Medical Policy

Policy Num:       11.003.107
Policy Name:     Germline Genetic Testing for Pancreatic Cancer Susceptibility Genes (ATM, BRCA1, BRCA2, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PALB2, PMS2, STK11, and TP53)

Policy ID:           [11.003.107]  [Ac / B / M +/ P+]  [2.04.148]


Last Review:        March 03, 2025
Next Review:        March 20, 2026

Related Policies:

11.003.030- Genetic Testing for BRCA1 or BRCA2 for Hereditary Breast/Ovarian Cancer Syndrome and Other High-Risk Cancers
11.003.028- Genetic Testing for Lynch Syndrome and Other Inherited Colon Cancer Syndromes
11.003.022- Genetic Testing for Li-Fraumeni Syndrome
11.003.027- Moderate Penetrance Variants Associated With Breast Cancer in Individuals at High Breast Cancer Risk
11.003.036- Genetic Testing for Familial Cutaneous Malignant Melanoma
11.003.019- Genetic Cancer Susceptibility Panels Using Next Generation Sequencing
11.003.059- Genetic Testing for Hereditary Pancreatitis

Germline Genetic Testing for Pancreatic Cancer Susceptibility Genes (ATM, BRCA1, BRCA2, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PALB2, PMS2, STK11, and TP53)

Population References No. Populations Interventions Comparators Outcomes
                                           1 Individuals:
  • With previously untreated, locally advanced or metastatic pancreatic cancer
Interventions of interest are:
  • Genetic testing for a BRCA1, BRCA2, or PALB2 variant to select treatment with platinum-based chemotherapy
Comparators of interest are:
  • Standard of care without genetic testing
Relevant outcomes include:
  • Overall survival
  • Disease-specific survival
  • Test accuracy
  • Test validity
                                          2 Individuals:
  • With metastatic pancreatic cancer
Interventions of interest are:
  • Genetic testing for a BRCA1 or BRCA2 variant to select targeted treatment
Comparators of interest are:
  • Standard of care without genetic testing
Relevant outcomes include:
  • Overall survival
  • Disease-specific survival
  • Test accuracy
  • Test validity
                                          3 Individuals:
  • With pancreatic cancer
Interventions of interest are:
  • Genetic testing for ATM, CDK2NA, EPCAM, MMR genes (MLH1, MSH2, MSH6, PMS2), , STK11, and TP53variants to guide treatment
Comparators of interest are:
  • Standard of care without genetic testing
Relevant outcomes include:
  • Overall survival
  • Disease-specific survival
  • Test accuracy
  • Test validity
                                          4 Individuals:
  • Who are asymptomatic and at high risk for hereditary pancreatic cancer
Interventions of interest are:
  • Testing for genes associated with hereditary pancreatic cancer to guide surveillance for pancreatic cancer
Comparators of interest are:
  • No genetic testing
Relevant outcomes include:
  • Overall survival
  • Disease-specific survival
  • Test accuracy
  • Test validity

Summary

Description

Pancreatic cancer is the fourth leading cause of cancer death in the United States, accounting for 8.3% of all cancer deaths in 2023. Multiple genetic syndromes are associated with an increased risk for pancreatic cancer, and approximately 10% to 15% of patients with pancreatic cancer are thought to have a hereditary susceptibility to the disease. Germline genetic testing for pancreatic cancer susceptibility genes is proposed to guide treatment decisions in patients with pancreatic cancer, and to inform decisions about surveillance in asymptomatic patients at high risk of pancreatic cancer.

Summary of Evidence

For individuals who have pancreatic cancer who receive testing for a BRCA1, BRCA2, or PALB2 variant to guide selection for first-line treatment, the evidence includes observational studies. Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1, BRCA2, and PALB2 variants, including among those who do not have a family history of pancreatic cancer. Observational studies have reported a survival advantage when patients with a BRCA or PALB2 variant were treated with platinum-based chemotherapy regimens compared to non-platinum-based regimens. Although these studies are limited by their small sample sizes and retrospective designs, the consistency and magnitude of benefit across studies suggests that genetic testing for these variants to aid in treatments decisions is a reasonable approach. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals who have pancreatic cancer who receive testing for a BRCA1 or BRCA2 variant to guide selection for targeted treatment, the evidence includes observational studies and 1 randomized controlled trial. Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1 or BRCA2 variants, including among those who do not have a family history of pancreatic cancer. A placebo-controlled trial of olaparib as maintenance therapy in patients with germline BRCA1 or BRCA2 variants and metastatic pancreatic cancer found longer progression-free survival with olaparib (7.4 months vs. 3.8 months; hazard ratio, 0.53; 95% confidence interval 0.35 to 0.82; p=.04 ). The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

For individuals with pancreatic cancer who receive genetic testing for ATM, CDK2NA, EPCAM, MMR genes (MLH1, MSH2, MSH6, PMS2), STK11, and TP53 to guide treatment, the evidence includes observational studies. Relevant outcomes are overall survival, disease-specific survival, test accuracy, and test validity. Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with disease-associated variants, including among those who do not have a family history of the disease. However, there is no direct evidence comparing health outcomes in patients tested or not tested for a variant. Additionally, there are no targeted treatments for pancreatic cancer based on these genes, and management changes that would result from testing these genes are unclear. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

For individuals who are asymptomatic and at high risk for hereditary pancreatic cancer who receive testing for genes associated with hereditary pancreatic cancer, the evidence includes observational studies. There is no direct evidence comparing health outcomes in patients tested or not tested for a variant. There is indirect evidence from 2 comparative observational studies of high-risk individuals under surveillance that the risk of progression to pancreatic cancer is higher among individuals with a known pathogenic variant than in individuals identified as at-risk based on family history alone. There is also evidence from prospective observational studies that surveillance of high-risk individuals can identify pancreatic cancer and precursor lesions. In 1 analysis of 76 high-risk individuals under surveillance, survival was better in those who had surgery due to detection of either low- or high-risk neoplastic precursor lesions (n=71) compared to those who had advanced to unresectable disease (n=5). Although observational studies have demonstrated that surveillance can identify pancreatic cancer and precursor lesions in asymptomatic individuals, it is not possible to conclude from this body of evidence that surveillance improves survival. Longer survival time observed in individuals undergoing surveillance could be due to earlier identification of the disease (downstaging) and not the effects of early intervention and treatment. Additionally, evidence is too limited to determine if selecting patients for surveillance based on genetic testing leads to better outcomes than using criteria such as family history alone. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Additional Information

Not applicable

Objective

The objective of this evidence review is to evaluate the clinical validity and clinical utility of genetic testing of individuals with or at high-risk for hereditary pancreatic cancer and to determine if its use improves the net health outcome.

Policy Statements

Genetic testing for BRCA1, BRCA2, and PALB2 variants to guide selection for treatment with platinum-based chemotherapy in previously untreated individuals with locally advanced or metastatic pancreatic cancer may be considered medically necessary.

Genetic testing for BRCA1 and BRCA2 variants to guide selection for treatment with olaparib (Lynparza) in individuals with pancreatic cancer may be considered medically necessary.

Genetic testing for ATM, CDK2NA, EPCAM, MMR genes (MLH1, MSH2, MSH6, PMS2), STK11, and TP53 in individuals with pancreatic cancer is considered investigational unless the individual meets criteria for testing as specified in another policy (see policy guidelines).

Genetic testing for ATM, BRCA1, BRCA2, CDK2NA, EPCAM, MMR genes (MLH1, MSH2, MSH6, PMS2), PALB2, STK11, and TP53 in asymptomatic individuals at high risk for hereditary pancreatic cancer is considered investigational unless the individual meets criteria for testing as specified in another policy (see policy guidelines).

Policy Guidelines

Related Policies on Hereditary Cancer Syndromes

Testing At-Risk Relatives

Individuals are considered at high risk for hereditary pancreatic cancer if they have:

For familial assessment, 1st-, 2nd-, and 3rd-degree relatives are blood relatives on the same side of the family (maternal or paternal).

At-risk relatives primarily refer to first-degree relatives. However, some judgment must be permitted, e.g., in the case of a small family pedigree, when extended family members may need to be included in the testing strategy.

Targeted Variant Testing

It is recommended that, when possible, initial genetic testing for variants associated with hereditary pancreatic cancer be performed in an affected family member so that testing in unaffected family members can focus on the pathogenic variant found in the affected family member. In unaffected family members of potential hereditary pancreatic cancer families, most test results will be negative and uninformative. Therefore, it is strongly recommended that an affected family member be tested first whenever possible to adequately interpret the test. Should a variant be found in an affected family member(s), DNA from an unaffected family member can be tested specifically for the same variant of the affected family member without having to sequence the entire gene.

Genetic Counseling

Experts recommend formal genetic counseling for individuals who are at risk for inherited disorders and who wish to undergo genetic testing. Interpreting the results of genetic tests and understanding risk factors can be difficult for some individuals ; genetic counseling helps individuals understand the impact of genetic testing, including the possible effects the test results could have on the individual or their family members. It should be noted that genetic counseling may alter the utilization of genetic testing substantially and may reduce inappropriate testing; further, genetic counseling should be performed by an individual with experience and expertise in genetic medicine and genetic testing methods.

Coding

See the Codes table for details.

Benefit Application

BlueCard/National Account Issues

Some Plans may have contract or benefit exclusions for genetic testing.

Benefits are determined by the group contract, member benefit booklet, and/or individual subscriber certificate in effect at the time services were rendered. Benefit products or negotiated coverages may have all or some of the services discussed in this medical policy excluded from their coverage.

Background

Pancreatic Cancer Epidemiology

Pancreatic cancer is the fourth leading cause of cancer death in the U.S., accounting for 8.3%8.5% of all cancer deaths in 20232024.1, The disease has a poor prognosis, with only 12.5%12.8% of patients surviving to 5 years. Five-year survival for localized pancreatic cancer is 41.6%44.0% but most symptomatic patients have advanced, incurable disease at diagnosis.

Genetics and Pancreatic Cancer

Approximately 10% to 15% of patients with pancreatic cancer are thought to have a hereditary susceptibility to the disease.2, Multiple genetic syndromes, including hereditary breast and ovarian cancer syndrome, are associated with an increased risk for pancreatic cancer. Five percent to 9% of pancreatic ductal adenocarcinomas (PDACs) develop in patients with a germline BRCA or PALB2 variant, with higher rates observed in those with a family or personal history of pancreatic cancer or other BRCA-related malignancies.3, The incidence of germline PALB2 variants in persons with PDAC is estimated to be between 0.6% and 2.1%.4,

Having a first-degree relative with pancreatic cancer increases an individual's risk of developing pancreatic cancer, and the degree of risk increases depending on the number of affected relatives (Table 1).5, Individuals are considered at high-risk for hereditary pancreatic cancer if they have 2 relatives with pancreatic cancer where 1 is a first-degree relative, have 3 or more relatives with pancreatic cancer, or have a history of hereditary pancreatitis. In 80% of pancreatic cancer patients with a family history of pancreatic cancer, the genetic basis of the inherited predisposition is unknown.6,

Table 1. Family History and Pancreatic Cancer Risk
Number of FDR with Pancreatic Cancer Increased Risk
1 affected FDR 4.6-fold
2 affected FDR 6.4-fold
3 affected FDR 32-fold
 Sources: American Society of Clinical Oncology; 2, American College of Gastroenterology 7, FDR: first-degree relative.

Germline genetic testing for pancreatic cancer susceptibility genes has several proposed purposes. In patients with pancreatic cancer, the purpose of genetic testing would be to guide treatment decisions (e.g., selection of platinum-based chemotherapy for first-line treatment, targeted treatment with a poly ADP ribose polymerase [PARP] inhibitor). In asymptomatic patients at high risk of pancreatic cancer (e.g., due to family history or other clinical factors), the purpose of genetic testing would be to inform decisions about surveillance for early detection of pancreatic cancer. Because the incidence of pancreatic cancer in the general population is low, with a lifetime risk of approximately 1.6%, screening is not recommended for patients who are not at high-risk, but patients with a family history of pancreatic cancer or a syndrome associated with increased risk of pancreatic cancer are potential targets for surveillance.

Regulatory Status

Testing for variants associated with pancreatic cancer is typically done by direct sequence analysis or next-generation sequencing. A number of laboratories offer to test for the relevant genes, either individually or as panels.

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 (CLIA). Laboratories that offer laboratory-developed tests must be licensed by the CLIA for high-complexity testing. To date, the U.S. Food and Drug Administration (FDA) has chosen not to require any regulatory review of this test.

In December 2019, the FDA approved olaparib (Lynparza, AstraZeneca Pharmaceuticals LP) for the maintenance treatment of adult patients with deleterious or suspected deleterious germline BRCA-mutated metastatic pancreatic adenocarcinoma, as detected by an FDA approved test, whose disease has not progressed on at least 16 weeks of a first-line platinum-based chemotherapy regimen. Also in 2019, BRACAnalysis CDx received expanded FDA approval for use as a companion diagnostic for Lynparza (olaparib) in pancreatic cancer patients.8,

Rationale

This evidence review was created in February 2020 and was updated with a search of the PubMed database. The most recent literature update was performed through December 23, 2024.

Evidence reviews assess whether a medical test is clinically useful. A useful test provides information to make a clinical management decision that improves the net health outcome. That is, the balance of benefits and harms is better when the test is used to manage the condition than when another test or no test is used to manage the condition. The first step in assessing a medical test is to formulate the clinical context and purpose of the test. The test must be technically reliable, clinically valid, and clinically useful for that purpose. Evidence reviews assess the evidence on whether a test is clinically valid and clinically useful. Technical reliability is outside the scope of these reviews, and credible information on technical reliability is available from other sources.

Promotion of greater diversity and inclusion in clinical research of historically marginalized groups (e.g., People of Color [African-American, Asian, Black, Latino and Native American]; LGBTQIA (Lesbian, Gay, Bisexual, Transgender, Queer, Intersex, Asexual); Women; and People with Disabilities [Physical and Invisible]) allows policy populations to be more reflective of and findings more applicable to our diverse members. While we also strive to use inclusive language related to these groups in our policies, use of gender-specific nouns (e.g., women, men, sisters, etc.) will continue when reflective of language used in publications describing study populations.

Population Reference No. 1

Genetic Testing for a BRCA1, BRCA2, or PALB2 Variant to Select First-Line Treatment

Clinical Context and Test Purpose

The purpose of genetic testing for a BRCA1, BRCA2, or PALB2 variant in individuals with pancreatic cancer is to identify individuals who might benefit from a platinum-containing chemotherapy regimen.

The following PICO was used to select literature to inform this review.

Population

The relevant population of interest is individuals with previously untreated, locally advanced or metastatic pancreatic cancer.

Interventions

The test being considered is genetic testing for a BRCA1, BRCA2, or PALB2 variant.

Comparators

Alternatives to genetic testing would be treatment as usual without genetic testing.

Outcomes

The potential beneficial outcomes of primary interest would be improvements in overall survival (OS) and disease-specific survival in individuals with pancreatic cancer.

Potential harmful outcomes are those resulting from false-positive or false-negative test results. False-positive test results can lead to unnecessary clinical management changes or unnecessary cascade testing for other cancers. False-negative test results can lead to the absence of clinical management changes.

Study Selection Criteria

For the evaluation of the clinical validity of the genetic test, studies that reported on the sensitivity and specificity and/or diagnostic yield of the test were considered, including curated sources of information on genes associated with increased risk of pancreatic cancer (eg, summaries from professional societies).

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1, BRCA2, and PALB2 variants.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Several retrospective observational studies and an uncontrolled subgroup analysis from a randomized controlled trial of veliparib have reported a survival advantage for pancreatic cancer patients with BRCA or PALB2 pathogenic variants who received platinum-containing chemotherapy (Tables 2 and 3).

Golan et al (2014) analyzed survival data and clinical characteristics from databases of pancreatic cancer patients treated at 3 institutions between 1994 and 2012, including 71 patients with BRCA1 or BRCA2 variants.9, Longer median overall survival was observed in patients with BRCA variants who received platinum-based chemotherapy compared to those who received non-platinum-based chemotherapies (22 months [range, 6 to 27] vs. 9 months [range, 4 to 12]; p=.039 ).

Three retrospective cohort studies used similar methods to compare survival outcomes in patients with or without BRCA or PALB2 variants who were treated with platinum-based chemotherapy.10,11,4, In these studies, patients with a pathogenic variant were matched to control patients on prognostic factors such as age at diagnosis, sex, and stage of disease. All of these studies reported a survival advantage when variant-positive patients were treated with platinum versus non-platinum-based regimens, while there was no advantage for platinum-based therapy in patients who did not harbor a BRCA or PALB2 variant (See Table 3).

Limitations of these studies are summarized in Tables 4 and 5. Major limitations include the studies' small sample sizes and retrospective designs. The timing of genetic testing varied within the patient cohorts (e.g. some patients were tested before and others after their pancreatic cancer diagnosis). It is possible that patients who survived their pancreatic ductal adenocarcinoma (PDAC) diagnosis longer were more likely to undergo genetic testing. Because many control patients were not tested, some may have been variant-positive. However, this is less of a concern because this would have biased results toward the null. There was also heterogeneity in the timing and type of chemotherapy regimens patients received. Although the studies attempted to control for confounding by matching patients on important prognostic factors or using statistical analysis methods, the potential for unmeasured confounding decreases confidence in the results. Despite these limitations, consistency in the magnitude and direction of results across studies suggest that a strategy of testing for these variants to aid in decision-making about first-line treatment is a reasonable approach.

O'Reilly et al (2020) conducted a randomized controlled trial (RCT) of platinum-based chemotherapy with or without the poly ADP ribose polymerase (PARP) inhibitor veliparib in patients with previously untreated, locally advanced or metastatic pancreatic cancer and a BRCA or PALB2 germline variant. Two-year OS rate for the entire cohort was 30.6% (95% confidence interval [CI], 17.8% to 44.4%), and 3-year OS rate for the entire cohort was 17.8% (95% CI, 8.1% to 30.7%).Overall survival did not differ significantly when veliparib was added to the platinum-based regimen. The trial was not designed to compare platinum-based versus standard chemotherapy, but it does provide uncontrolled evidence of the effectiveness of platinum-containing chemotherapy in patients with germline pathogenic BRCA or PALB2 variants. The major limitation of this analysis was the lack of a control group of patients who did not receive platinum-based chemotherapy.

Table 2. Platinum-based Chemotherapy for Pancreatic Cancer Treatment in Patients with a BRCA1, BRCA2, or PALB2 Variant: Study Characteristics
Study Study Type Location Dates Participants Pancreatic Cancer Treatment Regimen
Golan et al (2014)9, Retrospective cohort Canada and Israel, 3 sites Patients diagnosed between January 1994 and December 2012 71 patients with PDAC and BRCA1 (n=21), BRCA2 (n =49), or both (n=1) variants
Stage 1 (1.4%), stage 2 (27%), stage 3 (23%), stage 4 (48%); 1 missing data on stage
22 patients in the stage 3/4 group received
platinum-based treatment. The majority of platinum-treated patients received gemcitabine and cisplatin, 1 patient received gemcitabine and oxaliplatin and 3 patients received FOLFIRINOX
 
O'Reilly et al (2020)3, RCT (platinum-based chemotherapy + veliparib vs. platinum-based chemotherapy alone) US, Canada, Israel, 6 sites Patients enrolled between 2014 and 2018 52 patients with untreated locally advanced or metastatic PDAC and germline pathogenic variants in BRCA or PALB2 Arm A: cisplatin, gemcitabine, and veliparib
Arm B: cisplatin and gemcitabine
Reiss et al (2018)4, Retrospective cohort US, single site Patients diagnosed between 1995 and 2016 29 patients diagnosed with either locally advanced or metastatic PDAC with a known pathogenic germline BRCA1, BRCA2, or PALB2 variant.
58 controls were either confirmed variant noncarriers or had not been tested.
Cohorts matched by age at diagnosis, year of diagnosis, stage at diagnosis, and sex
Of the 87 patients, 4 variant-positive patients (13.8%) and 12 control patients (20.7%) received no systemic treatment of any kind. Treatment history for 1 control patient was unknown.
Patients who were variant-positive and did receive systemic therapy:18 of 25 (72.0%) received platinum-based therapy 48.0% oxaliplatin, 12.0% received cisplatin, 8.0% received both oxaliplatin and cisplatin, and the exact regimen was unknown for 1 patient.
Control patients, 60.8% received platinum-based therapy (96.4% oxaliplatin, 1 cisplatin (3.5%), regimen unknown for 1 patient..
Yu et al (2019)11, Retrospective cohort US, single site Patients diagnosed between January 1, 1995 and March 31, 2018 32 patients with nonmetastatic PDAC who had undergone curative intent surgical resection and had a known pathogenic germline variant in BRCA1, BRCA2, or PALB2
64 control patients who were either confirmed variant noncarriers or had not been tested.
Cohorts matched by age at diagnosis, year of diagnosis, sex, and disease stage.
42% in the variant-positive group and 17% in the variant-negative group received perioperative platinum chemotherapy (p=.01). Of these, 3 patients in the variant-positive group and 10 in the variant-negative group received perioperative FOLFIRINOX, the remaining patients received other platinum-containing regimens.
12 patients in the variant-positive group and 23 in the variant-negative group received palliative platinum chemotherapy upon recurrence.
Wattenberg et al (2020)10, Retrospective cohort US, single site Patients diagnosed between July 2011 and March 2018 26 patients with locally advanced or metastatic PDAC and pathogenic germline variants in BRCA1 (n=5), BRCA2 (n=17) or PALB2 (n=4) who had received platinum-based therapy
52 control patients who were either confirmed non-carriers or had not been tested
Cohorts matched by age at diagnosis, sex, and race.
Variant-positive patients: FOLFIRINOX (n=10; 38.5%), FOLFOX (n=10; 38.5%) and cisplatin plus gemcitabine (n=6; 23.0%). 1 patient received FOLFIRINOX followed by cisplatin plus gemcitabine.
Control patients: FOLFIRINOX (n=39; 75%), FOLFOX (n=11; 21.1%), cisplatin plus gemcitabine (n=1; 1.9%) and cisplatin plus gemcitabine plus nab-paclitaxel (n=1; 1.9%).
Platinum therapy was most commonly received in the first-line setting regardless of cohort 80.7% of variant-positive patients 67.3% of control patients (p=.21).
Significantly more control patients received FOLFIRINOX (75% vs. 38.5%; p=.0016) and significantly more variant-positive patients received cisplatin plus gemcitabine (23.1% vs. 1.9%; p=0.0021)
 FOLFIRINOX: folinic acid, fluorouracil, irinotecan and oxaliplatin; FOLFOX: folinic acid, fluorouracil and oxaliplatin, or cisplatin/gemcitabine; PDAC: pancreatic ductal adenocarcinoma; RCT: randomized controlled trial. 
Table 3. Platinum-based Chemotherapy for Pancreatic Cancer Treatment in Patients with a BRCA1, BRCA2, or PALB2 Variant: Study Results
Study Overall Survival Median Overall Survival Median Progression-Free Survival
Golan et al (2014)9, Probability of survival, platinum-based (n=22) vs. non-platinum-based (n=21) therapy:
12 months: 0.70 (95% CI, 0.44 to 0.85) vs. 0.26 (95% CI, 0.08 to 0.48)
36 months: 0.16 (95% CI, 0.01 to 0.46) vs. 0.07 (95% CI, 0.01 to 0.26)
Stage 3/4 patients treated with platinum-based chemotherapy vs. non-platinum-based chemotherapy (N=43):
22 months (95% CI, 6 to 27) vs. 9 months (95% CI, 4 to 12); p=.039
(Disease-free survival) Patients with stage 1 or 2 disease (n=20): 13 months (95% CI, 6 to 19 months)
Probability of remaining disease free:
1 year: 0.54 (95% CI, 0.29 to 0.74)
5 years: 0.27 (95% CI, 0.09 to 0.5)
O'Reilly et al (2020)3, 9/50 (18%) alive at final data cutoff
2-year OS: 30.6% (95% CI, 17.8% to 44.4%)
3-year OS: 17.8% (95% CI, 8.1% to 30.7%)
Arm A: 15.5 months (95% CI, 12.2 to 24.3 months)
Arm B: 16.4 months (95% CI, 11.7 to 23.4)
Arm A: 10.1 months (95% CI, 6.7 to 11.5 months)
Arm B: 9.7 months (95% CI, 4.2 to 13.6)
Reiss et al (2018)4, 1-year OS: 94% Control: 60%
HR, 0.25; 95% CI, 0.1 to 0.61; p=.002
In patients not treated with platinum, there was no significant difference in OS between groups (HR, 0.54; 95% CI, 0.25 to 1.17; p=.12).
BRCA-or PALB2 variant- positive: Undefined at a median follow-up of 20.1 months
Control: 15.5 months
 
Yu et al (2019)11,   Variant-positive group vs. control (all patients): 46.6 months vs. 23.2 months; HR, 0.49; 95% CI, 0.27 to 0.88
Subgroup who received platinum treatment at any time, variant-positive vs. control: 47.7 months vs. 23.1 months; HR, 0.30; 95% CI, 0.13 to 0.70
Subgroup who did not receive platinum treatment, variant-positive vs. control: HR, 0.52; 95% CI, 0.12 to 2.24
 
Wattenberg et al (2020)10,   Variant-positive group vs. control: 24.6 months vs. 18.8 months (p=.0467)
No difference in outcomes between groups when platinum was administered in the second line or later.
10.1 months vs 6.9 months
 CI: confidence interval; HR: hazard ratio; OS: overall survival.
Table 4. Study Relevance Limitations
Study Populationa Interventionb Comparatorc Outcomesd Duration of Follow-upe
Golan et al (2014)9, stage of disease varied 3. timing of testing varied      
O'Reilly et al (2020)3,     No variant-negative control group    
Reiss et al (2018)4, stage of disease varied

3. timing of testing varied

     
Yu et al (2019)11, stage of disease varied 3. timing of testing varied      
Wattenberg et al (2020)10, stage of disease varied 3. timing of testing varied      
 The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment.  a Population key: 1. Intended use population unclear; 2. Clinical context is unclear; 3. Study population is unclear; 4. Study population not representative of intended use. b Intervention key: 1. Not clearly defined; 2. Version used unclear; 3. Delivery not similar intensity as comparator; 4.Not the intervention of interest. c Comparator key: 1. Not clearly defined; 2. Not standard or optimal; 3. Delivery not similar intensity as intervention; 4. Not delivered effectively. d Outcomes key: 1. Key health outcomes not addressed; 2. Physiologic measures, not validated surrogates; 3. No CONSORT reporting of harms; 4. Not establish and validated measurements; 5. Clinical significant difference not prespecified; 6. Clinical significant difference not supported. e Follow-Up key: 1. Not sufficient duration for benefit; 2. Not sufficient duration for harms.
Table 5. Study Design and Conduct Limitations
Study Allocationa Blindingb Selective Reportingc Data Completenessd Powere Statisticalf
Golan et al (2014)9, 1. not randomized 1. not blinded        
O'Reilly et al (2020)3,            
Reiss et al (2018)4, 1. not randomized 1. not blinded   1. missing data on chemotherapy regimen received    
Yu et al (2019)11, 1. not randomized 1. not blinded        
Wattenberg et al (2020)10, 1. not randomized 1. not blinded        
 The study limitations stated in this table are those notable in the current review; this is not a comprehensive gaps assessment. a Allocation key: 1. Participants not randomly allocated; 2. Allocation not concealed; 3. Allocation concealment unclear; 4. Inadequate control for selection bias. b Blinding key: 1. Not blinded to treatment assignment; 2. Not blinded outcome assessment; 3. Outcome assessed by treating physician. c Selective Reporting key: 1. Not registered; 2. Evidence of selective reporting; 3. Evidence of selective publication. d Data Completeness key: 1. High loss to follow-up or missing data; 2. Inadequate handling of missing data; 3. High number of crossovers; 4. Inadequate handling of crossovers; 5. Inappropriate exclusions; 6. Not intent to treat analysis (per protocol for noninferiority trials). e Power key: 1. Power calculations not reported; 2. Power not calculated for primary outcome; 3. Power not based on clinically important difference. f Statistical key: 1. Analysis is not appropriate for outcome type: (a) continuous; (b) binary; (c) time to event; 2. Analysis is not appropriate for multiple observations per patient; 3. Confidence intervals and/or p values not reported; 4. Comparative treatment effects not calculated.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

There is no direct evidence from RCTs of the clinical utility of germline testing for BRCA or PALB2 variants in patients with pancreatic cancer.

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

The consistency and magnitude of benefit across studies suggests that genetic testing for these variants to aid in treatments decisions is a reasonable approach.

Section Summary: Genetic Testing for a BRCA1, BRCA2, or PALB2 Variant to Select First-Line Treatment

Retrospective cohort studies and an uncontrolled analysis from a randomized controlled trial have reported a survival advantage when patients with a BRCA or PALB2 variant were treated with platinum-based chemotherapy regimens compared to non-platinum-based regimens. Although these studies are limited by their small sample sizes and retrospective designs, the consistency and magnitude of benefit across studies suggests that genetic testing for these variants to aid in treatments decisions is a reasonable approach.

 

For individuals who have pancreatic cancer who receive testing for a BRCA1, BRCA2, or PALB2 variant to guide selection for first-line treatment, the evidence includes observational studies. Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1, BRCA2, and PALB2 variants, including among those who do not have a family history of pancreatic cancer. Observational studies have reported a survival advantage when patients with a BRCA or PALB2 variant were treated with platinum-based chemotherapy regimens compared to non-platinum-based regimens. Although these studies are limited by their small sample sizes and retrospective designs, the consistency and magnitude of benefit across studies suggests that genetic testing for these variants to aid in treatments decisions is a reasonable approach. The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

 

Population

Reference No. 1

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 2

Genetic Testing for a BRCA1 or BRCA2 Variant to Select Targeted Treatment

Clinical Context and Test Purpose

The purpose of genetic testing for a BRCA1 or BRCA2 variant in individuals with pancreatic cancer is to guide selection of targeted treatment for pancreatic cancer.

The following PICO was used to select literature to inform this review.

Population

The relevant population of interest is individuals with metastatic or recurrent pancreatic cancer.

Interventions

The test being considered is genetic testing for a BRCA1 or BRCA2 variant to select targeted treatment with PARP inhibitors such as olaparib.

Comparators

Alternatives to genetic testing would be treatment as usual without genetic testing.

Outcomes

The potential beneficial outcomes of primary interest would be improvements in OS and disease-specific survival in individuals with pancreatic cancer.

Potential harmful outcomes are those resulting from false-positive or false-negative test results. False-positive test results can lead to unnecessary clinical management changes or unnecessary cascade testing for other cancers. False-negative test results can lead to the absence of clinical management changes.

Study Selection Criteria

For the evaluation of the clinical validity of the genetic test, studies that reported on the sensitivity and specificity and/or diagnostic yield of the test were considered, including curated sources of information on genes associated with increased risk of pancreatic cancer (eg, summaries from professional societies).

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1 and BRCA2 variants.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Review of Evidence

Golan et al (2019) conducted a placebo-controlled RCT of olaparib as maintenance therapy in patients with germline BRCA1 or BRCA2 variants and metastatic pancreatic cancer (Tables 6 and 7).12, Of 3315 patients screened, 247 (7.5%) had a germline BRCA variant. Median progression-free survival was longer in the olaparib group, but there was no difference in OS.

Table 6. RCT of Targeted Treatment in Patients With Pancreatic Cancer: Study Characteristics
Study Countries Sites Dates Participants Interventions
          Active Comparator
Golan et al (2019)12,
NCT02184195
Multiple 119 2014-2019 N=144


Patients with a germline BRCA variant and metastatic pancreatic adenocarcinoma that had not progressed during first-line platinum-based chemotherapy
Olaparib Placebo
  NCT: National Clinical Trial 02184195, Multicentre Study of Maintenance Olaparib Monotherapy in Patients With gBRCA Mutated Metastatic Pancreatic Cancer Whose Disease Has Not Progressed on First Line Platinum Based Chemotherapy; RCT: randomized controlled trial.
Table 7. RCT of Targeted Treatment in Patients With Pancreatic Cancer: Results
Study Median Progression-free Survival Median Overall Survival Serious Adverse Events
Golan et al (2019)12,      
Olaparib 7.4 mos 18.9 mos 24%
Placebo 3.8 mos 18.1 mos 15%
HR (95% CI) 0.53 (0.35 to 0.82);
p=.004
0.91 (0.56 to 1.46)
p=.68
 
  CI: confidence interval; HR: hazard ratio; RCT: randomized controlled trial.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

There are no direct outcome data on the clinical usefulness of testing for confirmation of a BRCA1 or BRCA2 variant in patients with pancreatic cancer (ie, no studies have reported outcomes data for patients tested and not tested for a variant).

Chain of Evidence

Indirect evidence on clinical utility rests on clinical validity. If the evidence is insufficient to demonstrate test performance, no inferences can be made about clinical utility.

A chain of indirect evidence would demonstrate that genetic testing can identify individuals with pathogenic variants associated with pancreatic cancer who would not otherwise be identified, that treatments are available for these patients that would not otherwise be given to patients with pancreatic cancer, and that these treatments improve health outcomes.

Section Summary: Genetic Testing for a BRCA1 or BRCA2 Variant to Select Targeted Treatment

Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1 or BRCA2 variants, including among those who do not have a family history of pancreatic cancer. A placebo-controlled trial of olaparib as maintenance therapy in patients with germline BRCA1 or BRCA2 variants and metastatic pancreatic cancer found longer progression-free survival with olaparib (7.4 months vs. 3.8 months; hazard ratio [HR], 0.53; 95% CI 0.35 to 0.82; P=.04).

For individuals who have pancreatic cancer who receive testing for a BRCA1 or BRCA2 variant to guide selection for targeted treatment, the evidence includes observational studies and 1 randomized controlled trial. Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with BRCA1 or BRCA2 variants, including among those who do not have a family history of pancreatic cancer. A placebo-controlled trial of olaparib as maintenance therapy in patients with germline BRCA1 or BRCA2 variants and metastatic pancreatic cancer found longer progression-free survival with olaparib (7.4 months vs. 3.8 months; hazard ratio, 0.53; 95% confidence interval 0.35 to 0.82; p=.04 ). The evidence is sufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 2

Policy Statement

[X] MedicallyNecessary [ ] Investigational

Population Reference No. 3

Genetic Testing for ATM, CDKN2A , EPCAM, MLH1, MSH2, MSH6, PMS2, STK11, and TP53 to Guide Treatment in Individuals with Pancreatic Cancer

Clinical Context and Test Purpose

The purpose of genetic testing for genes associated with pancreatic cancer in individuals with pancreatic cancer is to guide treatment for pancreatic cancer.

The following PICO was used to select literature to inform this review.

Population

The relevant population of interest is individuals with pancreatic cancer.

Interventions

The test being considered is genetic testing for ATM, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PMS2, STK11, and TP53.

Comparators

Alternatives to genetic testing would be treatment as usual without genetic testing.

Outcomes

The potential beneficial outcomes of primary interest would be improvements in OS and disease-specific survival in individuals with pancreatic cancer.

Potential harmful outcomes are those resulting from false-positive or false-negative test results. False-positive test results can lead to unnecessary clinical management changes or unnecessary cascade testing for other cancers. False-negative test results can lead to the absence of clinical management changes.

Study Selection Criteria

For the evaluation of the clinical validity of the genetic test, studies that reported on the sensitivity and specificity and/or diagnostic yield of the test were considered, including curated sources of information on genes associated with increased risk of pancreatic cancer (eg, summaries from professional societies).

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Review of Evidence

Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with disease-associated variants; some recent studies are summarized in Table 8. A case-control analysis conducted by Hu et al (2018) compared the association of germline pathogenic variations in 3030 patients with pancreatic cancer to 176,241 controls from 2 public genome databases.13, There were significant associations between pancreatic cancer and pathogenic variations in 6 genes associated with pancreatic cancer (ATM, BRCA1, BRCA2, CDKN2A, MLH1, and TP53). Overall, pathogenic variants were identified in 5.5% of patients with pancreatic cancer.

Observational studies have reported that pathogenic variants are found in patients with pancreatic cancer who do not have a family history of the disease. In Hu et al (2018), pancreatic cancer associated variants were found in 7.9% of patients with a family history of pancreatic cancer and 5.2% of those without a family history of pancreatic cancer.13, Shindo et al (2017) reported that pathogenic variants were identified in 3.9% of a cohort of 854 patients with pancreatic adenocarcinoma.14, Of those with an identified pathogenic variant, only 3 (9.0%) reported a family history of pancreatic cancer.

Table 8. Study Characteristics: Clinical Validity of Genetic Tests in Patients with Pancreatic Cancer
Study Study Population Pathogenic Variants Identified, overall and by specific genes
Hu et al (2018)13, 3030 adults with pancreatic cancer enrolled in a registry
123,136 controls from the Genome Aggregation Database and 53,105 controls from the Exome Aggregation Consortium Database
Odds ratios (95% CI):
CDKN2A: 12.33 (5.43 to 25.61)
TP53: 6.70 (2.52 to 14.95)
MLH1: 6.66 (1.94 to 17.53)
BRCA2: 6.20 (4.62 to 8.17)
ATM: 5.71 (4.38 to 7.33)
BRCA1: 2.58 (1.54 to 4.05)
Brand et al (2018)15, 298 patients with newly diagnosed with pancreatic ductal adenocarcinoma 9.7%

Rate of pathogenic variants in specific genes:
ATM: 3.3%
BRCA1/2: 2.7%
CHEK2: 1.7%
Mandelker et al (2017)16, 1040 patients with advanced cancer (predominantly prostate, renal, pancreatic, breast and colon) referred for germline testing for hereditary cancer, who also had tumor DNA sequenced 44/176 (25%)

Pathogenic variants by gene
BRCA1: 6
BRCA2: 11
CDKN2A: 3
PALB2: 1
ATM: 5
CHEK2: 7
APC: 7
MUTYH: 3
FH (recessive): 1
Shindo et al (2017)14, 854 patients with pancreatic ductal adenocarcinoma;

Control groups: 288 patients with other pancreatic and periampullary neoplasms, and 51 patients with nonneoplastic diseases who underwent pancreatic resection
33/854 (3.9%; 95% CI, 3.0% to 5.8%)

Number of patients with deleterious variants in specific genes:
BRCA2: 12
ATM: 10
BRCA1 3
PALB2: 2
MLH1: 2
CDKN2A: 1
TP53: 1

3/33 patients had reported a family history of pancreatic cancer
Grant et al (2015)17, 708 individuals with pancreatic cancer consenting to be in a province-wide population-based registry, with available blood or saliva samples 11/290 (3.8%)

Number of pathogenic variants by gene:
ATM: 3
BRCA1: 1
BRCA2: 2
MLH1: 1
MSH2: 2
MSH6: 1
TP53: 1
 CI: confidence interval.

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

There are currently no targeted treatments for pancreatic cancer based on germline testing for ATM, CDKN2A , EPCAM, MLH1, MSH2, MSH6, PMS2, STK11, or TP53. It is unclear what management changes would be implemented based on results of such testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

There are no direct outcome data on the clinical usefulness of genetic testing for ATM, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PMS2, PALB, STK11, and TP53 (ie, no studies have reported outcomes data for patients tested and not tested).

Indirect Evidence

A chain of indirect evidence would demonstrate that genetic testing can identify individuals with pathogenic variants associated with pancreatic cancer who would not otherwise be identified, that treatments are available for these patients that would not otherwise be given to patients with pancreatic cancer, and that these treatments improve health outcomes.

Section Summary: Genetic Testing for ATM, CDKN2A , EPCAM, MLH1, MSH2, MSH6, PMS2, STK11, and TP53 in Individuals with Pancreatic Cancer

Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with disease-associated variants, including among those who do not have a family history of the disease. However, there is no direct evidence comparing health outcomes in patients tested or not tested for these variants. There are no targeted treatments for pancreatic cancer based on these variants.

For individuals with pancreatic cancer who receive genetic testing for ATM, CDK2NA, EPCAM, MMR genes (MLH1, MSH2, MSH6, PMS2), STK11, and TP53 to guide treatment, the evidence includes observational studies. Relevant outcomes are overall survival, disease-specific survival, test accuracy, and test validity. Multiple observational studies have demonstrated that testing patients with pancreatic cancer can identify individuals with disease-associated variants, including among those who do not have a family history of the disease. However, there is no direct evidence comparing health outcomes in patients tested or not tested for a variant. Additionally, there are no targeted treatments for pancreatic cancer based on these genes, and management changes that would result from testing these genes are unclear. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

 

Population

Reference No. 3

Policy Statement

[ ] MedicallyNecessary [X] Investigational

Population Reference No. 4

Genetic Testing in Asymptomatic Individuals who are at Risk for Hereditary Pancreatic Cancer

Clinical Context and Test Purpose

The purpose of genetic testing of asymptomatic individuals who are at high-risk for hereditary pancreatic cancer is to inform decisions about surveillance for early detection of pancreatic cancer. Given that most symptomatic pancreatic cancer is detected at an advanced stage and has a poor prognosis, targeted surveillance of high-risk individuals has the potential to identify tumors at an earlier stage that are more amenable to treatment.

The following PICO was used to select literature to inform this review.

Population

Individuals are considered at high-risk for hereditary pancreatic cancer if they have 2 relatives with pancreatic cancer where 1 is a first-degree relative, have 3 or more relatives with pancreatic cancer, or have a history of hereditary pancreatitis.

Interventions

The test being considered is testing for variants in genes associated with pancreatic cancer, including ATM, BRCA1, BRCA2, CDKN2A , EPCAM, MLH1, MSH2, MSH6, PALB2, STK11, and TP53.

For individuals without cancer who are at high-risk for hereditary pancreatic cancer, surveillance may be performed by endoscopic ultrasonography, magnetic resonance imaging (MRI), and/or computed tomography.

Comparators

Alternatives to genetic testing include risk assessment using criteria other than genetic testing (eg, family history).

Outcomes

The potential beneficial outcomes of primary interest would be improvements in OS and disease-specific survival.

Potential harmful outcomes are those resulting from false-positive or false-negative test results. False-positive test results can lead to unnecessary clinical management changes or unnecessary cascade testing for asymptomatic family members. False-negative test results can lead to the absence of clinical management changes or a lack of testing for asymptomatic family members.

Study Selection Criteria

For the evaluation of the clinical validity of the genetic test, studies that reported on the sensitivity and specificity and/or diagnostic yield of the test were considered, including curated sources of information on genes associated with increased risk of pancreatic cancer (eg, summaries from professional societies).

Clinically Valid

A test must detect the presence or absence of a condition, the risk of developing a condition in the future, or treatment response (beneficial or adverse).

Review of Evidence

Multiple genetic syndromes, including hereditary breast and ovarian cancer syndrome, are associated with an increased risk for pancreatic cancer (Table 9). Most of these are also associated with increased risk of other cancers. However, individual genes associated with the syndromes have been identified as increasing risk of pancreatic cancer, even in the absence of 1 of these syndromes.

Table 9. Pancreatic Cancer Susceptibility Genes and Associated Syndromes
Genes Associated Syndromes Absolute Risk of Pancreatic Cancer Relative Risk of Pancreatic Cancer Other Associated Cancers
ATM Ataxia-telangiectasia 1% to 5% 3-fold Breast, ovarian
BRCA1 Hereditary breast and ovarian 1.2% 3-fold Breast, ovarian, prostate
BRCA2 Hereditary breast and ovarian 2% to 5% 3.5 to 10-fold Breast, ovarian, prostate, melanoma
CDKN2A Familial atypical multiple mole melanoma 10% to 30% 13- to 39-fold Melanoma
MLH1, MSH2, MSH6, EPCAM Lynch 5% to 10% 9- to 11-fold Ovarian, colon, uterine, others
PALB2 Hereditary breast and ovarian 5% to 10% Unknown Breast, ovarian
PRSS1, SPINK1 Hereditary pancreatitis 40% to 45% 53-fold NA
STK11/LKB1 Peutz–Jeghers 10% to 30% Up to 132-fold Breast, ovarian, colorectal
Tp53 Li-Fraumeni Unknown Unknown Breast
 Sources: American Society of Clinical Oncology;2, American College of Gastroenterology7, NA: not available.

A prospective observational study of individuals under surveillance for pancreatic cancer on the basis of a family history of pancreatic cancer identified a known pathogenic variant in a pancreatic cancer susceptibility gene in 4.3% (15/345) (Table 10).18, In addition, 66 variants of unclear significance were identified. The cumulative incidence of pancreatic cancer in the germline variant group was higher than in the familial risk group, adjusted for age and sex and accounting for death as a competing event (HR, 2.85; 95% CI, 1.0 to 8.18; p=.05).

Table 10. Clinical Validity of Genetic Testing in Asymptomatic Individuals at High Risk for Hereditary Pancreatic Cancer
Study Study Population Prevalence of Pancreatic Cancer Pathogenic Variants Identified, overall and by specific genes
Abe et al (2019)18, 464 individuals enrolled in a high-risk pancreatic cancer surveillance program PDAC: 13/462 (2.8%)
PDAC or HGD:19/462 (4.1%)
PDAC or HGD or worrisome features on imaging: 42/446 (9.4%)
For patients with germline variants (n=134) compared to those with family history only with no known variant (n=330):
PDAC: HR, 2.85 (95% CI, 1 to 8.18, p=.05)
PDAC or HGD: HR, 2.81 (95% CI, 1.17 to 6.76, p=.02)
PDAC or HGD or worrisome features on imaging: HR, 3.27 (95% CI, 1.8 to 5.96, p<.001)
  CI: confidence interval; HGD: high-grade dysplasia; HR: hazard ratio; PDAC: pancreatic ductal adenocarcinoma.

Surveillance in Asymptomatic Individuals at High Risk for Hereditary Pancreatic Cancer

Recent prospective observational studies have reported the yield of screening and outcomes in high-risk individuals enrolled in pancreatic cancer surveillance programs (Table 11). Surveillance protocols varied somewhat and evolved over time, but typically included annual MRI and/or endoscopic ultrasound, with more frequent follow-up when a suspicious lesion was identified.

A 16-year follow-up study of surveillance in individuals at high-risk of pancreatic cancer due to family history or genetic factors was reported by Canto et al (2018).19, The overall detection rate over 16 years was 7%, including incident and prevalent neoplasms. Of 354 individuals under surveillance, 10 pancreatic cancers were detected, and 9 of 10 were resectable. Among these, 85% survived for 3 years.

Vasen et al (2016) found that surveillance of CDKN2A variant carriers detected most pancreatic adenocarcinomas at a resectable stage.20, In patients at risk for familial pancreatic cancer (those from families with 2 or 3 first-degree relatives with pancreatic cancer), however, the yield of screening was low.

Konings et al (2019) published a report of outcomes on 76 high-risk individuals from the International Cancer of the Pancreas Screening (CAPS) surveillance programs in 4 countries (U.S., the Netherlands, Israel, and Italy) who had either undergone pancreatic surgery because of the detection of a suspicious pancreatic lesion (n=71) or progressed to advanced unresectable malignant disease (n=5).21, Survival rate was significantly poorer for individuals with advanced pancreatic cancer compared with those who had surgery (40% vs. 83% respectively, p=.050; mean survival 9.5 vs. 54.3 months, p<.001).

Dbouk et al (2022) published results of the CAPS5 cohort, consisting of 1461 individuals who were determined to be at high risk for PDAC based either on presence of a germline pathogenic variant (48.5%) or family history without a known germline pathogenic variant (51.5%).22, A total of 9 individuals were diagnosed with a screen-detected pancreatic adenocarcinoma. The study authors concluded that their results "support current CAPS surveillance recommendations and argue against the notion of limiting pancreatic surveillance to those high-risk individuals with known pathogenic mutations."

In a cohort of 366 Dutch individuals at high risk of PDAC followed for 63 months (standard deviation, 43.2 months), Overbeek et al (2022) reported a 9.3% incidence of PDAC in the subset of individuals with a germline pathogenic variant and no PDAC in those with family history but no pathogenic variant.23, Three out of 10 (30%) individuals with PDAC were detected at an early stage. The resectability rate was 60% (6/10) overall and 50% (4/8) for incident cases.

Although these observational studies have demonstrated that surveillance can identify pancreatic cancer and precursor lesions in asymptomatic individuals, it is not possible to conclude from this body of evidence that surveillance improves survival. Longer survival time observed in individuals undergoing surveillance could be due to earlier identification of the disease (lead-time bias) and not the effects of early intervention and treatment.

Table 11. Studies of Surveillance in Individuals at High Risk of Pancreatic Cancer
Study Study Populations Surveillance Methods Results
Canto et al (2018) 19,(CAPS1, CAPS2, CAPS3, CAPS4) 354 individuals at high-risk for pancreatic cancer enrolled in Cancer of the Pancreas Screening cohort studies at tertiary care academic centers from 1998 through 2014
  • Patients who met clinical criteria for Peutz-Jeghers syndrome, or who had a variant in the STK11 gene, at least 30 yrs old
  • Individuals from an FPC kindred (at least 1 FDR with pancreatic cancer, at least 50 yrs old (CAPS 1 to 3) or at least 55 yrs old (CAPS 4), or 10 yrs younger than youngest pancreatic cancer in the family
  • Individuals with confirmed germline variants in BRCA1, BRCA2, PALB2.PRSS1, CDKN2A, or MLH1, MSH2, MSH6, PMS2 (Lynch syndrome), with at least 1 affected first- or second-degree relative, and at least 50 yrs old, or 10 yrs younger than the youngest pancreatic cancer in the family
EUS, MRI, and/or CT baseline screening with EUS intervals depended on the presence or absence of neoplastic-type pancreatic lesions. Normal pancreas or EUS features of chronic pancreatitis were followed annually. Those with pancreatic cysts or indeterminate radiologic lesions underwent more frequent imaging with EUS and/or MRI or CT, according to published international guidelines: every 6 to 12 months for those without a mural nodule or dilated pancreatic duct and every 3 to 6 months for larger cysts or cysts with worrisome features. Stable or improved appearance of pancreatic lesions resulted in decreased surveillance imaging frequency to every 12 mos. Median follow-up 5.6 yrs. Overall detection rate over 16 yrs was 7%; 9/10 cancers detected were resectable.
Dbouk et al (2022)22,
CAPS5
NCT02000089
1461 individuals with estimated elevated risk of developing PDAC.

Eligibility criteria:
Hereditary syndromes or germline variant carriers (BRCA2, ATM, BRCA1, PALB2, or Lynch syndrome–associated genes with family history of PDAC, FAMMM [CDKN2A], Peutz-Jeghers [STK11]). Family history of at least 1 first-degree and 1 second-degree relative with PDAC. Met age criteria for surveillance. Participants:
48.5% had a pathogenic germline variant; 51.5% had family history without known pathogenic germline variant; 31.1% had a personal history of cancer
Annual surveillance with EUS and/or MRI/MRCP, often alternating between the 2 methods (surveillance interval was modified when concerning lesions were detected) 9 patients were diagnosed with a screen-detected PDAC (either at baseline or at subsequent surveillance visits).

1 additional patient presented with symptomatic metastatic PDAC 4 years after their baseline and only surveillance.

7/9, (77.8%) were stage I by surgical pathology (4 stage IA, 3 stage IB); 1 patient had stage IIB cancer (case 8), and one (case 9) had a stage III cancer (clinically staged) with superior mesenteric artery involvement.

Overall, 8/9 (88.9%) of the screen-detected PDACs were resectable. Two of the stage I PDACs were surgically staged after neoadjuvant chemotherapy
(their stages at diagnosis by imaging, before neoadjuvant therapy were stage IA and IIA).

Among 7 of 9 patients diagnosed with a screen-detected PDAC still alive, median overall survival is 3.84 years.
Overbeek et al 202223, 366 asymptomatic individuals with an estimated 10% or greater lifetime risk of PDAC
201 with family history and no known germline pathogenic variant, 165 with a PDAC susceptibility gene
Annual surveillance with both EUS and MRI/MRCP at each visit, surveillance after 3 or 6 months when a concerning lesion detected. 9.3% in PDAC susceptibility gene carriers (cumulative incidence 6.5% at 5 years and 9.3% at 10 years). No cases identified in germline pathogenic variant-negative FPC kindreds. Median survival was 18 months (range, 1 to 32).
Vasen et al (2016)20,
  • 178 individuals with a CDKN2A variant
  • 214 individuals at high-risk for familial pancreatic cancer (from families with 2 or 3 first-degree relatives with pancreatic cancer)
  • 19 individuals with a BRCA1/2 or PALB2 variant
Annual MRI. Beginning in 2012, endoscopic ultrasound was also offered as an option in addition to annual MRI. In the event of a small lesion, MRI was repeated 3 to 6 months later. In cases where there was serious suspicion of pancreatic adenocarcinoma, additional endoscopic ultrasound and CT scanning was performed. Individuals with a CDKN2A variant:
  • 13/178 (7.3%)
  • Cumulative incidence of pancreatic cancer was 14% by the age of 70 yrs
Individuals at high-risk for familial pancreatic cancer
  • 3/214 (1.4%)
Individuals with a BRCA1/2 or PALB2 variant
  • 1/19 (3.8%)
 CAPS: Cancer of the Pancreas Screening; CT: computed tomography; EUS: endoscopic ultrasound; FAMMM: familial atypical multiple mole melanoma; FDR: first-degree relative; FPC: familial pancreatic cancer; MRCP, magnetic resonance cholangiopancreatography; MRI: magnetic resonance imaging; PDAC: pancreatic ductal adenocarcinoma.

Screening and Surveillance for Other Cancers in Asymptomatic Patients at High-Risk for Hereditary Pancreatic Cancer

Genes that are associated with pancreatic cancer are also associated with increased risk of other cancers and genetic cancer syndromes (see Table 9). For this reason, genetic testing in patients with pancreatic cancer has been proposed to identify patients who are candidates for surveillance, early treatment, and prevention of cancers such as breast, ovarian, colon, and melanoma. A review of the evidence in other cancers is beyond the scope of this review, and is addressed in the following policies:

Clinically Useful

A test is clinically useful if the use of the results informs management decisions that improve the net health outcome of care. The net health outcome can be improved if patients receive correct therapy, or more effective therapy, or avoid unnecessary therapy, or avoid unnecessary testing.

Direct Evidence

Direct evidence of clinical utility is provided by studies that have compared health outcomes for patients managed with and without the test. Because these are intervention studies, the preferred evidence would be from RCTs.

There is no direct evidence comparing health outcomes in asymptomatic patients tested or not tested for genes associated with hereditary pancreatic cancer.

Chain of Evidence

A chain of indirect evidence would demonstrate that genetic testing can identify individuals with pathogenic variants associated with hereditary pancreatic cancer who would not otherwise be identified, that treatments or increased surveillance are available for these patients that would not otherwise be given to patients with hereditary pancreatic cancer, and that these interventions improve health outcomes.

Section Summary: Genetic Testing in Asymptomatic Individuals who are at Risk for Hereditary Pancreatic Cancer

There is no direct evidence comparing health outcomes in patients tested or not tested for a variant. There is indirect evidence from 2 comparative observational studies of high-risk individuals under surveillance that the risk of progression to pancreatic cancer is higher among individuals with a known pathogenic variant than in individuals identified as at-risk based on family history alone. There is also evidence from prospective observational studies that surveillance of high-risk individuals can identify pancreatic cancer and precursor lesions. In 1 analysis of 76 high-risk individuals under surveillance, survival was better in those who had surgery due to detection of either low- or high-risk neoplastic precursor lesions (n=71) compared to those who had advanced to unresectable disease (n=5). Although observational studies have demonstrated that surveillance can identify pancreatic cancer and precursor lesions in asymptomatic individuals, it is not possible to conclude from this body of evidence that surveillance improves survival. Longer survival time observed in individuals undergoing surveillance could be due to earlier identification of the disease (lead-time bias) and not the effects of early intervention and treatment. Additionally, evidence is too limited to determine if selecting patients for surveillance based on genetic testing leads to better outcomes than using criteria such as family history alone.

For individuals who are asymptomatic and at high risk for hereditary pancreatic cancer who receive testing for genes associated with hereditary pancreatic cancer, the evidence includes observational studies. There is no direct evidence comparing health outcomes in patients tested or not tested for a variant. There is indirect evidence from 2 comparative observational studies of high-risk individuals under surveillance that the risk of progression to pancreatic cancer is higher among individuals with a known pathogenic variant than in individuals identified as at-risk based on family history alone. There is also evidence from prospective observational studies that surveillance of high-risk individuals can identify pancreatic cancer and precursor lesions. In 1 analysis of 76 high-risk individuals under surveillance, survival was better in those who had surgery due to detection of either low- or high-risk neoplastic precursor lesions (n=71) compared to those who had advanced to unresectable disease (n=5). Although observational studies have demonstrated that surveillance can identify pancreatic cancer and precursor lesions in asymptomatic individuals, it is not possible to conclude from this body of evidence that surveillance improves survival. Longer survival time observed in individuals undergoing surveillance could be due to earlier identification of the disease (downstaging) and not the effects of early intervention and treatment. Additionally, evidence is too limited to determine if selecting patients for surveillance based on genetic testing leads to better outcomes than using criteria such as family history alone. The evidence is insufficient to determine that the technology results in an improvement in the net health outcome.

Population

Reference No. 4

Policy Statement

[ ] MedicallyNecessary [X] Investigational

Supplemental Information

The purpose of the following information is to provide reference material. Inclusion does not imply endorsement or alignment with the evidence review conclusions.

Practice Guidelines and Position Statements

Guidelines or position statements will be considered for inclusion in ‘Supplemental Information’ if they were issued by, or jointly by, a US professional society, an international society with US representation, or National Institute for Health and Care Excellence (NICE). Priority will be given to guidelines that are informed by a systematic review, include strength of evidence ratings, and include a description of management of conflict of interest.

American College of Gastroenterology

In 2015, the American College of Gastroenterology Clinical Guideline on Genetic Testing and Management of Hereditary Gastrointestinal Cancer Syndromes includes the following recommendations on genetic testing for pancreatic cancer:7,

American Society of Clinical Oncology

In 2019, an American Society of Clinical Oncology (ASCO) opinion statement addressed the identification and management of patients and family members with a possible predisposition to pancreatic adenocarcinoma and made the following recommendations:2,

In 2020, ASCO published a guideline update on recommendations for second-line therapy options for metastatic pancreatic cancer.24, In patients who have a germline BRCA1 or BRCA2 mutation and who have received first-line platinum based chemotherapy without disease progression for at least 16 weeks, options for continued treatment include chemotherapy or the poly ADP ribose polymerase (PARP) inhibitor olaparib.

International Cancer of the Pancreas Screening Consortium

In 2020, the International Cancer of the Pancreas Screening Consortium published an updated consensus document on the management of patients with increased risk for familial pancreatic cancer.25, The panel recommended pancreatic cancer surveillance performed in a research setting for the following individuals:

The preferred surveillance tests are endoscopic ultrasound and magnetic resonance imaging (MRI). The recommended age to initiate surveillance depends on an individual's gene mutation status and family history, but no earlier than age 50 or 10 years earlier than the youngest relative with pancreatic cancer. There was no consensus on the age to end surveillance.

National Comprehensive Cancer Network

Two National Comprehensive Cancer Network (NCCN) guidelines address germline genetic testing in individuals with or at high risk for pancreatic cancer.26,6,

The Guidelines on Genetic/Familial High-risk Assessment: Breast, Ovarian, and Pancreatic ( v.2.2024v.2.2025) recommend germline testing for all individuals with exocrine pancreatic cancer, and specify that testing of first-degree relatives should only be done only if it is impossible to test the individual who has pancreatic cancer.26,

The Guideline on Treatment of Pancreatic Adenocarcinoma ( v.1.2024v.1.2025) recommends germline testing for any patient with confirmed pancreatic cancer using comprehensive gene panels for hereditary cancer syndromes.6, The guideline specifies the following genes as those typically tested for pancreatic cancer risk: ATM, BRCA1, BRCA2, CDKN2A, MLH1, MSH2, MSH6, ), PALB2, PMS2, STK11, and TP53.

For patients with locally advanced disease, preferred first-line therapy regimens include gemcitabine + cisplatin for patients with BRCA1/2 or PALB2 variants For patients with metastatic disease who have received previous platinum-based chemotherapy, olaparib is preferred only for patients with germline BRCA1/2 variants.

Genetic counseling is recommended for patients who test positive for a pathogenic variant, or for patients with a positive family history of pancreatic cancer, regardless of test results. The guidelines also recommend genetic counseling for patients who test positive for a pathogenic variant or for patients with a positive family history of pancreatic cancer, regardless of variant status.

U.S. Preventive Services Task Force Recommendation

The 2019 U.S. Preventive Services Task Force recommendation on screening for pancreatic cancer applies to asymptomatic adults not known to be at high-risk of pancreatic cancer.5, The recommendation does not apply to persons at high-risk of pancreatic cancer due to an inherited genetic syndrome or due to a history of hereditary pancreatic cancer.

Medicare National Coverage

There is no national coverage determination. In the absence of a national coverage determination, coverage decisions are left to the discretion of local Medicare carriers.

Ongoing and Unpublished Clinical Trials

Some currently unpublished trials that might influence this review are listed in Table 12.

Table 12. Summary of Key Trials
NCT No. Trial Name Planned Enrollment Completion Date
Ongoing      
NCT06760741 PREVENPANC Project: a Spanish Multicenter Study for Pancreatic Cancer Prevention 900 Dec 2026
NCT03060720 Systematic Hereditary Pancreatic Cancer Risk Assessment and Implications for Personalized Therapy 271 Feb 2026
NCT00835133 Biospecimen Resource for Familial Pancreas Research, a Data and Tissue Registry (Also Known as a Bio-repository, Bio-bank, Data and Tissue Database, Data and Tissue Bank, Etc.) to Help Advance Research in Familial Pancreas Disease 1122 (actual) Oct 2025
NCT02206360 Observational Study to Analyze the Outcomes of Subjects Who - Based Upon Their Sufficiently Elevated Risk for the Development of Pancreatic Adenocarcinoma- Elect to Undergo Early Detection Testing 100 Jun 2024
NCT00526578 Pancreatic Cancer Genetic Epidemiology (PACGENE) Study 4,770 Jun 2025
NCT05287347 Prospective Multicenter Observational Study for Validation of a Pancreatic Cancer Risk Model and Assessment of the Predictive Value of Blood Biomarkers in a High-risk Cohort 4,000 Oct 2027
NCT04548752 Randomized Phase II Clinical Trial of Olaparib + Pembrolizumab vs. Olaparib Alone as Maintenance Therapy in Metastatic Pancreatic Cancer Patients With Germline BRCA1 or BRCA2 Mutations 88 Mar 2025
 NCT: national clinical trial.

References

  1. National Cancer Institute: Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Pancreatic Cancer. https://seer.cancer.gov/statfacts/html/pancreas.html. Accessed January 8, 2025.
  2. Stoffel EM, McKernin SE, Khorana AA. Evaluating Susceptibility to Pancreatic Cancer: ASCO Clinical Practice Provisional Clinical Opinion Summary. J Oncol Pract. Feb 2019; 15(2): 108-111. PMID 30589608
  3. O'Reilly EM, Lee JW, Zalupski M, et al. Randomized, Multicenter, Phase II Trial of Gemcitabine and Cisplatin With or Without Veliparib in Patients With Pancreas Adenocarcinoma and a Germline BRCA/PALB2 Mutation. J Clin Oncol. May 01 2020; 38(13): 1378-1388. PMID 31976786
  4. Reiss KA, Yu S, Judy R, et al. Retrospective Survival Analysis of Patients With Advanced Pancreatic Ductal Adenocarcinoma and Germline BRCA or PALB2 Mutations. JCO Precision Oncology. Published online January 19, 2018. DOI: 10.1200/PO.17.00152.
  5. Owens DK, Davidson KW, Krist AH, et al. Screening for Pancreatic Cancer: US Preventive Services Task Force Reaffirmation Recommendation Statement. JAMA. Aug 06 2019; 322(5): 438-444. PMID 31386141
  6. National Comprehensive Cancer Network (NCCN). Clinical Practice Guidelines in Oncology: Pancreatic Adenocarcinoma. Version 1.2025. https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf. Accessed December 21, 2024.
  7. Syngal S, Brand RE, Church JM, et al. ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes. Am J Gastroenterol. Feb 2015; 110(2): 223-62; quiz 263. PMID 25645574
  8. Food & Drug Administration. Premarket Approval: BRACAnalysis CDx. 2019. https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id=P140020S019. Accessed January 8, 2025.
  9. Golan T, Kanji ZS, Epelbaum R, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer. Sep 09 2014; 111(6): 1132-8. PMID 25072261
  10. Wattenberg MM, Asch D, Yu S, et al. Platinum response characteristics of patients with pancreatic ductal adenocarcinoma and a germline BRCA1, BRCA2 or PALB2 mutation. Br J Cancer. Feb 2020; 122(3): 333-339. PMID 31787751
  11. Yu S, Agarwal P, Mamtani R, et al. Retrospective survival analysis of patients with resected pancreatic ductal adenocarcinoma and a Germline BRCA or PALB2 mutation. JCO Precision Oncol. Published online March 28, 2019. DOI: 10.1200/PO.18.00271
  12. Golan T, Hammel P, Reni M, et al. Maintenance Olaparib for Germline BRCA -Mutated Metastatic Pancreatic Cancer. N Engl J Med. Jul 25 2019; 381(4): 317-327. PMID 31157963
  13. Hu C, Hart SN, Polley EC, et al. Association Between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. JAMA. Jun 19 2018; 319(23): 2401-2409. PMID 29922827
  14. Shindo K, Yu J, Suenaga M, et al. Deleterious Germline Mutations in Patients With Apparently Sporadic Pancreatic Adenocarcinoma. J Clin Oncol. Oct 20 2017; 35(30): 3382-3390. PMID 28767289
  15. Brand R, Borazanci E, Speare V, et al. Prospective study of germline genetic testing in incident cases of pancreatic adenocarcinoma. Cancer. Sep 01 2018; 124(17): 3520-3527. PMID 30067863
  16. Mandelker D, Zhang L, Kemel Y, et al. Mutation Detection in Patients With Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing. JAMA. Sep 05 2017; 318(9): 825-835. PMID 28873162
  17. Grant RC, Selander I, Connor AA, et al. Prevalence of germline mutations in cancer predisposition genes in patients with pancreatic cancer. Gastroenterology. Mar 2015; 148(3): 556-64. PMID 25479140
  18. Abe T, Blackford AL, Tamura K, et al. Deleterious Germline Mutations Are a Risk Factor for Neoplastic Progression Among High-Risk Individuals Undergoing Pancreatic Surveillance. J Clin Oncol. May 01 2019; 37(13): 1070-1080. PMID 30883245
  19. Canto MI, Almario JA, Schulick RD, et al. Risk of Neoplastic Progression in Individuals at High Risk for Pancreatic Cancer Undergoing Long-term Surveillance. Gastroenterology. Sep 2018; 155(3): 740-751.e2. PMID 29803839
  20. Vasen H, Ibrahim I, Ponce CG, et al. Benefit of Surveillance for Pancreatic Cancer in High-Risk Individuals: Outcome of Long-Term Prospective Follow-Up Studies From Three European Expert Centers. J Clin Oncol. Jun 10 2016; 34(17): 2010-9. PMID 27114589
  21. Konings ICAW, Canto MI, Almario JA, et al. Surveillance for pancreatic cancer in high-risk individuals. BJS Open. Oct 2019; 3(5): 656-665. PMID 31592073
  22. Dbouk M, Katona BW, Brand RE, et al. The Multicenter Cancer of Pancreas Screening Study: Impact on Stage and Survival. J Clin Oncol. Oct 01 2022; 40(28): 3257-3266. PMID 35704792
  23. Overbeek KA, Levink IJM, Koopmann BDM, et al. Long-term yield of pancreatic cancer surveillance in high-risk individuals. Gut. Jun 2022; 71(6): 1152-1160. PMID 33820756
  24. Sohal DPS, Kennedy EB, Cinar P, et al. Metastatic Pancreatic Cancer: ASCO Guideline Update. J Clin Oncol. Sep 20 2020; 38(27): 3217-3230. PMID 32755482
  25. Goggins M, Overbeek KA, Brand R, et al. Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the International Cancer of the Pancreas Screening (CAPS) Consortium. Gut. Jan 2020; 69(1): 7-17. PMID 31672839
  26. National Comprehensive Cancer Network (NCCN). Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2025. https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf. Accessed December 22, 2024.

Codes

Codes Number Description
    There is no test specifically for pancreatic cancer. Genes associated with pancreatic cancer include BRCA1, BRCA2, PALB2, ATM, APC, MLH1, MLH2, MSH6, PMS2, EPCAM, CDKN2A, TP53, STK11. Tests or panels that include these genes may be reported and are listed below
CPT 0129U Hereditary breast cancer–related disorders (eg, hereditary breast cancer, hereditary ovarian cancer, hereditary endometrial cancer), genomic sequence analysis and deletion/duplication analysis panel (ATM, BRCA1, BRCA2, CDH1, CHEK2, PALB2, PTEN, and TP53)
  81162 BRCA1 (BRCA1, DNA repair associated), BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis and full duplication/deletion analysis (ie, detection of large gene rearrangements)
  81163 BRCA1 (BRCA1, DNA repair associated), BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis
  81164 BRCA1 (BRCA1, DNA repair associated), BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full duplication/deletion analysis (ie, detection of large gene rearrangements)
  81165 BRCA1 (BRCA1, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis
  81166 BRCA1 (BRCA1, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full duplication/deletion analysis (ie, detection of large gene rearrangements
  81167 BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full duplication/deletion analysis (ie, detection of large gene rearrangements)
  81201 APC (adenomatous polyposis coli) (eg, familial adenomatosis polyposis [FAP], attenuated FAP) gene analysis; full gene sequence
  81212 BRCA1 (BRCA1, DNA repair associated), BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; 185delAG, 5385insC, 6174delT variants
  81215 BRCA1 (BRCA1, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; known familial variant
  81216 BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; full sequence analysis
  81217 BRCA2 (BRCA2, DNA repair associated) (eg, hereditary breast and ovarian cancer) gene analysis; known familial variant
  81288 MLH1 (mutL homolog 1, colon cancer, nonpolyposis type 2) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; promoter methylation analysis
  81292 MLH1 (mutL homolog 1, colon cancer, nonpolyposis type 2) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis
  81293 MLH1 (mutL homolog 1, colon cancer, nonpolyposis type 2) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; known familial variants
  81294 MLH1 (mutL homolog 1, colon cancer, nonpolyposis type 2) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; duplication/deletion variants
  81295 MSH2 (mutS homolog 2, colon cancer, nonpolyposis type 1) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis
  81298 MSH6 (mutS homolog 6 [E. coli]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis
  81299 MSH6 (mutS homolog 6 [E. coli]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; known familial variants
  81300 MSH6 (mutS homolog 6 [E. coli]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; duplication/deletion variants
  81317 PMS2 (postmeiotic segregation increased 2 [S. cerevisiae]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; full sequence analysis
  81318 PMS2 (postmeiotic segregation increased 2 [S. cerevisiae]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; known familial variants
  81319 PMS2 (postmeiotic segregation increased 2 [S. cerevisiae]) (eg, hereditary non-polyposis colorectal cancer, Lynch syndrome) gene analysis; duplication/deletion variants
  81403 Molecular pathology procedure, Level 4
  81404 Molecular pathology procedure, Level 5
  81405 Molecular pathology procedure, Level 6
  81406 Molecular pathology procedure, Level 7
  81432 Hereditary breast cancer-related disorders (eg, hereditary breast cancer, hereditary ovarian cancer, hereditary endometrial cancer, hereditary pancreatic cancer, hereditary  prostate cancer); genomic sequence analysis panel, 5 or more genes, interrogation for sequence variants and copy number variants.
  81435 Hereditary colon cancer disorders (eg, Lynch syndrome, PTEN hamartoma syndrome, PTEN hamartoma syndrome, Cowden syndrome, familial adenomatosis polyposis), genomic sequence analysis panel, 5 or more genes, interrogation for sequence variants and copy number variants.
  81445 Targeted genomic sequence analysis panel, solid organ neoplasm, DNA analysis, and RNA analysis when performed, 5-50 genes (eg, ALK, BRAF, CDKN2A, EGFR, ERBB2, KIT, KRAS, NRAS, MET, PDGFRA, PDGFRB, PGR, PIK3CA, PTEN, RET), interrogation for sequence variants and copy number variants or rearrangements, if performed
  81455 Targeted genomic sequence analysis panel, solid organ or hematolymphoid neoplasm, DNA analysis, and RNA analysis when performed, 51 or greater genes (eg, ALK, BRAF, CDKN2A, CEBPA, DNMT3A, EGFR, ERBB2, EZH2, FLT3, IDH1, IDH2, JAK2, KIT, KRAS, MLL, NPM1, NRAS, MET, NOTCH1, PDGFRA, PDGFRB, PGR, PIK3CA, PTEN, RET), interrogation for sequence variants and copy number variants or rearrangements, if performed
HCPCS No code  
ICD10-CM C25.0-C25.9 Malignant neoplasm of the pancreas code range
  D01.7 Carcinoma in situ of other specified digestive organs (pancreas)
  Z12.89 Encounter for screening for malignant neoplasm of other sites
  Z15.09 Genetic susceptibility to other malignant neoplasm
  Z80.0 Family history of malignant neoplasm of digestive organs
  Z85.07 Personal history of malignant neoplasm of pancreas
PCS   Not applicable. ICD-10-PCS codes are only used for inpatient services. There are no ICD procedure codes for laboratory tests.
TOS Laboratory  
POS Outpatient  

Applicable Modifiers

N/A

Policy History

Date Action Description
03/03/2025 Annual Review

Policy updated with literature review through December 23, 2024; no references added. Policy statements unchanged.

12/16/2024 Code review

Code 81432, 81435 contains revised text. Delete code effective 1/01/2025 81433, 81436.

03/11/2024 Annual Review Policy updated with literature review through January 3, 2024; no references added. Policy statements unchanged.
03/15/2023 Annual Review Policy updated with literature review through January 6, 2023; references added. Minor editorial refinements to policy statements; intent unchanged. Policy title changed to include gene names (ATM, BRCA1, BRCA2, CDKN2A, EPCAM, MLH1, MSH2, MSH6, PALB2, PMS2, STK11, and TP53).
03/09/2022 Annual Review Policy updated with literature review through January 3, 2022; no references added. Policy statements unchanged.
03/04/2021 Policy Reviewed Policy updated with literature review through December 16, 2020; references added. New indication and medically necessary statement added for BRCA1, BRCA2, and PALB2 variant testing to select first-line treatment with platinum chemotherapy. PALB2 testing removed from indication 3. Indication 4 (genetic testing in asymptomatic individuals) unchanged. Title changed to "Germline Genetic Testing for Pancreatic Cancer Susceptibility Genes."
11/13/2020 Policy Reviewed Policy Statement Unchanged
10/27/2020 Policy Created New Policy