53 mutations in a cohort with childhood sarcoma: sex differences in cancer risk. Am J Hum Genet 2003; 72(4):975-83.

Kratz CP, Achatz MI, Brugieres L, et al(2017) Cancer screening recommendations for individuals with Li-Fraumeni syndrome. Clin Cancer Res. Jun 1 2017;23(11):e38-e45. PMID 28572266

Kratz CP, Achatz MI, Brugieres L, et al.(2017) Cancer screening recommendations for individuals with Li-Fraumeni syndrome. Clin Cancer Res. Jun 1 2017;23(11):e38-e45. PMID 28572266

Mai PL, Malkin D, Garber JE et al.(2012) Li-Fraumeni syndrome: report of a clinical research workshop and creation of a research consortium. Cancer Genet 2012; 205(10):479-87.

National Comprehensive Cancer Network (NCCN).(2018) NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment Breast and Ovarian. Version 1.2018. https://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf. Accessed June 20, 2018.

National Comprehensive Cancer Network (NCCN).(2018) NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment Breast and Ovarian. Version 1.2018. https://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf. Accessed June 20, 2018.

O'Shea R, Clarke R, Berkley E, et al.(2018) Next generation sequencing is informing phenotype: a TP53 example. Fam Cancer. Jan 2018;17(1):123-128. PMID 28509937

O'Shea R, Clarke R, Berkley E, et al.(2018) Next generation sequencing is informing phenotype: a TP53 example. Fam Cancer. Jan 2018;17(1):123-128. PMID 28509937

Ognjanovic S, Olivier M, Bergemann TL et al.(2012) Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer 2012; 118(5):1387-96.

Petitjean A, Mathe E, Kato S, et al.(2007) Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. Jun 2007;28(6):622-629. PMID 17311302

Qian M, Cao X, Devidas M, et al.(2018) TP53 Germline Variations Influence the Predisposition and Prognosis of B-Cell Acute Lymphoblastic Leukemia in Children. J Clin Oncol. Feb 20 2018; 36(6): 591-599. PMID 29300620

Rana HQ, Gelman R, LaDuca H, et al.(2018) Differences in TP53 mutation carrier phenotypes emerge from panel-based testing. J Natl Cancer Inst. Feb 26 2018. PMID 29529297

Raymond VM, Else T, Everett JN et al.(2013) Prevalence of germline TP53 mutations in a prospective series of unselected patients with adrenocortical carcinoma. J Clin Endocrinol Metab 2013; 98(1):E119-25.

Schneider K, Zelley K, Nichols KE, et al(2013) Li-Fraumeni Syndrome. In: Pagon RA, Adam MP, Bird TD, et al., eds. GeneReviews. Seattle, WA: University of Washington; 2013.

Singh AD, Medina CA, Singh N, et al.(2016) Fine-needle aspiration biopsy of uveal melanoma: outcomes and complications. Br J Ophthalmol. Apr 2016;100(4):456-462. PMID 26231747

Sorrell AD, Espenschied CR, Culver JO et al.(2013) Tumor protein p53 (TP53) testing and Li-Fraumeni syndrome : current status of clinical applications and future directions. Mol Diagn Ther 2013; 17(1):31-47.

Villani A, Tabori U, Schiffman J et al.(2011) Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: a prospective observational study. Lancet Oncol 2011; 12(6):559-67.

Wagner J, Portwine C, Rabin K, et al.(1994) High frequency of germline p53 mutations in childhood adrenocortical cancer. J Natl Cancer Inst. Nov 16 1994;86(22):1707-1710. PMID 7966399

Wasserman JD, Novokmet A, Eichler-Jonsson C, et al.(2015) Prevalence and functional consequence of TP53 mutations in pediatric adrenocortical carcinoma: a children's oncology group study. J Clin Oncol. Feb 20 2015;33(6):602-609. PMID 25584008


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.