E-Mail Us Close
Please note that this email should only be used for feedback and comments specifically related to this particular medical policy.
  
Horizon BCBSNJ
Uniform Medical Policy ManualSection:Radiology
Policy Number:166
Effective Date: 03/10/2020
Original Policy Date:02/23/2016
Last Review Date:02/11/2020
Date Published to Web: 04/13/2016
Subject:
Pediatric Oncology Imaging Policy

Description:
_______________________________________________________________________________________

IMPORTANT NOTE:
Medical Policies have been migrated to the new medical policy platform, the EMM (Evidence Management Module) and the new Horizonblue.com Medical Policy web page, effective (11/23/20). Medical Policies will no longer be updated on the Lotus Notes Horizon Medical Policy Database but current content will remain accessible to current readers.

For a new medical policy or the most current version of a medical policy, please copy and paste this link horizonblue.com/medicalpolicies to your browser or go to horizonblue.com > Providers > Policies & Procedures > Policies > Medical Policies.

Employees who need access to previous versions of policies may continue to request a copy of the previous version by emailing the Medical Policy Team at: MedPol_Grp_@horizonblue.com

All other sections of this Lotus Notes Medical Policy Database, such as the System Directive Sheet (Codes), PQR Summary and Integration BRD, will continue to be maintained on this database until further notice.

__________________________________________________________________________________________________________________________

Table of Contents

Abbreviations for Pediatric Oncology Imaging Guidelines
PEDONC-1: General Guidelines
PEDONC-2: Screening Imaging in Cancer Predisposition Syndromes
PEDONC-3: Pediatric Leukemias
PEDONC-4: Pediatric CNS Tumors
PEDONC-5: Pediatric Lymphomas
PEDONC-6: Neuroblastoma
PEDONC-7: Pediatric Renal Tumors
PEDONC-8: Pediatric Soft Tissue Sarcomas
PEDONC-9: Bone Tumors
PEDONC-10: Pediatric Germ Cell Tumors
PEDONC-11: Pediatric Liver Tumors
PEDONC-12: Retinoblastoma
PEDONC-13: Pediatric Nasopharyngeal Carcinoma
PEDONC-14: Pediatric Adrenocortical Carcinoma
PEDONC-15: Pediatric Melanoma and Other Skin Cancers
PEDONC-16: Pediatric Salivary Gland Tumors
PEDONC-17: Pediatric Breast Masses
PEDONC-18: Histiocytic Disorders
PEDONC-19: Long Term Pediatric Cancer Survivors

ABBREVIATIONS for PEDIATRIC ONCOLOGY IMAGING POLICY

AFPAlpha-fetoprotein (tumor marker)
ALCLAnaplastic large cell lymphoma
ALLAcute lymphoblastic leukemia
AMLAcute myelogenous leukemia
β-hCGHuman chorionic gonadotropin beta-subunit (tumor marker)
BKLBurkitt’s lymphoma
BWTBilateral Wilms tumor
CCSKClear cell sarcoma of the kidney
CNSCentral nervous system
COGChildren’s Oncology group
CPT®Current procedural terminology; trademark of the American Medical Association
CSFCerebrospinal fluid
CTComputed tomography
CXRChest x-ray
DAWTDiffuse anaplasia Wilms tumor
ESFTEwing sarcoma family of tumors
FAWTFocal anaplasia Wilms tumor
FHWTFavorable histology Wilms tumor
HLHodgkin lymphoma
HSCTHematopoietic stem cell transplant (bone marrow or peripheral blood)
HVAHomovanillic acid
LLLymphoblastic lymphoma
MIBGMetaiodobenzylguanidine (nuclear scan using 123i or 131i)
MPNSTMalignant peripheral nerve sheath tumor
MRI Magnetic resonance imaging
NBLNeuroblastoma
NEDNo evidence of disease
NHLNon-hodgkin lymphoma
NPCNasopharyngeal carcinoma
NRSTSNonrhabdomyosarcomatous soft tissue sarcomas
OSOsteosarcoma
PETPositron emission tomography
PMBCLPrimary mediastinal b-cell lymphoma
PNETPrimitive neuroectodermal tumor
RCCRenal cell carcinoma
RMSRhabdomyosarcoma
USUltrasound
VMAVannilylmandelic acid
WBCWhite blood cell count
XRTRadiation therapy


Policy:
( NOTE: For Medicare Advantage, Medicaid and FIDE-SNP, please refer to the Coverage Sections below for coverage guidance.)

PEDONC-1: General Guidelines

PEDONC-1.1: Age Considerations
PEDONC-1.2: Appropriate Clinical Evaluations
PEDONC-1.3: Modality General Considerations
PEDONC-1.4: PET Imaging in Pediatric Oncology
PEDONC-1.5: Diagnostic Radiation Exposure in Pediatric Oncology

This General Policy section provides an overview of the basic criteria for which pediatric oncology imaging may be medically necessary. Details regarding specific conditions or clinical presentations and the associated criteria for which imaging is medically necessary are described in subsequent sections.

This General Policy section provides an overview of the basic criteria for which Pediatric Oncology imaging may be medically necessary. Details regarding specific conditions or clinical presentations and the associated criteria for which imaging is medically necessary are described in subsequent sections.

PEDONC-1.1: Age Considerations

For this condition imaging is medically necessary based on the following criteria:

The majority of malignancies occurring in the pediatric population are different diagnoses than those occurring in the adult population. For those diseases which occur in both pediatric and adult populations, minor differences may exist in management between pediatric and adult medical oncologists due to member age, comorbidities, and differences in disease natural history between children and adults.

Members who are < 18 years old at initial diagnosis should be imaged according to the Pediatric Oncology Imaging Guidelines, and members who are ≥ 18 years at initial diagnosis should be imaged according to the adult Oncology Imaging Guidelines, except where directed otherwise by a specific guideline section.

Members who are 15 to 39 years old at initial diagnosis are defined as Adolescent and Young Adult (AYA) Oncology members. There is significantly more overlap between cancer types in this age group.

    ® When unique guidelines for a specific cancer type exist only in either Oncology or Pediatric Oncology, AYA members should be imaged according to the guideline section for their specific cancer type, regardless of the member’s age.
PEDONC-1.2: Appropriate Clinical Evaluations

For this condition imaging is medically necessary based on the following criteria:

In general, a recent (within 60 days) detailed history and physical examination and appropriate laboratory studies should be performed prior to considering advanced imaging, unless the member is undergoing guideline-supported scheduled off-therapy surveillance evaluation.

    ® Because of the relatively small number of childhood cancer treatment centers, it is common to combine off-therapy visits with imaging and other subspecialist visits to accommodate families traveling long distances for their child’s care.

The majority of pediatric oncology imaging indications are listed in the diagnosis-specific guideline sections, but for rare malignancies and other circumstances not specifically addressed elsewhere in the pediatric oncology guidelines, the following general principles apply:
    ® Routine imaging of brain, spine, neck, chest, abdomen, pelvis, bones, or other body areas is not indicated in the absence of localizing symptoms or abnormalities on plain radiography or ultrasound.

The overwhelming majority of pediatric oncology members treated in the United States will be enrolled on or treated according to recent Children’s Oncology Group (COG) protocols. These imaging guidelines are consistent with evaluations recommended by COG protocols commonly used for direct member care (whether formally enrolled on study or not).

For members enrolled on a COG study, imaging recommended by COG protocols should generally be approved unless the imaging is being performed solely to address a study objective and would not be indicated in usual clinical care.

Phases of Pediatric Oncology Imaging:

Screening:

    ® All imaging studies requested for members at increased risk for a particular cancer in the absence of any clinical signs or symptoms.
    ® Screening using advanced imaging is only supported for conditions listed in PEDONC-2: Screening Imaging In Cancer Predisposition Syndromes.

Initial staging:
    ® All imaging studies requested from the time cancer is first clinically suspected until the initiation of specific treatment (which may be surgical resection alone)
    ® Pediatric malignancies in general behave more aggressively than adult cancers, and the time from initial suspicion of cancer to specific therapy initiation can be measured in hours to days for most pediatric cancers
      ¡ It is recommended that children with pediatric solid tumors undergo CT evaluation of the Chest prior to general anesthesia for biopsy or resection due to the risk of post-operative atelectasis mimicking pulmonary metastasis resulting in inaccurate staging and/or delay in therapy initiation
      ¡ If CTs of other body areas are indicated, (Neck, Abdomen, Pelvis), they should be performed concurrently with Chest CT to avoid overlapping fields and the resulting increase in radiation exposure
      ¡ Metastatic CNS imaging and nuclear medicine imaging are generally deferred until after a histologic diagnosis is made, with the exception of aggressive non-Hodgkin Lymphomas
Treatment response:
    ® All imaging studies completed during any type of active treatment (chemotherapy or other medications, radiation therapy, or surgery), including evaluation at the end of planned active treatment
    ® Unless otherwise stated in the diagnosis-specific guidelines, imaging for treatment response can be approved after every 2 cycles, which is usually ~6 weeks of therapy for solid tumors and usually ~8 to 12 weeks for CNS tumors

Surveillance:
    ® All routine imaging studies requested for a member who is not receiving any active treatment, even if residual imaging abnormalities are present
    ® Unlike adult cancers, in most pediatric cancers surveillance does not begin until all planned multimodal therapy is completed. Pediatric cancers where surgical resection is considered curative are listed in the diagnosis-specific guideline sections
    ® The recommended timing for surveillance imaging studies in these guidelines refers to members who are asymptomatic or have stable chronic symptoms
    ® Certain tumor types do not require surveillance with advanced imaging as member outcomes following relapse are not improved by surveillance imaging. See diagnosis-specific guideline sections for details.
    ® PET imaging is not supported for surveillance imaging unless specifically stated in elsewhere in the diagnosis-specific guideline sections
    ® Members with new or changing clinical signs or symptoms suggesting recurrent disease should have symptom-appropriate imaging requests approved even when surveillance timing recommendations are not met.

Recurrence:
    ® All imaging studies completed at the time a recurrence or progression of a known cancer is documented or is strongly suspected based on clinical signs or symptoms, laboratory findings, or results of basic imaging studies such as plain radiography or ultrasound
    ® Following documented recurrence of childhood cancer, any studies recommended for initial staging of that cancer type in the diagnosis-specific imaging guideline section should be approved
    ® During active treatment for recurrent pediatric cancer, conventional imaging evaluation (CT or MRI, should use the same modality for ongoing monitoring as much as possible) of previously involved areas should be approved according to the treatment response imaging in the diagnosis-specific guideline section:
      ¡ Imaging may be indicated more frequently than recommended by guidelines with clinical documentation that the imaging results are likely to result in a treatment change for the member, including a change from active treatment to surveillance
    ® Unless otherwise specified for a specific cancer type, PET is generally not indicated for routine treatment response evaluation during active treatment for recurrent pediatric cancer
      ¡ In rare circumstances, PET may be appropriate when results are likely to result in a treatment change for the member, including a change from active treatment to surveillance.
      ¡ These requests will be forwarded for Medical Director review.
    ® If a member with recurrent pediatric cancer completes active treatment with no evidence of disease (NED), s/he should be imaged according to the diagnosis-specific surveillance guideline sections

Radiation Treatment Planning In Pediatric Oncology:

Imaging performed in support of radiation therapy treatment planning should follow guidelines outlined in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-1.5: Unlisted Procedure Codes in Oncology.

Cardiac Function Assessment in Pediatric Oncology During Active Treatment:

Echocardiography (CPT® 93306, CPT® 93307, or CPT® 93308) is preferred for evaluation of cardiac function prior to cardiotoxic chemotherapy and can be performed as often as each chemotherapy cycle at the discretion of the treating pediatric oncologist based on:

    ® Cumulative cardiotoxic therapy received to date
    ® Member’s age and gender
    ® Most recent echocardiogram results
    ® New or worsening cardiac symptoms

Multi-gated acquisition (MUGA, CPT® 78472) blood pool nuclear medicine scanning should not be approved for cardiac function monitoring in pediatric oncology members unless one of the following applies:
    ® Echocardiography yielded a borderline shortening fraction (< 30%) and additional left ventricular function data are necessary to make a chemotherapy decision
    ® Echocardiography windowing is suboptimal due to body habitus or tumor location

Immunosuppression during Pediatric Cancer therapy and imaging ramifications:

Members may be severely immunocompromised during active chemotherapy treatment and any conventional imaging request to evaluate for infectious complications during this time frame should be approved immediately

Imaging requests for infectious disease concerns for all members with absolute neutrophil count (ANC) < 500 or inconclusive findings on Chest x-ray or US at any ANC during active treatment should be approved as requested

Additionally, members may have therapy-induced hypogammaglobulinemia which requires supplemental intravenous immune globulin (IVIG) during maintenance therapy. Members receiving supplemental IVIG should be treated similarly to members with ANC < 500 with regards to imaging for infectious disease.

Some members are treated with very intensive chemotherapy regimens (including autologous stem cell transplantation - See Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-29: Hematopoietic Stem Cell Transplantation and spend the majority of their chemotherapy treatment phase in the hospital. Due to the high risk of invasive infections, frequent CT may be indicated to evaluate known sites of invasive fungal infection, and in general these should be approved as requested.

    ® Surveillance imaging of asymptomatic members to detect invasive fungal infection has not been shown to impact member outcomes. Imaging requests in these circumstances should only be approved when acute clinical decisions will be made based on the imaging.

Hematopoietic Stem Cell Transplant (HSCT) in Pediatric Oncology:

Transplantation of hematopoietic stem cells from bone marrow, peripheral blood, or cord blood is commonly used in the following clinical situations in pediatric hematology and oncology members:

    ® High risk or recurrent leukemia (allogeneic)
    ® Recurrent lymphoma (allogeneic or autologous)
    ® Hemophagocytic lymphohistiocytosis (allogeneic)
    ® High risk sickle cell disease (allogeneic)
    ® High risk neuroblastoma (autologous)
    ® High risk CNS tumors (autologous)
    ® Recurrent Ewing sarcoma family of tumors (autologous, rarely allogeneic)

Imaging considerations for HSCT should follow guidelines in: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-29: Hematopoietic Stem Cell Transplantation.


PEDONC-1.3: Modality General Considerations

For this condition imaging is medically necessary based on the following criteria:

Plain radiography

    ® Chest x-ray (CXR) can provide a prompt means to evaluate primary intrathoracic tumors and continues to be the initial imaging study recommended to detect complications, such as suspected infection, in symptomatic members undergoing treatment.
    ® CXR continues to be the initial imaging study recommended for pulmonary surveillance for some pediatric cancers. See diagnosis-specific guideline sections for details.
    ® Plain radiography continues to be the initial imaging study recommended for evaluation of lesions involving the appendicular skeleton, both during and after completion of treatment. See diagnosis-specific guideline sections for details.
    ® Plain abdominal radiographs have been replaced by ultrasound, CT, or MRI

Ultrasound
    ® Ultrasound is not widely used in pediatric oncology for staging, but is frequently used for surveillance in members who have successfully treated (primarily abdominal or pelvic) tumors with little or no residual disease. See diagnosis-specific guideline sections for details.

CT
    ® CT with contrast is the imaging study of choice in pediatric members with lymphomas or solid tumors of the neck, thorax, abdomen, and/or pelvis
      ¡ If CT contrast use is contraindicated due to allergy or impaired renal function, either CT without contrast or MRI without and with contrast may be substituted at the discretion of the ordering physician
MRI
    ® MRI without and with contrast is the study of choice for CNS and musculoskeletal tumors
      ¡ If MRI contrast use is contraindicated due to allergy or impaired renal function, MRI without contrast may be substituted at the discretion of the ordering physician
    ® Due to the length of time for image acquisition and the need for stillness, anesthesia is required for almost all infants and young children (age < 7 years), as well as older children with delays in development or maturity. In this member population, MRI imaging sessions should be planned with a goal of avoiding a short-interval repeat anesthesia exposure due to insufficient information using the following considerations:
      ¡ MRI should always be performed without and with contrast unless there is a specific contraindication to gadolinium use, since the member already has intravenous access for anesthesia.
        § Recent evidence based literature demonstrates the potential for gadolinium deposition in various organs including the brain, after the use of MRI contrast.
        § The U.S. food and drug administration (FDA) has noted that there is currently no evidence to suggest that gadolinium retention in the brain is harmful and restricting gadolinium-based contrast agents (GBCAS) use is not warranted at this time. It has been recommended that GBCA use should be limited to circumstances in which additional information provided by the contrast agent is necessary and the necessity of repetitive MRIs with GBCAS should be assessed.
        § If requesting clinicians indicate that a non-contrast study is being requested due to concerns regarding the use of gadolinium, the exam can be approved.
      ¡ If multiple body areas are supported by Horizon BCBSNJ guidelines for the clinical condition being evaluated, MRI of all necessary body areas should be obtained concurrently in the same anesthesia session
Nuclear medicine
    ® General PET imaging consideration can be found in PEDONC-1.4: PET Imaging in Pediatric Oncology.
    ® Bone scan is frequently used for evaluation of bone metastases during initial treatment, treatment response, and surveillance in pediatric oncology
      ¡ For the purposes of these guidelines, any of the following codes can be approved where “bone scan” is indicated:
        § CPT® 78300
        § CPT® 78305
        § CPT® 78306
        § CPT® 78803
        § CPT® 78305 and CPT® 78803
        § CPT® 78306 and CPT® 78803
        § If CPT® 78300 and CPT® 78803are requested together, only CPT® 78803 should be approved
        § CPT® 78315 has no specific indications for evaluation of malignant disease
    ® 123I-metaiodobenzylguanidine (MIBG) scintigraphy is the preferred metabolic imaging for neuroblastoma and is positive in 90 to 95% of neuroblastomas, and is also used for evaluation of pheochromocytomas, paragangliomas, ganglioneuromas, and ganglioneuroblatomas
      ¡ For the purposes of these guidelines, any of the following codes can be approved where “MIBG” is indicated:
        § CPT® 78800
        § CPT® 78801
        § CPT® 78802
        § CPT® 78803
        § CPT® 78804
    ® Octreotide and gallium scans use the same CPT codes as MIBG
    PEDONC-1.4: PET Imaging in Pediatric Oncology

    For this condition imaging is medically necessary based on the following criteria:

    Note: Some payors have specific restrictions on PET imaging, and those coverage policies may supersede the recommendations for PET imaging in these guidelines.

    Throughout these guidelines, the term “PET” refers specifically to 18F-FDG-PET imaging and also applies to PET/CT fusion studies.

    PET imaging in pediatric Oncology should use PET/CT fusion imaging (CPT® 78815 or CPT® 78816) unless there is clear documentation that the treating facility does not have fusion capacity, in which case PET alone (CPT® 78812 or CPT® 78813) can be approved along with the appropriate CT studies. Unbundling PET/CT imaging into separate PET and diagnostic CT codes is otherwise not supported.

    The decision whether to use skull base to mid-femur (“eyes to thighs”) procedure code for PET (CPT® 78812 or CPT® 78815) or whole body PET (CPT® 78813 or CPT® 78816) is addressed in the diagnosis-specific guideline sections.

    PET imaging is not reliable for the detection of anatomic lesions smaller than 8 mm in size.

    PET imaging using isotopes other than 18F-FDG and 68Ga-DOTATATE is considered investigational at this time.

    PET has not been shown to be diagnostically useful in all forms of childhood cancer. PET is supported for pediatric malignancies with significant published evidence regarding its diagnostic accuracy and importance in accurately directing member care decisions. See diagnosis-specific guideline sections for details.

    PET imaging is not specific to cancer, and has a high rate of false positivity. Inflammation, infection (especially granulomatous), trauma, and post-operative healing may show high levels of FDG uptake and be false-positive for malignant lesions.

    PET for rare malignancies not specifically addressed by Horizon BCBSNJ guidelines is generally not indicated, due to lack of available evidence regarding diagnostic accuracy of PET in the majority of rare cancers. Conventional imaging studies should be used for initial staging and treatment response for these diagnoses. PET can be approved if all of the following apply:

      ® Conventional imaging (CT, MRI, US, plain film) reveals findings that are equivocal or suspicious
      ® No other specific metabolic imaging (MIBG, octreotide, technetium, etc.) Is appropriate for the cancer type
      ® The submitted clinical information describes a specific decision regarding the member’s care that will be made based on the PET results
      ® These requests will be forwarded for Medical Director review

    PET imaging is not supported for surveillance imaging unless specifically stated elsewhere in the diagnosis-specific guideline sections

    Unless otherwise specified for a specific cancer type, once PET has been documented to be negative for a given member’s cancer or all PET-avid disease has been surgically resected, PET should not be used for continued disease monitoring or surveillance unless one of the following applies:

      ® Conventional imaging (CT, MRI, US, plain film) reveals findings that are inconclusive or suspicious for recurrence
        ¡ Residual mass that has not changed in size since the last conventional imaging does not justify PET imaging
        ¡ PET avidity in a residual mass at the end of planned therapy is not an indication for PET imaging during surveillance.
      ® Very rare circumstances where tumor markers or obvious clinical symptoms show strong evidence suggesting recurrence and PET would replace conventional imaging modalities
      ® The member is undergoing salvage treatment for a recurrent solid tumor with residual measurable disease on conventional imaging and confirmed repeat negative PET imaging will allow the member to transition from active treatment to surveillance.
      ® These requests will be forwarded for Medical Director review.
    PEDONC-1.5: Diagnostic Radiation Exposure in Pediatric Oncology

    For this condition imaging is medically necessary based on the following criteria:

    1. Young children are presumed to be at increased risk for malignancy from diagnostic radiation exposure, most commonly from CT and nuclear medicine imaging. They are more sensitive to radiation than adults and generally live longer after receiving radiation doses from medical procedures, resulting in a larger number of years during which to manifest a cancer.

    Because of this presumed increased risk in young children, requests to substitute MRI without and with contrast for CT with contrast to avoid radiation exposure can be approved if all of the following criteria apply:

      ® The member is presently a young child and the ordering physician has documented the reason for MRI, rather than CT, is to avoid radiation exposure.
      ® The disease-specific guidelines do not list CT as superior to MRI for the current disease and time point, meaning the MRI will provide equivalent or superior information relative to CT.
      ® The request is for a body area other than Chest as MRI is substantially inferior to CT for detection of small pulmonary metastases.
    The guidelines listed in this section for certain specific indications are not intended to be all-inclusive; clinical judgment remains paramount and variance from these guidelines may be appropriate and warranted for specific clinical situations.

    References

    1. Krishnamurthy R, Daldrup-Link HE, Jones JY, et al. Imaging studies in the diagnosis and management of pediatric malignancies. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:185-238.
    2. Allen-Rhoades W, Steuber CP. Clinical assessment and differential diagnosis of the child with suspected cancer. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:101-112.
    3. Gottschalk S, Naik S, Hegde M, et al. Hematopoietic stem cell transplantation in pediatric oncology. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:419-440.
    4. Freedman JL, Rheingold SR, and Fisher MJ. Oncologic emergencies. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:967-991.
    5. Andrews J, Galel SA, Wong W, et al. Hematologic supportive care for children with cancer. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:992-1009.
    6. Ardura MI, Koh AY. Infectious complications in pediatric cancer patients. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:1010-1057.
    7. Shaikh R, Prabhu SP, Voss SD. Imaging in the evaluation and management of childhood cancer. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:2146-2254.
    8. Sung L, Fisher BT, Koh AY. Infectious Disease in the pediatric cancer patient. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:2257-2266.
    9. Mullen EA, Gratias E. Oncologic emergencies. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:2267-2291.
    10. Weiser DA, Kaste SC, Siegel MJ, et al. Imaging in childhood cancer: A society for Pediatric Radiology and Children’s Oncology Group joint task force report. Pediatr Blood Cancer. 2013;60(8):1253-1260. doi:10.1002/pbc.24533.
    11. American College of Radiology. ACR-ASER-SCBT-MR-SPR Practice Parameter for the Performance of Pediatric Computed Tomography (CT). 2014. https://www.acr.org/-/media/ACR/Files/Practice-Parameters/CT-Ped.pdf .
    12. ACR-SPR Practice Parameter for the Performance and Interpretation of Pediatric Magnetic Resonance Imaging (MRI), Revised 2015 (Resolution 11). https://www.acr.org/~/media/CB384A65345F402083639E6756CE513F.pdf
    13. Smith EA, Dillman JR. Current role of body MRI in pediatric oncology. Pediatr Radiol. 2016;46(6):873-880. doi:10.1007/s00247-016-3560-8.
    14. The Center for Drug Evaluation and Research. Meeting of the Medical Imaging Drugs Advisory Committee, presented September 8, 2017. https://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/MedicalImagingDrugsAdvisoryCommittee/UCM574746.pdf.
    15. Uslu L, Doing J, Link M, et al. Value of 18F-FDG PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
    16. McCarville MB. PET-CT imaging in pediatric oncology. Cancer Imaging. 2009(1);9:35-43. doi:10.1102/1470-7330.2009.0008.
    17. Matthews JD, Forsythe AV, Brady Z, et al. Cancer risk in 680 000 people exposed to computed tomography scans in childhood or adolescence: data linkage study of 11 million Australians. BMJ. 2013;346:f2360. doi:10.1136/bmj.f2360.
    18. Erdi YE. Limits of tumor detectability in nuclear medicine and PET. Mol Imaging Radio Nucl Ther. 2012;21(1):23-28. doi:10.4274/Mirt.138.
    19. Ing C, DiMaggio C, Whitehouse A, et al. Long-term differences in language and cognitive function after childhood exposure to anesthesia. Pediatrics. 2012;130(3):e476-e485. doi:10.1542/peds.2011-3822.
    20. Monteleone M, Khandji A, Cappell J, et al. Anesthesia in children: perspectives from nonsurgical pediatric specialists. J Neurosurg Anesthesiol. 2014;26(4):396-398. doi:10.1097/ANA.0000000000000124.
    21. DiMaggio C, Sun LS, Li G. Early childhood exposure to anesthesia and risk of developmental and behavioral disorders in a sibling birth cohort. Anesth Analg. 2011;113(5):1143-1151. doi:10.1213/ANE.0b013e3182147f42.
    22. Brody AS, Guillerman RP. Don’t let radiation scare trump patient care: 10 ways you can harm your patients by fear of radiation-induced cancer from diagnostic imaging. Thorax. 2014;69(8):782-784. doi:10.1136/thoraxjnl-2014-205499.
    23. Meulepas JM, Ronckers CM, Smets AMJB, et al. Radiation exposure from pediatric CT scans and subsequent cancer risk in the Netherlands, JNCI J Natl Cancer Inst. 2019;111:256-2634. doi: 10.1093/jnci/djy104.
    24. Bhatia S, Pappo AS, Acquazzino M, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 1.2020—July 11, 2019, Adolescent and Young Adult (AYA) Oncology, available at: https://www.nccn.org/professionals/physician_gls/pdf/aya.pdf, referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Adolescent and Young Adult (AYA) Oncology V1.2020 7/11/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org.


    PEDONC-2: Screening Imaging in Cancer Predisposition Syndromes

    PEDONC-2.1: Screening Imaging in Cancer Predisposition Syndromes – General Considerations
    PEDONC-2.2: Li-Fraumeni Syndrome (LFS)
    PEDONC-2.3: Neurofibromatosis 1 and 2 (NF1 and NF2)
    PEDONC-2.4: Beckwith-Wiedemann Syndrome (BWS)
    PEDONC-2.5: Denys-Drash Syndrome (DDS)
    PEDONC-2.6: Wilms Tumor-Aniridia-Growth Retardation (WAGR)
    PEDONC-2.7: Familial Adenomatous Polyposis (FAP) and Related Conditions
    PEDONC-2.8: Multiple Endocrine Neoplasias (MEN)
    PEDONC-2.9: Tuberous Sclerosis Complex (TSC)
    PEDONC-2.10: Von Hippel-Lindau Syndrome (VHL)
    PEDONC-2.11: Rhabdoid Tumor Predisposition Syndrome
    PEDONC-2.12: Familial Retinoblastoma Syndrome
    PEDONC-2.13: Hereditary Paraganglioma-Pheochromocytoma (HPP) Syndromes
    PEDONC-2.14: Costello Syndrome
    PEDONC-2.15: Constitutional Mismatch Repair Deficiency (CMMRD or Turcot Syndrome)
    PEDONC-2.1: General Considerations

    PEDONC-2.1: Screening Imaging in Cancer Predisposition Syndromes – General Considerations

    For this condition imaging is medically necessary based on the following criteria:

    This section is intended to give guidance for screening imaging prior to diagnosis with a specific malignancy. Once a member with a cancer predisposition syndrome has been diagnosed with a malignant disease, future imaging decisions should be guided by the appropriate disease-specific guidelines except as explicitly stated elsewhere in this section.

    This section’s guidelines are limited to cancer predisposition syndromes with screening imaging considerations. Syndromes requiring only clinical or laboratory screening are not discussed here.

    In general, a recent (within 60 days) detailed history and physical examination and appropriate laboratory studies should be performed prior to considering advanced imaging, unless the member is undergoing guideline-supported scheduled screening evaluation identified in this section

    Many of these cancer predisposition syndromes also affect adults as survival continues to improve for these members. Adults with syndromes covered in this section may follow these imaging guidelines except where contradicted by specific statements in the adult imaging guidelines or payor-specific coverage policies.
    Documentation of genetic or molecular confirmation of the appropriate syndrome with increased cancer risk is preferred for any member to qualify for screening imaging. There are a number of complex ethical, social, and financial issues involved in the decision to complete genetic testing in a pediatric member:

    Note: Some payors consider certain genetic tests to be investigational, and those coverage policies supersede the recommendations for genetic testing in this section.

    From the 2013 AAP Policy Statement, “Predictive genetic testing for adult-onset conditions generally should be deferred unless an intervention initiated in childhood may reduce morbidity or mortality.” Imaging surveillance is one such intervention and should not be performed without justifiable cause.

    Genetic testing should be performed in conjunction with genetic counseling for appropriate communication of risks identified by testing

    When genetic testing is not possible or not supported by health plan coverage policies, formal diagnosis after evaluation by a physician with significant training and/or experience in cancer predisposition syndromes (most commonly a geneticist or oncologist) is generally sufficient to confirm eligibility for screening imaging.

    Due to the length of time for image acquisition and the need for stillness, anesthesia is required for almost all infants and young children (age < 7 years), as well as older children with delays in development or maturity. In this member population, MRI imaging sessions should be planned with a goal of avoiding a short-interval repeat anesthesia exposure due to insufficient information using the following considerations:

      ® MRI should always be performed without and with contrast unless there is a specific contraindication to gadolinium use, since the member already has intravenous access for anesthesia.
        ¡ Recent evidence based literature demonstrates the potential for gadolinium deposition in various organs including the brain, after the use of MRI contrast.
        ¡ The U.S. food and drug administration (FDA) has noted that there is currently no evidence to suggest that gadolinium retention in the brain is harmful and restricting gadolinium-based contrast agents (GBCAS) use is not warranted at this time. It has been recommended that GBCA use should be limited to circumstances in which additional information provided by the contrast agent is necessary and the necessity of repetitive MRIs with GBCAS should be assessed.
        ¡ If requesting clinicians indicate that a non-contrast study is being requested due to concerns regarding the use of gadolinium, the exam can be approved.
      ® If multiple body areas are supported by Horizon BCBSNJ guidelines for the clinical condition being evaluated, MRI of all necessary body areas should be obtained concurrently in the same anesthesia session
    PEDONC-2.2: Li-Fraumeni Syndrome (LFS)

    For this condition imaging is medically necessary based on the following criteria:

    Syndrome inherited in an autosomal dominant manner (50% risk to offspring) associated with germline mutations in TP53 resulted in an increased susceptibility to a variety of cancers.

    Eighty percent of individuals will have germline TP53 mutation:

      ® Tumor-specific TP53 mutations are much more common than germline TP53 mutations and are not associated with an increased risk for subsequent cancers
      ® If TP53-negative, formal diagnosis of LFS should be assigned by a physician with significant training and/or experience in LFS (most commonly a geneticist or oncologist) based on specified clinical criteria prior to beginning a screening imaging program
      ® TP53 mutations may be present in 50 to 80% of pediatric adrenocortical carcinoma, 10% of pediatric rhabdomyosarcoma, and 10% of pediatric osteosarcoma members

    Members with LFS have an increased sensitivity to ionizing radiation, so screening strategies resulting in significant radiation exposure are not appropriate (CT and nuclear medicine).

    The following imaging studies should be considered appropriate in members with LFS:

    Annual complete detailed physical examinations, complete blood counts, and urinalyses form the backbone of LFS cancer screening.
    Annual MRI Brain without and with contrast (CPT® 70553) for all members

    Annual whole-body MRI (WBMRI, CPT® 76498) for all members

    Substantial variation continues to exist in WBMRI techniques, and a specific CPT code for WBMRI has not yet been assigned. As a result, CPT® 76498 is the only approvable code for a WBMRI study at this time.

    Abdominal (CPT® 76700) and Pelvic (CPT® 76856) ultrasound every 3 months from birth to age 18 (for adrenocortical carcinoma screening)

    Annual Breast MRI (CPT® 77049) alternating every 6 months with breast ultrasound for breast cancer screening is appropriate for LFS members beginning at age 20 (See BR-6: Breast MRI Indications)

    Targeted MRI imaging without and with contrast of any body area(s) with documented signs or symptoms suggestive of possible malignancy

    When a specific malignancy is suspected, the member should be imaged according to the Horizon BCBSNJ imaging guideline specific to the suspected cancer type

    Studies ordered as part of a screening imaging program based on specific family cancer history that has been developed for an individual member in conjunction with a multidisciplinary team including at least genetics and Oncology

    Specifics of the program should be obtained and available for the Medical Director reviewing the case

    PEDONC-2.3: Neurofibromatosis 1 and 2 (NF1 and NF2)

    For this condition imaging is medically necessary based on the following criteria:

    NF1:

    Common syndrome inherited in an autosomal dominant manner (50% risk to offspring) affecting 1 in 2500 people. The diagnosis is commonly made based on established clinical criteria including café-au-lait spots, lisch nodules of the iris, axillary freckling, family history, and the presence of NF-associated tumors.

    Genetic testing is encouraged for children with possible NF1 and no family history prior to assigning a diagnosis, but will not identify a mutation for all members with NF1. The majority of tumors are benign in nature, but malignant degeneration can occur. The most frequent neoplasms associated with NF1 in children are malignant peripheral nerve sheath tumor (MPNST), glioma, pheochromocytoma, and leukemia.

    NF1-affected persons have increased sensitivity to ionizing radiation, so CT and nuclear medicine imaging are not appropriate screening or surveillance studies for these members. CT and/or nuclear medicine studies may be indicated for acute clinical situations and should be judged on a case-by-case basis. These requests will be forwarded for Medical Director review.

    Annual ophthalmology evaluation is strongly recommended beginning at the time of diagnosis of NF1 to evaluate for optic pathway abnormalities:

    Screening MRIs of the Brain (CPT® 70553) and Orbits (CPT® 70543) for asymptomatic individuals are not generally recommended due to the ~60% rate of unidentified bright objects (UBOs, T2-weighted signal abnormalities) which mostly disappear by age 30

      ® A one-time MRI Brain (CPT® 70553) and Orbits (CPT® 70543) without and with contrast can be approved to clarify the diagnosis of NF1 if evaluation by a physician with significant training and/or experience in neurofibromatosis is inconclusive (most commonly a neurologist, geneticist, ophthalmologist, or oncologist)
      ® MRI Brain (CPT® 70553) and Orbits (CPT® 70543) without and with contrast can be approved for any new or worsening symptoms
      ® Routine follow up imaging of UBOs is not warranted in the absence of acute symptoms suggesting new or worsening intracranial disease
      ® Children with negative brain and orbital screening at age 15 months generally do not develop optic pathway gliomas

    Members with NF1 and documented optic pathway gliomas should be imaged according to PEDONC-4.2: Intracranial Low Grade Gliomas (LGG).


    NF1 members are at increased risk for plexiform neurofibromas (PN) and malignant peripheral nerve sheath tumors (MPNST— a high grade sarcoma).

    Screening imaging of asymptomatic members for these tumors is not supported by evidence. PET imaging is not supported for PN surveillance in asymptomatic members at this time as the positive predictive value is only 60 to 65% even in symptomatic members.

    MRI imaging without and with contrast is appropriate for any clinical symptoms suggestive of change in a known PN in a member with NF1 (examples include pain, rapid growth, and neurologic dysfunction).

    Although PET imaging has a positive predictive value of only 61 to 63% in NF1 members with suspected transformation to MPNST, the negative predictive value is high (96 to 99%)

      ® PET imaging is indicated for evaluating NF1 members with clinical symptoms concerning for malignant transformation of a known PN when all of the following conditions exist:
        ¡ Recent MRI is inconclusive regarding transformation or progression
        ¡ Negative PET will result in a decision to avoid biopsy in a difficult or morbid location
      ® Inconclusive PET findings should lead to biopsy of the concerning lesion
        ¡ Repeat PET studies are not indicated due to the poor positive predictive value in this setting
    Members with NF1 and known plexiform neurofibromas should be imaged according guidelines in Pediatric Peripheral Nerve Disorder Imaging Policy (Policy #168 in the Radiology Section); PEDPN-2.1: Neurofibromatosis 1.

    Members with NF1 and new soft tissue masses should be imaged according to Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-12: Sarcoma or PEDONC-8.3: Non-Rhabdomyosarcoma Soft Tissue Sarcomas, depending on the member’s age at the time the mass is discovered.

    Members with NF1 and new bone masses should be imaged according to PEDONC-9: Bone Tumors.

    NF2:

    NF2 is substantially less common than NF1. It is inherited in an autosomal dominant manner (50% risk to offspring) affecting ~1 in 25000 people. NF2 is associated with increased risk for meningiomas (50% of affected individuals), vestibular schwannomas, and spinal tumors (75% of affected individuals).

    Members with NF2 and known vestibular schwannomas should be imaged according to guidelines in Pediatric Peripheral Nerve Disorder Imaging Policy (Policy #168 in the Radiology Section); PEDPN-2.2: Neurofibromatosis 2.

    Members with NF2 and known meningioma should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-2.8: Meningiomas

    Members with NF2 and known ependymoma should be imaged according to guidelines in PEDONC-4: Ependymoma.

    Recommended cancer screening imaging includes:

    Annual MRI Brain without and with contrast (CPT® 70553) beginning at age 10 years

    MRI spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved every 3 years beginning at age 10 years for members without spinal tumors

      ® Annual MRI spine can be approved for all members with NF2 and a history of spinal tumors

    Additional appropriate imaging requests include:

    MRI Brain without and with contrast (CPT® 70553) should be approved for any member with NF2 and clinical symptoms of intracranial mass or vestibular disease

    MRI spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) should be approved for any member with NF2 and:

      ® Clinical symptoms suggestive of new or progressive spinal or paraspinal tumors, including uncomplicated back pain or radiculopathy
      ® Recent diagnosis with a meningioma or vestibular schwannoma
    PEDONC-2.4: Beckwith-Wiedemann Syndrome (BWS)

    For this condition imaging is medically necessary based on the following criteria:

    Inherited syndrome characterized by macroglossia, hemihypertrophy, macrosomia, organomegaly, and neonatal hypoglycemia. Members with isolated hemihypertrophy are also imaged according to this guideline.

    Caused by mutation at chromosome 11p15, affected children are predisposed to Wilms tumor, hepatoblastoma, rhabdomyosarcoma, and adrenal tumors.

    Recommended cancer screening imaging includes:

    Abdominal ultrasound (CPT® 76700) every 3 months from birth to the 8th birthday

      ® Members found to have adrenal masses on screening ultrasound should receive additional imaging as follows:
        ¡ Purely cystic mass:
          § Continue screening ultrasound every 3 months without additional imaging
        ¡ Solid or mixed mass in members age 0 to 5 months:
          § If mass 0 to 3 cm in diameterMIBG imaging and either CT or MRI Abdomen (contrast as requested)
            · If no evidence of malignancy based on MIBG, CT or MRI, Urine HVA/VMA, and serum ACTH, then repeat abdominal ultrasound every 6 weeks for 2 years
          § If mass > 3 cm in diameterMIBG imaging and MRI Abdomen (contrast as requested)
        ¡ Solid or mixed mass in members age 6 months or greater:
          § MIBG imaging prior to biopsy or resection
          § If no evidence of malignancy on biopsy or resection, resume screening abdominal ultrasound every 3 months
    Members with BWS and known renal tumors should be imaged according to guidelines in PEDONC-7: Pediatric Renal Tumors.

    Members with BWS and known hepatoblastoma should be imaged according to guidelines in PEDONC-11.2: Hepatoblastoma.

    Members with BWS and known neuroblastoma should be imaged according to guidelines in PEDONC-6: Neuroblastoma.

    Members with BWS and known adrenocortical carcinoma should be imaged according to guidelines in PEDONC-14: Pediatric Adrenocortical Carcinoma.

    Members with BWS and known pheochromocytoma should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-15: Neuroendocrine Cancers and Adrenal Tumors.

    PEDONC-2.5: Denys-Drash Syndrome (DDS)

    For this condition imaging is medically necessary based on the following criteria:

    Characterized by pseudohermaphroditism, early renal failure, and > 90% risk of Wilms tumor development in each kidney. Associated with mutations at 11p13, risk of renal failure after detection of symptomatic Wilms tumor is 62%, so early detection may allow for renal-sparing surgical approaches.

    Recommended cancer screening imaging includes:

    Abdominal ultrasound (CPT® 76700) every 3 months from birth to the 8th birthday

    Members with DDS and known renal tumors should be imaged according to guidelines in PEDONC-7: Pediatric Renal Tumors.

    PEDONC-2.6: Wilms Tumor-Aniridia-Growth Retardation (WAGR)

    For this condition imaging is medically necessary based on the following criteria:

    Named for the components of the disorder, it is associated with mutations at 11p13. As the name suggests, members are predisposed to Wilms tumor, with 57% of members in one cohort developing Wilms tumor. Risk of renal failure after detection of symptomatic Wilms tumor is 38%, so early detection may allow for renal-sparing surgical approaches

    Recommended cancer screening imaging includes:

    Abdominal US (CPT® 76700) every 3 months from birth to the 8th birthday

    Members with WAGR and known renal tumors should be imaged according to guidelines in PEDONC-7: Pediatric Renal Tumors.

    PEDONC-2.7: Familial Adenomatous Polyposis (FAP) and Related Conditions

    For this condition imaging is medically necessary based on the following criteria:

    Inherited in an autosomal dominant manner (50% risk to offspring), it is also known as Adenomatous Polyposis Coli (APC). It is associated with the development of thousands of colonic polyps by age 20 and > 90% risk of colorectal carcinoma. Prophylactic total colectomy is recommended by age 20 for most members. FAP is also associated with hepatoblastoma, tumors of the pancreas and small bowel, medulloblastoma, and thyroid cancer.

    Members with Lynch, Gardner, and Turcot syndromes should also be imaged according to these guidelines.

    Recommended cancer screening imaging includes:

    Abdominal US (CPT® 76700) every 3 months from birth to the 6th birthday

      ® Annual Abdominal US for life after age 6 with family history of desmoid tumors

    Serum AFP every 3 months to the 6th birthday

    Annual colonoscopy beginning at age 10

    Annual esophagogastroduodenoscopy beginning at age 10

    Annual thyroid ultrasound (CPT® 76536) beginning at age 12

    Annual pelvic ultrasound (CPT® 76856) beginning at age 30

    Members with FAP and known colorectal tumors should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-16: Colorectal Cancer.

    Members with FAP and known desmoid tumors should be imaged according to guidelines in PEDONC-8.3: Non-Rhabdomyosarcoma Soft Tissue Sarcomas (NRSTS).

    PEDONC-2.8: Multiple Endocrine Neoplasias (MEN)

    For this condition imaging is medically necessary based on the following criteria:

    Inherited in an autosomal dominant manner (50% risk to offspring)

    MEN1 is characterized by parathyroid, pancreatic islet cell, and pituitary gland tumors (3 P’s), as well as carcinoid tumors in the chest and abdomen, and 28% of members will develop at least one tumor by age 15.

    MEN2a is characterized by medullary thyroid carcinoma, parathyroid adenomas, and pheochromocytomas.

    MEN2b is characterized by ganglioneuromas of the GI tract and skeletal abnormalities presenting in infancy.

    Recommended cancer screening imaging includes:

    MEN1

      ® Annual MRI Brain without and with contrast (CPT® 70553) can be approved beginning at age 5
      ® Annual MRI Abdomen without and with contrast (CPT® 74183), CT Abdomen with contrast (CPT® 74160), or ultrasound (CPT® 76700) can be approved beginning at age 5
      ® Annual MRI Chest without and with contrast (CPT® 71552) or CT Chest with contrast (CPT® 71260) can be approved beginning at age 15
      ® Annual Octreotide study (CPT® 78800, CPT® 78801, CPT® 78802, CPT® 78803, or CPT® 78804) can be approved beginning at age 5

    Members with MEN1 and known thyroid cancer should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-6: Thyroid Cancer

    Members with MEN1 and known pheochromocytoma should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-15: Neuroendocrine Cancers and Adrenal Tumors

    MEN2a and MEN2b

      ® Annual measurement of catecholamines for pheochromocytoma screening
      ® MRI Abdomen without and with contrast (CPT® 74183) can be approved every 3 years beginning at age 5
      ® Octreotide study (CPT® 78800, CPT® 78801, CPT® 78802, CPT® 78803, or CPT® 78804) or Adrenal Nuclear Imaging (CPT® 78075) can be approved for elevated catecholamines or inconclusive adrenal mass on MRI

    Members with MEN2a or MEN2b and known pheochromocytoma should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-15: Neuroendocrine Cancers and Adrenal Tumors

    PEDONC-2.9: Tuberous Sclerosis Complex (TSC)

    For this condition imaging is medically necessary based on the following criteria:

    Inherited in an autosomal dominant manner (50% risk to offspring), affecting ~1 in 6000 individuals, it is associated with benign tumors, hypopigmented skin macules (ash leaf spots), pulmonary lymphangioleiomyomatosis, developmental delay, and epilepsy.

    Malignancies associated with this syndrome include:

    Subependymal giant cell astrocytomas (SEGA tumors)

      ® Historically, early surgery was important to reduce morbidity related to these tumors
      ® More recently, everolimus has been successfully used to treat these tumors without surgery, and early detection remains an important feature for success

    Renal cell carcinoma

    Cardiac rhabdomyosarcoma

    Pulmonary lymphangioleiomyomatosis

    Recommended cancer screening imaging includes:

    Annual ophthalmologic and dermatologic evaluation

    Annual Brain MRI without and with contrast (CPT® 70553) beginning at age 3 until age 25

    Annual Renal US (CPT® 76770) beginning at age 3

      ® Annual MRI Abdomen without and with contrast (CPT® 74183) can be substituted for Renal US in members with documented renal lesions

    Annual Echocardiography

    CT Chest without contrast (CPT® 71250) every 5 years beginning at age 18 years

      ® Additional CTs may be approved every 1 year for members with documented abnormalities
      ® CT Chest without contrast should be approved for evaluation of any new pulmonary symptoms or worsening pulmonary function testing

    Members with TSC and known SEGA tumors should be imaged according to PEDONC-4.2: Intracranial Low Grade Gliomas (LGG)

    Members with TSC and known renal cell carcinoma should be imaged according to PEDONC-7.4: Pediatric Renal Cell Carcinoma (RCC)



    PEDONC-2.10: Von Hippel-Lindau Syndrome (VHL)

    For this condition imaging is medically necessary based on the following criteria:

    Inherited in an autosomal dominant manner (50% risk to offspring), it is associated with CNS hemangioblastomas, retinal angiomas, endolymphatic sac tumors (ELST), gastrointestinal stromal tumor (GIST), renal cell carcinoma (RCC), and pheochromocytomas and other neuroendocrine tumors (NETs). Pediatric members are at risk of developing hemangioblastomas and pheochromocytomas that can remain clinically occult until symptoms become severe. Historically, substantial mortality was attributable to RCC, pancreatic NET, and CNS hemangioblastoma.

    Recommended cancer screening imaging includes:

    Annual ophthalmologic evaluation beginning at birth

    Annual measurement of catecholamines beginning at age 2

      ® Octreotide study (CPT® 78800, CPT® 78801, CPT® 78802, CPT® 78803, or CPT® 78804) or Adrenal Nuclear imaging (CPT® 78075) can be approved for elevated catecholamines or inconclusive adrenal mass on MRI

    Audiology assessment every 2 years beginning at age 5
      ® If frequent ear infections are present, MRI Brain without and with contrast (CPT® 70553) with attention to internal auditory canals can be approved

    MRI Brain without and with contrast (CPT® 70553) every 2 years beginning at age 8
      ® Members with known hemangioblastoma that has not been resected can have MRI Brain every 1 year or for any new or worsening symptoms

    MRI Spine without and with contrast (Cervical-CPT® 72156), Thoracic-CPT® 72157, and Lumbar-CPT® 72158) every 2 years beginning at age 8
        ¡ Members with known hemangioblastoma that has not been resected can have MRI Spine every 1 year or for any new or worsening symptoms
    Annual Abdominal US (CPT® 76700) beginning at age 5

    MRI Abdomen without and with contrast (CPT® 74183) every 2 years beginning at age 10

    Members with VHL and known CNS Hemangioblastoma should be imaged according to PEDONC-4.2: Intracranial Low Grade Gliomas (LGG)

    Members with VHL and known renal cell carcinoma should be imaged according to PEDONC-7.4: Pediatric Renal Cell Carcinoma (RCC)

    Members with VHL and known pheochromocytoma or other neuroendocrine tumors should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-15: Neuroendocrine Cancers And Adrenal Tumors



    PEDONC-2.11: Rhabdoid Tumor Predisposition Syndrome

    For this condition imaging is medically necessary based on the following criteria:

    Inherited in an autosomal dominant manner (50% risk to offspring), it is associated with malignant rhabdoid tumors of the kidney and extrarenal locations, and atypical teratoid/rhabdoid tumors (ATRT) of the CNS. It is caused by a germline mutation in INI1 or SMARCB1, and is associated with a more variable prognosis than de novo rhabdoid tumors.

    Targeted advanced imaging should be approved for any member with this syndrome and any clinical symptoms to suggest malignancy

    Ultrasound of the head (CPT® 76506), abdomen (CPT® 76700), and pelvis (CPT® 76856) monthly from birth to 12 months of age

    MRI can be approved for clarification of inconclusive findings on ultrasound, and should be used in place of ultrasound for remainder of planned screening

    MRI Brain (CPT® 70553) and Spine (CPT® 72156, 72157, & 72158) without and with contrast every 3 months from age 1 to 5 years

    MRI (CPT® 74183 & 72197) or Ultrasound Abdomen and Pelvis (CPT® 76700 & 76856) every 3 months from age 1 to 5 years

      ® Whole-body MRI resolution may not be sufficient to detect small rhabdoid tumors, so is not recommended in lieu of conventional MRI studies

    PEDONC-2.12: Familial Retinoblastoma Syndrome

    For this condition imaging is medically necessary based on the following criteria:

    This syndrome is inherited in an autosomal dominant manner (50% risk to offspring). As the name suggests, it is associated with retinoblastoma, as well as osteosarcoma, pediatric melanoma, and a significantly increased risk for radiation-related malignancies.

    Regular physical and ophthalmologic evaluations under anesthesia (EUA) are the hallmark of surveillance strategies for these members, and asymptomatic screening imaging does not have a defined role at this time.

    Members with retinomas (premalignant retinal lesions) can have annual MRI Orbits (CPT® 70543)

    When advanced imaging is necessary for evaluation of inconclusive EUA findings or new symptoms, ultrasound or MRI should be used if at all possible in lieu of CT or nuclear imaging if at all possible to avoid radiation exposure in these members


    PEDONC-2.13: Hereditary Paraganglioma-Pheochromocytoma (HPP) Syndromes

    For this condition imaging is medically necessary based on the following criteria:

    Caused by mutations in SDHx genes, this syndrome is inherited in an autosomal dominant manner (50% risk to offspring), and is associated with pheochromocytomas and paragangliomas.

    Members with multiple endocrine neoplasias should not use this guideline and should be imaged according to PEDONC-2.8: Multiple Endocrine Neoplasias (MEN).

    Cancer screening should begin at age 6. The following recommended imaging can be approved:

    All members with SDHx mutations:

      ® Annual measurement of catecholamines
      ® One of the following every 2 years:
        ¡ Whole body MRI (CPT® 76498)
        ¡ MRI Neck (CPT® 70543), Chest (CPT® 71552), Abdomen (CPT® 74183), Pelvis (CPT® 72197) without and with contrast
        ¡ CT Neck (CPT® 70491), Chest (CPT® 71260), and Abdomen/Pelvis (CPT® 74177) with contrast
        ¡ MRI is preferred to CT to minimize radiation exposure given these members’ lifelong need for screening
    Members with HPP and known pheochromocytoma or other neuroendocrine tumors should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-15: Neuroendocrine Cancers and Adrenal Tumors


    PEDONC-2.14: Costello Syndrome

    For this condition imaging is medically necessary based on the following criteria:

    Caused by mutations in HRAS genes, this syndrome is inherited in an autosomal dominant manner (50% risk to offspring), and is associated with rhabdomyosarcoma and neuroblastoma in early childhood, and transitional cell cancer of the bladder in older children and adults.

    Recommended Screening Imaging Includes:

    Following initial diagnosis, any or all of the following are indicated:

      ® Echocardiogram (CPT® 93306)
      ® MRI Brain (CPT® 70553) without and with contrast
      ® MRI Cervical (CPT® 72156) and Thoracic Spine (CPT® 72157) without and with contrast

    Ultrasound of the Abdomen (CPT® 76700) and Pelvis (CPT® 76856) every 3 months from birth to 10th birthday

    Echocardiogram (CPT® 93306) as requested for members with Costello syndrome and known cardiac disease

    Members with Costello syndrome and known rhabdomyosarcoma should be imaged according to guidelines in PEDONC-8.2: Rhabdomyosarcoma (RMS)

    Members with Costello syndrome and known neuroblastoma should be imaged according to guidelines in PEDONC-6: Neuroblastoma

    PEDONC-2.15: Constitutional Mismatch Repair Deficiency (CMMRD or Turcot Syndrome)

    For this condition imaging is medically necessary based on the following criteria:

    A highly penetrant and aggressive cancer predisposing syndrome resulting from autosomal recessive inheritance of biallelic mutations in mismatch repair genes, CMMRD syndrome leads to substantial risk for several commonly fatal childhood malignancies - high-grade CNS tumors (glioma, PNET, medulloblastoma) and hematologic malignancies (non-Hodgkin lymphoma, acute lymphoblastic leukemia). CMMRD members are also at increased risk for gastrointestinal tumors.

    Recommended Screening Imaging Includes:

    MRI Brain without and with contrast (CPT® 70553) every 6 months after CMMRD diagnosis is confirmed

    Annual whole body MRI (CPT® 76498) beginning at age 6 years

    Annual esophagogastroduodenoscopy and colonoscopy beginning at age 4 years

    References

    1. Plon SE, Malkin D. Childhood Cancer and Heredity. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:13-31.
    2. Aplan PD, Shern JF, Khan J. Molecular and genetic basis of childhood cancer. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:32-62.
    3. Zelley K, Lindell RB, Schiffman JD. et al. Genetic predisposition to cancer. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1270-1293.
    4. Garber JE, Offit K. Hereditary cancer predisposition syndromes. J Clin Oncol. 2005;23(2):276-292. doi:10.1200/JCO.2005.10.042.
    5. Holman JD, Dyer JA. Genodermatoses with malignant potential. Current Opin Pediatr. 2007;19(4):446-454. doi:10.1097/MOP.0b013e3282495939.
    6. Foulkes WD. Inherited susceptibility to common cancers. N Engl J Med. 2008;359:2143-2153. doi:10.1056/NEJMra0802968.
    7. Schiffman JD, Geller JI, Mundt E, et al. Update on pediatric cancer predisposition syndromes. Pediatr Blood Cancer. 2013;60(8):1247-1252. doi:10.1002/pbc.24555.
    8. American Academy of Pediatrics. Ethical and policy issues in genetic testing and screening of children. Pediatrics. 2013;131(3):620-622. doi:10.1542/peds.2012-3680.
    9. Monsalve J, Kapur J, Malkin D, Babyn PS. Imaging of cancer predisposition syndromes in children. Radiographics. 2011;31(1):263-280. doi:10.1148/rg.311105099.
    10. D’Orazio JA. Inherited cancer syndromes in children and young adults. J Pediatr Hematol Oncol. 2010;32(3):195-228. doi:10.1097/MPH.0b013e3181ced34c.
    11. Lu KH, Wodd ME, Daniels M, et al. American Society of Clinical Oncology Expert Statement: collection and use of a cancer family history for oncology providers. J Clin Oncol. 2014;32(8):833-841. doi:10.1200/JCO.2013.50.9257.
    12. Farid M, Ngeow J. Sarcomas associated with genetic cancer predisposition syndromes: a review. Oncologist. 2016;21(8):1002-1013. doi:10.1634/theoncologist.2016-0079.
    13. Tiwari R, Singh AK, Somwaru AS, et al. Radiologist’s primer on imaging of common hereditary cancer syndromes. RadioGraphics. 2019;39:759-778. doi:10.1148/rg.2019180171.
    14. Schneider K, Zelley K, Nichols KE, Garber J. Li-Fraumeni Syndrome. In: Adam MP, Ardinger HH, Pagon RA, et al., ed. GeneReviews® [Internet]. Seattle, WA: University of Washington, Seattle; 1993-2019. https://www.ncbi.nlm.nih.gov/books/NBK1311/.
    15. Villani A, Tabori U, Schiffman J, et al. Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: a prospective observational study. Lancet Oncol. 2011;12(6):559-567. doi:10.1016/S1470-2045(11)70119-X.
    16. Villani A, Shore A, Wasserman JD, et al. Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: 11 year follow-up of a prospective observational study. Lancet Oncol. 2016;17(9):1295-1305. doi:10.1016/S1470-2045(16)30249-2.
    17. Ballinger ML, Best A, Mai PL, et al. Baseline surveillance in Li-Fraumeni Syndrome using whole-body magnetic resonance imaging a meta analysis. JAMA Oncol. 2017;3(12):1634-1639. doi:10.1001/jamaoncol.2017.1968.
    18. Ruijs MWG, Loo CE, van Buchem CAJM, et al. Surveillance of Dutch patients with Li-Fraumeni Syndrome: the life-guard study. JAMA Oncol. 2017;3(12):1733-1734. doi:10.1001/jamaoncol.2017.1346.
    19. Mai PL, Khincha PP, Loud JT, et al. Prevalence of Cancer at Baseline screening in the National Cancer Institute Li-Fraumeni Syndrome Cohort. JAMA Oncol. 2017;3(12):1640-1645. doi:10.1001/jamaoncol.2017.1350.
    20. Kratz CP, Achatz MI, Brugiéres L, et al. Cancer screening recommendations for individuals with Li-Fraumeni Syndrome. Clin Cancer Res. 2017;23(11):e38-e45. doi:10.1158/1078-0432.CCR-17-0408.
    21. Greer MC, Voss SD, States LJ. Pediatric cancer predisposition imaging: focus on whole-body MRI. Clin Cancer Res. 2017;23(11):e6-e13. doi:10.1158/1078-0432.CCR-17-0515.
    22. Asdahl PH, Ojha RP, Hasle H. Cancer screening in Li-Fraumeni Syndrome. JAMA Oncol. 2017;3(12):1645-1646. doi:10.1001/jamaoncol.2017.2459.
    23. Eutsler EP, Khanna G. Whole-body magnetic resonance imaging in children: technique and clinical applications. Pediatr Radiol. 2016;46(6):858-872. doi: 10.1007/s00247-016-3586-y.
    24. Gottumukkalla RV, Gee MS, Hampilos PJ, Greer MC. Current and emerging roles of whole-body MRI in evaluation of pediatric cancer patients. RadioGraphics. 2019;39:516-534. doi:10.1148/rg.2019180130.
    25. Daly MB, Pilarski R, Berry MP, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 3.2019—January 18, 2019, Genetic/Familial High-Risk Assessment: Breast and Ovarian, available at: https://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Genetic/Familial High-Risk Assessment: Breast and Ovarian V3.2019 1/18/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org.
    26. Miller DT, Freedenberg D, Schorry E, et al. Health supervision for children with neurofibromatosis type 1. Pediatrics. 2019;143:e20190660. doi:10.1541/peds.2019-0660.
    27. Sahin M. Neurocutaneous syndromes. In: Kliegman RM, Stanton BF, St. Geme JW, Schor NF, Behrman RE, eds. Nelson Textbook of Pediatrics. Philadelphia, PA: Elsevier Saunders; 2011:2046-2048.
    28. Ullrich NJ. Neurocutaneous syndromes and brain tumors. J Child Neurol. 2016;31(12):1399-1411. doi:10.1177/0883073815604220.
    29. Korf BR and Bebin EM. Neurocutaneous disorders in Children. Pediatr Rev. 2017; 38(3):119-127. doi:10.1542/pir.2015-0118.
    30. Campian J, Gutmann DH. CNS tumors in neurofibromatosis. J Clin Oncol. 2017; 35(21):2378-2385. doi:10.1200/JCO.2016.71.7199.
    31. Friedman JM. Neurofibromatosis 1. In: Pagon RA, Adam MP, Ardinger HH et al., eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle;1998-2017. Version June 6, 2019. https://www.ncbi.nlm.nih.gov/books/NBK1109/.
    32. Karajannis MA, Ferner RE. Neurofibromatosis-related tumors: emerging biology and therapies. Curr Opin Pediatr. 2015;27(1):26-33. doi:10.1097/MOP.0000000000000169.
    33. Meany H, Dombi E, Reynolds J, et al. 18-Fluorodeoxyglucode-Positron Emission Tomography (FDG-PET) evaluation of nodular lesions in patients with neurofibromatosis Type 1 and Plexiform Neurofibromas (PN) or Malignant Peripheral Nerve Sheath Tumors (MPNST). Pediatr Blood Cancer. 2013;60(1):59-64. doi:10.1002/pbc.24212.
    34. Tsai LL, Druback L, Fahey F, et al. [18F]-Fluorodeoxyglucose positron emission tomography in children with neurofibromatosis type 1 and plexiform neurofibromas: correlation with malignant transformation. J Neuro Oncol. 2012;108(3):469-475. doi:10.1007/s11060-012-0840-5.
    35. Corbemale P, Valeyrie-Allanore L, Giammarile F, et al. Utility of 18F-FDG PET with a semi-quantitative index in the detection of sarcomatous transformation in patients with neurofibromatosis Type 1. PLoS ONE. 2014;9(2):e85954. doi:10.1371/journal.pone.0085954.
    36. Chirindel A, Chaudhry M, Blakeley JO, Wahl R. 18F-FDG PET/CT Qualitative and quantitative evaluation in neurofibromatosis Type 1 Patients for detection of malignant transformation: comparison of early to delayed imaging with and without liver activity normalization. J Nucl Med. 2015;56(3):379-385. doi:10.2967/jnumed.114.142372.
    37. Prada CE, Hufnagel RB, Hummel TR, et al. The use of magnetic resonance imaging screening for optic pathway gliomas in children with neurofibromatosis Type 1. J Pediatr. 2015;167(4):851-856. doi:10.1016/j.jpeds.2015.07.001.
    38. Blakeley JO, Evans DG, Adler J, et al. Consensus recommendations for current treatments and accelerating clinical trials for patients with neurofibromatosis Type 2. Am J Med Genet A. 2012;158A(1):24-41. doi:10.1002/ajmg.a.34359.
    39. Evans DG. Neurofibromatosis 2. In: Pagon RA, Adam MP, Bird TD et al., eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 2011. Version March 15, 2018. https://www.ncbi.nlm.nih.gov/books/NBK1201/.
    40. Evans DG. Neurofibromatosis type 2 (NF2): a clinical and molecular review. Orphanet J Rare Dis 2009;4:16. doi:10.1186/1750-1172-4-16.
    41. Ardern-Holmes S, Fisher G, North K. Neurofibromatosis Type 2: presentation, major complications, and management, with a focus on the pediatric age group. J Child Neurol. 2016;32(1):9-22. doi:10.1177/0883073816666736.
    42. Shuman C, Beckwith JB, Weksberg R. Beckwith-Wiedemann syndrome. In: Pagon RA, Adam MP, Bird TD et al., eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 2016. Version August 11, 2016. https://www.ncbi.nlm.nih.gov/pubmed/20301568.
    43. Choyke PL, Siegel MJ, Craft AW, Green DM, DeBaun MR. Screening for Wilms Tumor in children with Beckwith-Wiedemann syndrome or idiopathic hemihypertrophy. Med Pediatr Oncol. 1999;32(3):196-200. doi:10.1002/(SICI)1096-911X(199903)32:3<196::AID-MPO6>3.0.CO;2-9.
    44. Mussa A, Molinatto C, Baldassare G, et al. Cancer risk in Beckwith-Wiedemann syndrome: A systematic review and meta-analysis outlining a novel (epi)genotype specific histotype targeted screening protocol. J Pediatr. 2016 September;176:142-149. doi:10.1016/j.jpeds.2016.05.038.
    45. MacFarland SP, Mostoufi-Moab S, Zelley K, et al. Management of adrenal masses in patients with Beckwith-Wiedemann syndrome. Pediatr Blood Cancer. 2017;64(8):e26432. doi:10.1002/pbc.26432.
    46. Dome JS and Huff V. Wilms tumor predisposition. In: Pagon RA, Adam MP, Bird TD et al., eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 2016. https://www.ncbi.nlm.nih.gov/pubmed/20301471 Accessed January 3, 2018
    47. Fischbach BV, Trout KL, Lewis J, Luis CA, Sika M. WAGR Syndrome: A Clinical Review of 54 Cases. Pediatrics. 2005;116(4):984-988. doi:10.1542/peds.2004-0467.
    48. Provenzale D, Gupta S, Ahnen DJ, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 3.2017—October 10, 2017, Genetic/Familial High-Risk Assessment: Colorectal, available at: https://www.nccn.org/professionals/physician_gls/pdf/genetics_colon.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Genetic/Familial High-Risk Assessment: Colorectal V2.2019 8/8/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org
    49. Kennedy RD, Potter DD, Moir CR, Mounif E. The natural history of familial adenomatous polyposis syndrome: A 24 year review of a single center experience in screening, diagnosis, and outcomes, J Pediatr Surg 2014;49(1):82-86. doi:10.1016/j.jpedsurg.2013.09.033.
    50. Jasperson KW, Patel SG, Ahnen DJ. APC-Associated Polyposis Conditions. In: Pagon RA, Adam MP, Bird TD, et al., ed. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 2017. Version February 2, 2017. https://www.ncbi.nlm.nih.gov/books/NBK1345/.
    51. Stoffel EM, Mangu PB, Gruber SB, et al. Hereditary colorectal cancer syndromes: American Society of Clinical Oncology clinical practice guideline endorsement of the familial risk-colorectal cancer: European Society for Medical Oncology clinical practice guidelines. J Clin Oncol. 2015;33(2):209-217. doi:10.1200/JCO.2014.58.1322.
    52. Giusti F, Marini F, Brandi ML. Multiple endocrine neoplasia Type 1. In: Pagon RA, Adam MP, Ardinger HH et al, ed. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 1993-2019. Version December 14, 2017. https://www.ncbi.nlm.nih.gov/books/NBK1538/.
    53. Thakker RV, Newey PJ, Walls GV, et al. Clinical Practice guidelines for multiple endocrine neoplasia type 1 (MEN1). J Clin Endocrinol Metab. 2012;97(9):2990-3011. doi:10.1210/jc.2012-1230
    54. Giri D, McKay V, Weber A, Blair JC. Multiple endocrine neoplasia syndromes 1 and 2: manifestations and management in childhood and adolescence. Arch Dis Child. 2015;100(10):994-999. doi:10.1136/archdischild-2014-307028.
    55. Eng C. Multiple endocrine neoplasia Type 2. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 1999-2019. Version August 15, 2019. https://www.ncbi.nlm.nih.gov/books/NBK1257/.
    56. Northrup H, Koenig MK, Pearson DA, Au KS. Tuberous Sclerosis Complex. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; Version September 3, 2015. https://www.ncbi.nlm.nih.gov/pubmed/20301399.
    57. Northrup H, Koenig MK, Pearson DA, Au KS. Tuberous sclerosis complex. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle;1993-2019. Version July 12, 2018. https://www.ncbi.nlm.nih.gov/books/NBK1220/.
    58. Krueger DA, Care MM, Agricola K, Tudor C, Mays M, Franz DN. Everolimus long-term safety and efficacy in subependymal giant cell astrocytoma. Neurology. 2013;80(6):574-580. doi:10.1212/WNL.0b013e3182815428.
    59. Wheless JW, Klimo P. Subependymal giant cell astrocytomas in patients with tuberous sclerosis complex: considerations for surgical or pharmacotherapeutic intervention. J Child Neurol. 2014;29(11):1562-1571. doi:10.1177/0883073813501870.
    60. van Leeuwaarde RS, Ahmad S, Links TP, Giles RH. Von Hippel-Lindau Syndrome. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle;1993-2019. Version September 6, 2018. https://www.ncbi.nlm.nih.gov/books/NBK1463/.
    61. Nielsen SM, Rhodes L, Blanco I, et al. Von Hippel-lindau Disease: genetics and role of genetic counseling in a Multiple Neoplasia Syndrome. J Clin Oncol. 2016;34(18):2172-2181.
    62. Rednam SP, Erez A, Druker H, et al. Von Hippel-Lindau and hereditary pheochromocytoma/paraganglioma syndromes: clinical features, genetics, and surveillance recommendations in childhood. Clin Cancer Res. 2017;23:e68-e75. doi: 10.1158/1078-0432.CCR-17-0547.
    63. Kordes U, Bartelheim K, Modena P, et al. Favorable outcome of patients affected by rhabdoid tumors due to Rhabdoid Tumor Predisposition Syndrome (RTPS). Pediatr Blood Cancer. 2014;61(5):919-921. doi:10.1002/pbc.24793.
    64. Sredni ST, Tomita T. Rhabdoid Tumor Predisposition Syndrome. Pediatr Dev Pathol. 2015;18(1):49-58. doi:10.2350/14-07-1531-MISC.1.
    65. Nemes K, Bens S, Bourdeaut F, et al. Rhabdoid tumor predisposition syndrome. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 1993-2019. Version December 7, 2017. https://www.ncbi.nlm.nih.gov/books/NBK469816/.
    66. Lohmann DR, Gallie BL. Retinoblastoma. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 1993-2019. Version November 21, 2018. https://www.ncbi.nlm.nih.gov/books/NBK1452/.
    67. Kirmani S, Young WF. Hereditary Paraganglioma-Pheochromocytoma Syndromes. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 2008-2014. Version November 6, 2014. https://www.ncbi.nlm.nih.gov/pubmed/20301715.
    68. Gripp KW, Lin AE. Costello Syndrome. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 1999-2019. Version January 12, 2012. https://www.ncbi.nlm.nih.gov/books/NBK1507/.
    69. Tabori U, Hansford JR, Achatz MI, et al. Clinical management and tumor surveillance recommendations of inherited mismatch repair deficiency in childhood. Clin Cancer Res. 2017;23:e32-e37. doi: 10.1158/1078-0432.CCR-17-0574.


    PEDONC-3: Pediatric Leukemias

    PEDONC-3.1: Pediatric Leukemia General Considerations
    PEDONC-3.2: Acute Lymphoblastic Leukemia (ALL)
    PEDONC-3.3: Acute Myeloid Leukemia (AML)

    PEDONC-3.1: Pediatric Leukemia General Considerations

    For this condition imaging is medically necessary based on the following criteria:

    The overwhelming majority of leukemias occurring in children are acute. Chronic myelogenous leukemia (CML) is rare in children, and the occurrence of chronic lymphocytic leukemia (CLL) appears to have only been reported once in pediatric members to date.

    MRI Brain without and with contrast (CPT® 70553) can be performed in members exhibiting CNS symptoms and in members found to have high tumor burden on CSF cytology.

    There is not sufficient evidence to support the use of PET imaging for any indication in the management of acute lymphoblastic leukemia, acute myeloid leukemia, or chronic myeloid leukemia.

    Routine advanced imaging is not indicated in the evaluation and management of chronic myeloid leukemia in the absence of specific localizing clinical symptoms or clearance for hematopoietic stem cell transplantation. See Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-29: Hematopoietic Stem Cell Transplantation for imaging guidelines related to transplant.



    PEDONC-3.2: Acute Lymphoblastic Leukemia (ALL)

    For this condition imaging is medically necessary based on the following criteria:

    The majority of ALL members have B-precursor ALL and routine advanced imaging is not necessary.

    Members with B-precursor or T-cell lymphoblastic lymphoma without bone marrow involvement are treated similarly to leukemia members of the same cell type and should be imaged according to this guideline section.

    This section does not apply to members with mature B-cell histology (primarily Burkitt’s in children). Please refer to PEDONC-5.3: Pediatric Aggressive Mature B-Cell Non-Hodgkin Lymphomas (NHL) for guidelines for these members.

    CXR should be performed to evaluate for mediastinal mass in suspected cases or upon initial diagnosis.

      ® If mediastinal widening is seen on CXR, CT Chest with contrast (CPT® 71260) is indicated immediately to evaluate for airway compression and anesthesia safety prior to attempting histologic diagnosis.
      ® Members with known or strongly suspected T-cell histology or other suspected lymphoblastic lymphoma involvement can have either of the following approved for initial staging purposes:
        ¡ CT Neck (CPT® 70491), CT Chest (CPT® 71260) and CT Abdomen/Pelvis (CPT® 74177) with contrast OR
        ¡ PET/CT (CPT® 78815 or 78816)
    MRI Brain without and with contrast (CPT® 70553) can be performed in members exhibiting CNS symptoms and in members found to have high tumor burden on CSF cytology.

    Additional imaging in lymphoblastic lymphoma:

    Follow up CT to assess response to therapy is indicated only for members with known bulky nodal disease (usually with T-cell histology) at the end of induction (~4 to 6 weeks). Members with residual masses can be evaluated with every new therapy phase (Consolidation, Interim maintenance, etc., generally every 8 to 12 weeks) until disease resolution is seen.

      ® PET/CT (CPT® 78815) can be approved when residual mass ≥ 8 mm in diameter is present on recent CT imaging and there is documentation of how PET findings will affect immediate treatment decision making. These requests should be forwarded for Medical Director review.

    Once CT imaging shows no evidence of disease, further surveillance should use CXR or Abdominal Ultrasound (CPT® 76700) only, as indicated by site(s) of bulky disease present at diagnosis.
      ® Members with persistent residual masses can have CT of all involved bulky nodal areas performed as part of an end of therapy evaluation

    Immunosuppression during ALL therapy and imaging ramifications:

    ALL members are severely immunocompromised during the first 4 to 6 weeks of treatment (induction) and any conventional imaging request to evaluate for infectious complications during this time frame should be approved immediately.

    CT or MRI imaging requests for infectious disease concerns for ALL members with absolute neutrophil count (ANC) < 500 or inconclusive findings on chest x-ray or US at any ANC during active treatment should be approved as requested.

    Additionally, members may have therapy-induced hypogammaglobulinemia which requires supplemental intravenous immune globulin (IVIG) during maintenance therapy. Members receiving supplemental IVIG should be treated similarly to members with ANC < 500 with regards to imaging for infectious disease.

    Imaging during therapy for relapsed ALL:

    Relapsed ALL members are treated with very intensive chemotherapy regimens and most members spend the majority of their chemotherapy treatment phase in the hospital. Due to the high risk of invasive infections, frequent CT or MRI imaging may be indicated to evaluate known or suspected new sites of invasive fungal or other aggressive infections, and in general these should be approved as requested.

      ® Surveillance imaging of asymptomatic members to detect invasive fungal infection has not been shown to impact member outcomes. Imaging requests in these circumstances should only be approved when acute clinical decisions will be made based on the imaging.

    Imaging of known or suspected osteonecrosis in ALL:

    Osteonecrosis (ON) in ALL members is a relatively common complication of ALL and its treatment, primary corticosteroids. Approximately 3% of younger children and 12 to 15% of adolescents are affected by ON at some point during therapy. The peak incidence occurs approximately one year from the time of diagnosis.

      ® For members with symptoms suggesting osteonecrosis, MRI without contrast or without and with contrast of the affected joint(s) can be approved.
        ¡ CT without contrast can be approved when MRI is contraindicated or unavailable, or for diagnosis of suspected subchondral fracture
      ® Screening MRI of asymptomatic members age ≤ 10 years to detect osteonecrosis has not been shown to impact member outcomes, and it is not standard to alter treatment based on imaging findings alone without symptoms
        ¡ A single screening MRI Bilateral Hips (CPT® 73721 or CPT® 73723 with modifier -50) can be approved 6 to 9 months after diagnosis for members age ≥11 years
      ® If osteonecrosis is detected on initial MRI, corticosteroids are often withheld during maintenance chemotherapy (but continued in earlier phases of therapy).
      ® In members whose symptoms have resolved and are still receiving active treatment, repeat MRI without contrast of the affected joint(s) can be approved every 2 cycles of maintenance (~every 6 months) if reintroduction of corticosteroids is being considered.
      ® MRI without contrast of the affected joint(s) can be approved if requested for preoperative planning in members undergoing core decompression
      ® See PEDONC-19.4: Osteonecrosis In Long Term Cancer Survivors for information on osteonecrosis in ALL members who have completed therapy
    PEDONC-3.3: Acute Myeloid Leukemia (AML)

    For this condition imaging is medically necessary based on the following criteria:

    The majority of AML members do not have any bulky disease and routine advanced imaging is not necessary.

    Advanced imaging may be indicated for rare members with bulky tumor masses (commonly referred to as chloromas, leukemic sarcomas, or myeloid sarcomas) noted on physical examination or other imaging such as plain film or ultrasound.

    AML members are treated with very intensive chemotherapy regimens and spend the majority of their chemotherapy treatment phase in the hospital. Due to the high risk of invasive infections, frequent CT or MRI imaging may be indicated to evaluate known sites of invasive fungal infection, and in general these should be approved as requested.

      ® Surveillance imaging of asymptomatic members to detect invasive fungal infection has not been shown to impact member outcomes. Imaging requests in these circumstances should only be approved when acute clinical decisions will be made based on the imaging.

    References

    1. Rabin KR, Gramatges MM, Margolin JF, et al. Acute Lymphoblastic Leukemia. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:463-497.
    2. Arceci RJ, Meshinchi S. Acute Myeloid Leukemia and Myelodysplastic disorders. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:498-544.
    3. Rau R, Loh ML. Myeloproliferative neoplasms of childhood. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:545-567.
    4. Gutierrez A, Silverman LB. Acute Lymphoblastic Leukemia. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1527-1554.
    5. Berman JN, Look AT. Pediatric Myeloid Leukemia, Myelodysplasia, and Myeloproliferative Disease. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1555-1613.
    6. Kobos R, Shukla N, and Armstrong SA. Infant Leukemias. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1614-1625.
    7. Kaplan JA. Leukemia in children. Pediatr Rev. 2019;40:319-331. doi: 10.1542/pir.2018-0192.
    8. Brown P, Inaba H, Annesley C, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 1.2020—May 30, 2019, Pediatric Acute Lymphoblastic Leukemia, available at: https://www.nccn.org/professionals/physician_gls/pdf/ped_all.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Pediatric Acute Lymphoblastic Leukemia V1.2020 5/30/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org.
    9. Ranta S, Palomäki M, Levinsen M, et al. Role of neuroimaging in children with Acute Lymphoblastic Leukemia and central nervous system involvement at diagnosis. Pediatr Blood Cancer. 2016;64:64-70. doi:10.1002/pbc.26182/epdf.
    10. Agrawal AK, Saini N, Gildengorin G et al. Is routine computed tomographic scanning justified in the first week of persistent febrile neutropenia in children with malignancies? Pediatr Blood Cancer. 2011;57(4):620-624. doi:10.1002/pbc.22974/epdf.
    11. Pui C-H, Yang JJ, Hunger SP, et al. Childhood Acute Lymphoblastic Leukemia: progress through collaboration. J Clin Oncol. 2015;33(27):2938-2948. doi: 10.1200/JCO.2014.59.1636.
    12. Kawedia JD, Kaste SC, Pei D, et al. Pharmacokinetic, pharmacodynamic, and pharmacogenetic determinants of osteonecrosis in children with acute lymphoblastic leukemia. Blood Journal. 2011;117(8):2340-2347. doi:10.1182/blood-2010-10-311969.
    13. Marcucci G, Beltrami G, Tamburini A, et al. Bone health in childhood cancer: review of the literature and recommendations for the management of bone health in childhood cancer survivors. Ann Oncol 2019;30:908-920. doi:10.1093/annonc/mdz120.
    14. Vora A. Management of osteonecrosis in children and young adults with acute lymphoblastic leukaemia. Br J Haematol. 2011;155(5):549-560. doi:10.1111/j.1365-2141.2011.08871.x.
    15. Kaste SC, Pei D, Cheng C, et al. Utility of early screening magnetic resonance imaging for extensive hip osteonecrosis in pediatric patients treated with glucocorticoids. J Clin Oncol. 2015;33(6):610-615. doi:10.1200/JCO.2014.57.5480.
    16. Niinimäki T, Harila-Saari A, Niinimäki R. The diagnosis and classification of osteonecrosis in patients with childhood Leukemia. Pediatr Blood Cancer. 2015;62(2):198-203. doi:10.1002/pbc.25295.
    17. Murphey MD, Roberts CC, Bencardino JT, et al. Osteonecrosis of the hip. ACR Appropriateness Criteria® 2015;1-12. https://acsearch.acr.org/docs/69420/Narrative/.
    18. Karol SE, Mattano LA, Yang W, et al. Genetic risk factors for the development of osteonecrosis in children under age 10 treated for acute lymphoblastic leukemia. Blood Journal. 2016;127(5):558-564. doi:10.1182/blood-2015-10-673848.
    19. Chavhan GB, Babyn PS, Nathan PC, et al. Imaging of acute and subacute toxicities of cancer therapy in children. Pediatr Radiol. 2016;46(1):9-20. doi:10.1007/s00247-015-3454-1.


    PEDONC-4: Pediatric CNS Tumors

    PEDONC-4.1: Pediatric CNS Tumors General Considerations
    PEDONC-4.2: Intracranial Low Grade Gliomas (LGG)
    PEDONC-4.3: High Grade Gliomas (HGG)
    PEDONC-4.4: Medulloblastoma (MDB), Supratentorial Primitive Neuroectodermal Tumors (sPNET), and Pineoblastoma
    PEDONC-4.5: Atypical Teratoid/Rhabdoid Tumors (ATRT)
    PEDONC-4.6: Pineocytomas
    PEDONC-4.7: CNS Germinomas and Non-Germinomatous Germ Cell Tumors (NGGCT)
    PEDONC-4.8: Ependymoma
    PEDONC-4.9: Malignant Tumors of the Spinal Cord
    PEDONC-4.10: Craniopharyngioma and Other Hypothalamic/Pituitary Region Tumors
    PEDONC-4.11: Primary CNS Lymphoma
    PEDONC-4.12: Meningiomas
    PEDONC-4.13: Choroid Plexus Tumors
    PEDONC-4.1: Pediatric CNS Tumors General Considerations

    For this condition imaging is medically necessary based on the following criteria:

    Central nervous system tumors are the second most common form of childhood cancer, accounting for ~20% of all pediatric malignancies.
    Red flag symptoms raising suspicion for CNS tumors include:
    Any headache complaint from a child age ≤ 5 years
    Headaches awakening from sleep
    Focal findings on neurologic exam
    Clumsiness (common description of gait or coordination problems in young children)
    Headaches associated with morning nausea/vomiting
    New onset of seizure activity with focal features
    Papilledema on physical exam
    MRI is the preferred imaging modality for all pediatric CNS tumors. The primary imaging study for pediatric brain tumors is MRI Brain without and with contrast (CPT® 70553).

      ® For children able to undergo MRI without sedation, MRI Brain without contrast (CPT® 70551) can be approved if requested for initial evaluation of suspected CNS tumor.
      ® Younger members requiring sedation for MRI should have their initial MRI performed without and with contrast in order to avoid a second anesthesia exposure.

    CT can be approved for evaluation of ventriculomegaly or other operative considerations, or for children who cannot undergo MRI safely.
      ® Because of the significant percentage of pediatric CNS tumors occurring in the posterior fossa, CT is not a recommended study for evaluation of pediatric headache when brain tumor is clinically suspected because of its limited diagnostic accuracy in this area. MRI should be used as first line imaging in these cases.
      ® CT should not be used in place of MRI to avoid sedation in young children when red flag symptoms for CNS tumors are present
      ® CT can also be approved for evaluation of headaches related to head trauma or evaluation of skull or facial bone abnormalities

    MRA or CTA are not routinely indicated in pediatric CNS tumors but can be approved for preoperative planning or to clarify inconclusive findings on MRI or CT.

    Definitive imaging should be completed prior to considering biopsy given the high degree of morbidity associated with operating on the CNS

      ® Occasionally biopsy is not necessary because the imaging findings provide a definitive diagnosis. Examples include diffuse intrinsic pontine glioma and optic pathway gliomas in a member with known neurofibromatosis.

    Perioperative imaging frequency
      ® Children may undergo very frequent imaging in the immediate perioperative period around resection or debulking of a CNS tumor due to the small anatomic spaces involved. Requests for imaging during this time period to specifically evaluate postoperative course or ventriculoperitoneal shunt functioning should, in general, be approved as requested.
      ® A one-time MRI Brain without and with contrast (CPT® 70553) can be approved in the immediate preoperative period (even if another study has already been completed) to gain additional information which can be important in optimizing member outcomes, such as:
        ¡ Completion of additional specialized MRI sequences such as diffusion-tensor imaging, perfusion imaging, tractography, or other sequences not reported under a separate CPT® code but not part of a routine MRI Brain series
      ® Repeat MRI Brain that is being requested solely for loading into operative navigation software should not be requested as a diagnostic code, but can be approved under a treatment planning code (CPT® 76498). These requests should be forwarded for Medical Director review.

    MR Spectroscopy (MRS, CPT® 76390):

    Note: Some payors have specific restrictions on MR Spectroscopy, and those coverage policies may supersede the recommendations for MRS in these guidelines.

    MRS is only supported for use in brain tumors of specified histologies where diagnostic accuracy has been established in peer-reviewed literature

      ® See diagnosis-specific guidelines for MRS indications

    MRS is considered investigational for all other histologies and indications not listed in a diagnosis-specific guideline section

    MR spectroscopy is not indicated for routine surveillance

    Requests for MRS should be forwarded for Medical Director review

    PET Brain Imaging (CPT® 78608 and CPT® 78609):

    Note: Some payors have specific restrictions on PET Brain Metabolic imaging, and those coverage policies may supersede the recommendations for this study in these guidelines.

    PET Brain Metabolic imaging (CPT® 78608) is only supported for use in brain tumors of specified histologies where diagnostic accuracy has been established in peer-reviewed literature

      ® See diagnosis-specific guidelines for PET indications

    PET Brain Metabolic imaging is considered investigational for all other histologies and indications not listed in a diagnosis-specific guideline section

    PET Brain Perfusion imaging (CPT® 78609) is not indicated in the evaluation or management of primary CNS tumors

    Fusion PET/CT studies (CPT® 78814, CPT® 78815, or CPT® 78816) are not indicated in the evaluation or management of primary CNS tumors

    PET Brain Metabolic is not indicated for routine surveillance

    Requests for PET Brain Metabolic should be forwarded for Medical Director review



    PEDONC-4.2: Intracranial Low Grade Gliomas (LGG)

    For this condition imaging is medically necessary based on the following criteria:

    Account for 40 to 60% of pediatric CNS tumors. These tumors are defined as having a WHO histologic grade of I or II (out of IV), can occur anywhere in the CNS, and includes the following tumors:


      ® Pilocytic Astrocytoma
      ® Fibrillary (or Diffuse) Astrocytoma
      ® Optic Pathway Gliomas
      ® Pilomyxoid Astrocytoma
      ® Oligodendroglioma
      ® Oligoastrocytoma
      ® Oligodendrocytoma
      ® Subependymal Giant Cell Astrocytoma (SEGA)
      ® Ganglioglioma
      ® Gangliocytoma
      ® Dysembryoplastic Infantile Astrocytoma (DIA)
      ® Dysembryoplastic Infantile Ganglioglioma (DIG)
      ® Dysembryoplastic Neuroepithelial Tumor (DNT)
      ® Tectal Plate Gliomas
      ® Cervicomedullary Gliomas
      ® Pleomorphic Xanthoastrocytoma (PXA)
      ® Any other glial tumor with a WHO grade of I or II

    PET Brain Metabolic imaging (CPT® 78608) can be approved in the following circumstances:
      ® To determine need for biopsy when transformation to high grade glioma is suspected based on clinical symptoms or recent MRI findings
      ® To evaluate a brain lesion of indeterminate nature when the PET findings will be used to determine whether biopsy/resection can be safely postponed

    MR spectroscopy (MRS, CPT® 76390) can be approved in the following circumstances:
      ® To distinguish low grade from high grade gliomas
      ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed
      ® To distinguish radiation-induced tumor necrosis from progressive disease within 18 months of completing radiotherapy.

    Note: Some payors have specific restrictions on PET Brain Metabolic imaging and/or MR Spectroscopy, and those coverage policies may supersede the recommendations for PET Brain or MRS in these guidelines

    Low Grade Gliomas Initial Staging:

    MRI Brain without and with contrast (CPT® 70553) is indicated for all LGG

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved for all LGG members if requested, and spinal imaging is particularly recommended for members with:

      ® Multicentric tumors
      ® Intracranial leptomeningeal disease
      ® Clinical signs or symptoms suggesting spinal cord involvement
      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain.

    Members with neurofibromatosis and small optic pathway tumors may not undergo biopsy or resection and will proceed directly to treatment or surveillance.

    Low Grade Gliomas Treatment Response:

    Children who have resection of the tumor can have a single MRI Brain without and with contrast (CPT® 70553) approved following resection to establish baseline imaging and those with a complete resection should then be imaged according to surveillance guidelines

    Children with neurofibromatosis and small optic pathway gliomas may be observed without specific treatment and should be imaged according to surveillance guidelines for LGG.

    Members age > 10 years with incompletely resected tumors usually receive adjuvant radiation therapy and can have a single MRI Brain without and with contrast (CPT® 70553) approved at completion of radiotherapy and should then be imaged according to surveillance guidelines

    Members age ≤ 10 years with incompletely resected tumors are commonly treated with chemotherapy and can have MRI Brain without and with contrast (CPT® 70553) approved every 2 cycles during active treatment and at the end of planned chemotherapy

    Spinal imaging is not indicated during treatment response for members without evidence of spinal cord involvement at initial diagnosis

    Spinal imaging is appropriate every 2 cycles during induction chemotherapy for members with measurable spinal cord disease on MRI

    Low Grade Gliomas Surveillance:

    MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for 2 years, then every 6 months for 3 years, then annually thereafter.

      ® MRI Orbits without and with contrast (CPT® 70543) can be approved for members with optic pathway glioma and either a history of intra-orbital involvement or a history of NF1

    MRI Spine is not indicated during surveillance in members without prior history of spinal involvement except to evaluate symptoms suspicious for spinal cord recurrence

    For members with cord involvement at diagnosis, MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved after completion of therapy every 3 months for 2 years, then every 6 months for 3 years, then annually thereafter.

      ® MRI Spine with contrast only can be approved (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) if being performed immediately following a contrast-enhanced MRI Brain

    MR Spectroscopy and PET Brain Metabolic are not indicated for routine surveillance

    PEDONC-4.3: High Grade Gliomas (HGG)

    For this condition imaging is medically necessary based on the following criteria:

    Rare in children compared with the adult population, but represent 10 to 20% of pediatric CNS tumors. Prognosis is very poor, and survival significantly beyond 3 years from diagnosis is rare, even with complete surgical resection at initial diagnosis.

    These tumors are defined as having a WHO histologic grade of III or IV (out of IV) can occur anywhere in the CNS (though the majority occur in the brain), and includes the following tumors:


      ® Anaplastic astrocytoma
      ® Glioblastoma multiforme
      ® Diffuse intrinsic pontine glioma (DIPG, or “Brainstem glioma”)
      ® Gliomatosis cerebri
      ® Gliosarcoma
      ® Anaplastic oligodendroglioma
      ® Anaplastic ganglioglioma
      ® Anaplastic mixed glioma
      ® Anaplastic mixed ganglioneuronal tumors
      ® Any other glial tumor with a WHO grade of III or IV

    PET Brain Metabolic Imaging (CPT® 78608) can be approved in the following circumstances:
      ® To distinguish radiation-induced tumor necrosis from progressive disease within 18 months of completing radiotherapy
      ® To evaluate inconclusive MRI findings when the PET findings will be used to determine need for biopsy or change in therapy, including a change from active therapy to surveillance
      ® To evaluate a brain lesion of indeterminate nature when the PET findings will be used to determine whether biopsy/resection can be safely postponed
      ® PET Brain is not indicated in gliomas occurring in the brain stem due to poor uptake and lack of impact on member outcomes

    MR Spectroscopy (MRS, CPT® 76390) can be approved in the following circumstances:
      ® To distinguish low grade from high grade gliomas
      ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed
      ® To distinguish radiation-induced tumor necrosis from progressive disease within 18 months of completing radiotherapy.

    Note: Some payors have specific restrictions on MR Spectroscopy, and those coverage policies may supersede the recommendations for MRS in these guidelines

    High Grade Gliomas Initial Staging:

    MRI Brain without and with contrast (CPT® 70553) is indicated for all HGG

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved for all HGG members if requested, and spinal imaging is particularly recommended for members with:

      ® Multicentric tumors
      ® Intracranial leptomeningeal disease
      ® Clinical signs or symptoms suggesting spinal cord involvement
      ® MRI spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    High Grade Gliomas Treatment Response:

    Members who have resection of the tumor can have a single MRI Brain without and with contrast (CPT® 70553) approved following resection to establish baseline imaging and those with a complete resection should then be imaged according to surveillance guidelines

    If receiving adjuvant radiotherapy after a completely resected tumor, an additional MRI Brain without and with contrast (CPT® 70553) can be approved at the end of radiotherapy

    Members with incompletely resected tumors are commonly treated with chemotherapy and can have MRI Brain without and with contrast (CPT® 70553) approved every 2 cycles during active treatment and at the end of planned chemotherapy

    Spinal imaging is not indicated during treatment response for members without evidence of spinal cord involvement at initial diagnosis

    Spinal imaging is appropriate every 2 cycles during induction chemotherapy for members with measurable spinal cord disease on MRI

    High Grade Gliomas Surveillance:

    MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for 3 years, then every 6 months thereafter

    MRI Spine is not indicated during surveillance in members without prior history of spinal involvement except to evaluate symptoms suspicious for spinal cord recurrence

    For members with cord involvement at diagnosis, MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved after completion of therapy every 3 months for 3 years, then every 6 months thereafter

      ® MRI Spine can be performed with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) if being performed immediately following a contrast-enhanced MRI Brain

    MR Spectroscopy and PET Brain Metabolic are not indicated for routine surveillance


    PEDONC-4.4: Medulloblastoma (MDB), Supratentorial Primitive Neuroectodermal Tumors (sPNET), and Pineoblastoma

    For this condition imaging is medically necessary based on the following criteria:

    Account for 15 to 25% of pediatric CNS tumors, prognosis is generally favorable. Leptomeningeal spread is common and can occur after initial diagnosis.
    Includes the following tumors:
    Medulloblastoma and Pineoblastoma
    sPNET
      ® Medulloepithelioma
      ® Cerebral or cerebellar neuroblastoma
      ® Cerebral or cerebellar ganglioneuroblastoma
      ® Ependymoblastoma
    Risk assessment is important in determining optimal treatment
    High risk features include the following:
    Spinal metastasis (including cytology positive only)
    Multifocal intracranial tumors
    Anaplastic histology
    All sPNET and pineoblastomas
    > 1.5 cm2 residual tumor area on postoperative MRI and age < 3 years
    Members without any high risk features are considered “average risk”
    PET Brain Metabolic Imaging (CPT® 78608) can be approved in the following circumstances:

      ® To distinguish radiation-induced tumor necrosis from progressive disease within 18 months of completing radiotherapy
      ® To evaluate inconclusive MRI findings when the PET findings will be used to determine need for biopsy or change in therapy, including a change from active therapy to surveillance
      ® To evaluate a Brain lesion of indeterminate nature when the PET findings will be used to determine whether biopsy/resection can be safely postponed

    MR Spectroscopy (CPT® 76390) can be approved in the following circumstances:
      ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed

    Medulloblastoma, sPNET, Pineoblastoma Initial Staging:

    Preoperative MRI Brain without and with contrast (CPT® 70553) is indicated for all members

    Postoperative MRI Brain without and with contrast (CPT® 70553) is required (preferably within 48 hours of surgery) to quantify residual tumor volume

    MRI spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is required for all members either preoperatively or within 28 days postoperatively

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    Medulloblastoma, sPNET, Pineoblastoma Treatment Response:

    Members generally proceed to chemoradiotherapy within 31 days of surgical resection. All members receive adjuvant chemotherapy lasting 6 to 12 months that begins ~6 weeks after completion of chemoradiotherapy.

    MRI Brain without and with contrast (CPT® 70553) and MRI spine without and with contrast (Cervical-CPT® 72156, Thoracic CPT® 72157, Lumbar-CPT® 72158) is appropriate at the start of adjuvant chemotherapy and every 2 cycles until therapy is completed

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
      ® Children age < 3 years are often treated with multiple cycles of high dose chemotherapy with autologous stem cell rescue in lieu of radiotherapy, and disease evaluations may occur prior to each cycle (every 4 to 6 weeks) if needed for response determination.

    End of treatment evaluation should include MRI Brain without and with contrast (CPT® 70553) and MRI Spine (with or without and with contrast)

    Medulloblastoma, sPNET, Pineoblastoma Surveillance:

    MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for 2 years, then every 6 months for 3 years, then annually for 10 years

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved after completion of therapy every 3 months for 2 years, then every 6 months for 3 years, then annually for 10 years

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    MR Spectroscopy and PET Brain Metabolic are not indicated for routine surveillance

    PEDONC-4.5: Atypical Teratoid/Rhabdoid Tumors (ATRT)

    For this condition imaging is medically necessary based on the following criteria:

    Highly aggressive tumor occurring primarily in very young children that has a clinical presentation very similar to medulloblastoma with a much higher rate of leptomeningeal spread. Metastases can occur outside the CNS, and associated tumors can also arise in the kidneys (Malignant Rhabdoid Tumor of the Kidney, MRT). Rhabdoid malignancies occurring outside the CNS should be imaged according to PEDONC-7.6: Malignant Rhabdoid Tumor of the Kidney (MRT) and Other Extracranial Sites.

    Overall prognosis is poor, with < 20% of members surviving beyond 2 years from diagnosis.


    Atypical Teratoid/Rhabdoid Tumor Initial Staging:

    Preoperative MRI Brain without and with contrast (CPT® 70553) is indicated for all members

    Postoperative MRI Brain without and with contrast (CPT® 70553) is required (preferably within 48 hours of surgery) to quantify residual tumor volume

    MRI Spine without and with contrast (Cervical-CPT®72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is required for all members either preoperatively or within 28 days postoperatively

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    Renal US (CPT® 76770) is indicated to evaluate for renal masses at initial diagnosis
      ® CT Abdomen/Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be approved if a renal lesion is detected on US.
      ® If a renal lesion is also present, imaging guidelines for MRT should be followed
        (See: PEDONC-7.6: Malignant Rhabdoid Tumor of the Kidney (MRT) and Other Extracranial Sites)
    PET Brain Metabolic does not have a defined role in the evaluation of ATRT at this time

    MR Spectroscopy (CPT® 76390) can be approved in the following circumstances:

      ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed

    Atypical Teratoid/Rhabdoid Tumor Treatment Response:

    Members generally proceed to induction chemotherapy shortly following surgical resection or biopsy.

    MRI Brain without and with contrast (CPT® 70553) and MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate after every 2 cycles of induction chemotherapy

      ® MRI spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
      ® Children with ATRT are often treated using consolidation chemotherapy with 2 to 4 cycles of high dose chemotherapy with autologous stem cell rescue. Disease evaluation is indicated following the end of the planned stem cell rescues but may occur prior to each cycle (every 4 to 6 weeks) if needed for response determination.

    Following completion of chemotherapy some members will proceed to radiotherapy. MRI performed at the end of consolidation therapy should serve as the diagnostic MRI prior to radiotherapy.

    MRI Brain without and with contrast (CPT® 70553) and MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate at the end of all planned therapy

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain.

    Atypical Teratoid/Rhabdoid Tumor Surveillance:

    MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for 2 years, then every 6 months for 3 years, then annually for 10 years

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved after completion of therapy every 3 months for 2 years, then every 6 months for 3 years, then annually for 10 years

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain.

    MR Spectroscopy is not indicated for routine surveillance



    PEDONC-4.6: Pineocytomas

    For this condition imaging is medically necessary based on the following criteria:

    Low grade malignancy that is similar in presentation to low grade glioma (LGG).

    PET Brain Metabolic imaging and MR Spectroscopy do not have a defined role in the evaluation of pineocytoma.

    Pineocytomas Initial Staging:

    MRI Brain without and with contrast (CPT® 70553) is indicated for all members

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved for members with:

      ® Multicentric tumors
      ® Atypical histology including pineoblastoma-like elements
      ® Clinical signs or symptoms suggesting spinal cord involvement
      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    Pineocytomas Treatment Response:

    Surgical resection is curative for most members. Members who have resection of the tumor can have a single MRI Brain without and with contrast (CPT® 70553) approved following resection to establish baseline imaging and those with a complete resection should then be imaged according to surveillance guidelines

    Members with incompletely resected tumors may receive adjuvant radiation therapy and can have a single MRI Brain without and with contrast (CPT® 70553) approved at completion of radiotherapy and should then be imaged according to surveillance guidelines

      ® Spinal imaging is not indicated for members without evidence of spinal cord involvement at initial diagnosis
      ® Spinal imaging is appropriate at completion of radiotherapy for members with measurable spinal cord disease on MRI

    Pineocytomas Surveillance:

    MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually thereafter

    MRI Spine is not indicated during surveillance in members without prior history of spinal involvement except to evaluate symptoms suspicious for spinal cord recurrence

    For members with cord involvement at diagnosis, MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved after completion of therapy every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually thereafter

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    PEDONC-4.7: CNS Germinomas and Non-Germinomatous Germ Cell Tumors (NGGCT)

    For this condition imaging is medically necessary based on the following criteria:

    More common in older school age children and younger adolescents, but can occur throughout the pediatric age range. Although leptomeningeal spread is common, prognosis is excellent due to high sensitivity to chemotherapy and radiotherapy.
    Includes the following tumors:
    CNS Germinoma
    Non-Germinomatous Germ Cell Tumors (NGGCT)
      ® Embryonal carcinoma
      ® Yolk sac tumor
      ® Choriocarcinoma
      ® Teratoma
      ® Mixed germ cell tumor
    PET Metabolic Brain imaging does not have a defined role in the evaluation of CNS GCT.

    MR Spectroscopy (CPT® 76390) can be approved in the following circumstances:

      ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed

    CNS Germinoma & NGGCT Initial Staging:

    MRI Brain without and with contrast (CPT® 70553) is indicated for all members

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is indicated for all members

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    CNS Germinoma & NGGCT Treatment Response:

    Members generally proceed to chemotherapy shortly following surgical resection or biopsy and will usually receive 2 to 4 cycles.

    MRI Brain without and with contrast (CPT® 70553) is appropriate after every 2 cycles of induction chemotherapy

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate at the end of induction chemotherapy for members with localized intracranial tumors

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
      ® Spinal imaging is appropriate every 2 cycles during induction chemotherapy for members with measurable spinal cord disease on MRI

    Following completion of chemotherapy, members with residual disease will proceed to second-look surgery and/or radiotherapy
      ® MRI of all known sites of measurable disease can be performed prior to surgery and prior to radiotherapy, if necessary

    MRI Brain without and with contrast (CPT® 70553) and MRI Spine (with or without and with contrast) is appropriate at the end of all planned therapy

    CNS Germinoma & NGGCT Surveillance:

    MRI Brain without and with contrast (CPT® 70553) can be approved every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually until 5 years after completion of therapy

      ® For additional imaging guidelines for members in long term follow up after CNS tumor treatment that included radiation therapy, See PEDONC-19.3: Second Malignant Neoplasms (SMN)

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually until 5 years after completion of therapy
      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
    Members with new or worsening neurologic symptoms (including worsening of diabetes insipdus:
      ® MRI Brain without and with contrast (CPT® 70553)
      ® MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158)
        ¡ MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
    PEDONC-4.8: Ependymoma

    For this condition imaging is medically necessary based on the following criteria:

    Occur primarily intracranially, roughly 2/3 in the posterior fossa. Overall prognosis is very good, with supratentorial tumors faring better. Primary spinal tumors can also occur, and are more common in adult members than pediatric members.

    Surgery is the primary treatment modality. Radiotherapy +/- chemotherapy is used for:

      ® Incompletely resected tumors
      ® Anaplastic histology
      ® Infratentorial location

    PET Brain Metabolic imaging does not have a defined role in the evaluation of ependymoma.

    MR Spectroscopy (CPT® 76390) can be approved in the following circumstances:

      ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed

    Ependymoma Initial Staging:

    MRI Brain without and with contrast (CPT® 70553) is indicated for all members

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is indicated for all members

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    Ependymoma Treatment Response:

    Members who have resection of the tumor can have a single MRI Brain without and with contrast (CPT® 70553) or MRI without and with contrast of involved spinal level(s) approved following resection to establish baseline imaging and those with a complete resection should then be imaged according to surveillance guidelines

    Members with incomplete resection or high risk histology receiving adjuvant radiation therapy can have a single MRI Brain without and with contrast (CPT® 70553) or involved spinal level(s) approved at completion of radiotherapy and should then be imaged according to surveillance guidelines

    Members treated with chemotherapy can have MRI Brain without and with contrast (CPT® 70553) or involved spinal level(s) approved every 2 cycles during active treatment and at the end of planned chemotherapy

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate at the end of induction chemotherapy for members with localized intracranial tumors

      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    MRI Brain without and with contrast (CPT® 70553) is appropriate at the end of induction chemotherapy for members with localized intraspinal tumors

    Following completion of chemotherapy some members will proceed to second-look surgery and/or radiotherapy

      ® MRI of all known sites of measurable disease can be performed prior to surgery and prior to radiotherapy, if necessary

    MRI Brain without and with contrast (CPT® 70553) and MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate at the end of all planned therapy for all members
      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    Ependymoma Surveillance:

    For members with primary intracranial ependymoma:

      ® MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for1 year, then every 6 months for 1 year, then annually for 10 years
      ® MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved annually for 2 years after completion of therapy for members with no history of spinal cord involvement
      ® For members with metastatic cord involvement at diagnosis, MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved after completion of therapy every 3 months for 1 year, then every 6 months for 1 year, then annually for 10 years
        ¡ MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
    For members with primary intraspinal ependymoma:
      ® MRI without and with contrast of the involved spinal level(s) can be approved after completion of therapy every 3 months for1 year, then every 6 months for 1 year, then annually for 10 years
      ® MRI Brain without and with contrast (CPT® 70553) can be approved annually for 2 years after completion of therapy for members with no history of intracranial involvement
      ® For members with metastatic intracranial involvement at diagnosis, MRI Brain without and with contrast (CPT® 70553) can be approved after completion of therapy every 3 months for 1 year, then every 6 months for 1 year, then annually for 10 years

    MR Spectroscopy is not indicated for routine surveillance


    PEDONC-4.9: Malignant Tumors of the Spinal Cord

    For this condition imaging is medically necessary based on the following criteria:

    Treatment principles are the same as tumors of the brain, and should follow imaging guidelines according to the specific histologic type.

    Multiple spinal cord tumors should raise suspicion for neurofibromatosis.

    Common histologies of primary spinal cord tumor in children include:


      ® Low Grade Glioma, See PEDONC-4.2: Intracranial Low Grade Glioma (LGG) for guidelines
      ® Ependymoma, See PEDONC-4.8: Ependymoma for guidelines
      ® Any type can occur, but other histologies are rare

    Primary site imaging should always include MRI Spine without and with contrast of all involved levels (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT®72158)
      ® Entire spine imaging may be indicated based on the histologic type

    MRI Brain without and with contrast (CPT® 70553) is indicated at initial diagnosis, but may be not be necessary during treatment response and surveillance
      ® Given the rarity of primary spinal cord tumors in children, MRI Brain requests should, in general, be approved for surveillance after recent evaluation by a physician with significant training and/or experience in pediatric spinal cord tumors (most commonly a pediatric neurosurgeon or pediatric oncologist) as the need for intracranial surveillance is highly individualized

    Asymptomatic surveillance imaging should generally end at the time point appropriate for the specific tumor type


    PEDONC-4.10: Craniopharyngioma and Other Hypothalamic/Pituitary Region Tumors

    For this condition imaging is medically necessary based on the following criteria:

    Imaging guidelines and treatment approaches for pediatric pituitary tumors other than craniopharyngioma are consistent with those used for adults with pituitary tumors. For these tumors follow guidelines in Adult Head Imaging Policy (Policy #151 in the Radiology Section); HD-19: Pituitary

    Craniopharyngiomas are less common, accounting for 6 to 8% of pediatric CNS tumors. Most commonly affects children in the preadolescent ages. Several key imaging findings can be used to differentiate the tumors in this region including the presence of calcifications, cysts, and T1/T2 enhancement patterns in craniopharyngiomas. These are best evaluated using a COMBINATION of both MRI and CT modalities. Preoperative prediction is much more successful when BOTH modalities are obtained prior to biopsy.

    Other less common tumors in the optic chiasm, sella, and suprasella region may include Germ Cell Tumors (GCT, see PEDONC-4.7) and Langerhans Cell Histiocytosis (LCH, see PEDONC-18).

    PET Brain Metabolic Imaging and MR Spectroscopy do not have a defined role in the evaluation of craniopharyngioma.

    Craniopharyngioma Initial Staging:

    MRI Brain without and with contrast (CPT® 70553) is indicated for all members

    Concurrent CT Head without contrast (CPT® 70450) can be approved in addition to MRI if cranipharyngioma is suspected

    MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved for members with:

      ® Multicentric tumors
      ® Clinical signs or symptoms suggesting spinal cord involvement
      ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

    Craniopharyngioma Treatment Response:

    Surgical resection is curative for many members. Members who have resection of the tumor can have a single MRI Brain without and with contrast (CPT® 70553) approved following resection to establish baseline imaging and those with a complete resection should then be imaged according to surveillance guidelines.

    Members with incomplete resection and receiving adjuvant radiation therapy can have a single MRI Brain (CPT® 70553) approved at completion of radiotherapy and should then be imaged according to surveillance guidelines

    Those rare members who are treated with chemotherapy can have MRI Brain without and with contrast (CPT® 70553) approved every 2 cycles during active treatment and at the end of planned chemotherapy

      ® Spinal imaging is appropriate every 2 cycles during induction chemotherapy for members with measurable spinal cord disease on MRI
      Craniopharyngioma Surveillance:

      MRI Brain without and with contrast (CPT® 70553) can be approved every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually until 10 years after completion of therapy as late progressions can occur

        ® For additional imaging guidelines for members in long term follow up after CNS tumor treatment that included radiation therapy, See PEDONC-19.3: Second Malignant Neoplasms (SMN)

      MRI Spine is not indicated during surveillance in members without prior history of spinal involvement except to evaluate symptoms suspicious for spinal cord recurrence

      PEDONC-4.11: Primary CNS Lymphoma

      For this condition imaging is medically necessary based on the following criteria:

      Primary CNS lymphoma is a solitary or multifocal mass occurring in the brain without evidence of systemic (bone marrow or lymph node) involvement. Usually associated with immunodeficiency, this is a very rare entity in pediatrics accounting for < 0.1% of pediatric malignancies, so age-specific guidelines have not been established.

      Primary CNS lymphoma imaging indications in pediatric members are identical to those for adult members. See Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-2.7: CNS Lymphoma for imaging guidelines.

      CNS lymphomas also involving bone marrow and/or lymph nodes should be imaged according to: PEDONC-5.3: Pediatric Aggressive Mature B-Cell Non-Hodgkin Lymphomas (NHL).

      PEDONC-4.12: Meningiomas

      For this condition imaging is medically necessary based on the following criteria:

      Account for 1 to 3% of pediatric CNS tumors. Usually associated with neurofibromatosis type 2 (NF-2) or prior therapeutic radiation exposure to the brain. Lifetime risk may be as high as 20% for young children receiving whole brain radiotherapy, most commonly occurring 15 to 20 years after radiation exposure.

      Meningioma imaging indications in pediatric members are identical to those for adult members. See Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-2.8: CNS Meningioma for imaging guidelines.



      PEDONC-4.13: Choroid Plexus Tumors

      For this condition imaging is medically necessary based on the following criteria:

      As a group these account for 1 to 4% of pediatric CNS tumors, and 70% of choroid plexus tumors present within the first 2 years of life.

      Includes the following tumors:

        ® Choroid plexus papilloma
        ® Choroid plexus adenoma, or atypical choroid plexus papilloma
        ® Choroid plexus carcinoma

      PET Metabolic Brain imaging does not have a defined role in the evaluation of choroid plexus tumors.

      MR Spectroscopy (CPT® 76390) can be approved in the following circumstances:

        ® To evaluate a brain lesion of indeterminate nature when the MRS findings will be used to determine whether biopsy/resection can be safely postponed

      Choroid Plexus Papilloma

      Choroid plexus papillomas outnumber other choroid plexus tumors by 4 to 5 times. These ventricular tumors commonly present with hydrocephalus caused by increased CSF production, resulting in signs of increased intracranial pressure. Appearance on MRI Brain without and with contrast (CPT® 70553) is typical, and they are usually treated by excision.

      Regrowth is rare, but repeat MRI Brain without and with contrast (CPT® 70553) is indicated if return of hydrocephalus is suspected or seen on CT imaging

      Choroid Plexus Adenoma or Atypical Choroid Plexus Papilloma

      These are extremely rare tumors with features midway in the malignant spectrum between papillomas and carcinomas. They are more prone to local invasion, but rarely to metastasis. Presenting symptoms are similar to papillomas. Appearance on MRI Brain with and without contrast (CPT® 70553) is typical, and they are usually treated by excision.

      Spinal imaging may be approved if requested at initial diagnosis

      Regrowth is rare, but repeat MRI Brain without and with contrast is indicated if return of hydrocephalus is suspected or Seen on CT imaging

      Choroid Plexus Carcinoma

      This is a very aggressive malignancy, with high rates of metastasis to other parts of the CNS. Prognosis is significantly less favorable than for papillomas with overall survival rates of 35 to 40%. Overall incidence of metastases in choroid plexus carcinoma is 12–50%, which is associated with a worse outcome. TP53 mutations and alternative lengthening telomeres (ALT) are common in members with choroid plexus carcinoma.

      Choroid Plexus Carcinoma Initial Staging:

      MRI Brain without and with contrast (CPT® 70553) is indicated for all members

      MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is indicated for all members

        ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

      Choroid Plexus Carcinoma Treatment Response:

      Surgical gross total resection is curative for many members. Members who have gross or subtotal resection of the tumor can have a single MRI Brain without and with contrast (CPT® 70553) approved following resection to establish baseline imaging.

        ® Members with confirmed gross total resection should then be imaged according to surveillance guidelines.

      Members with incomplete resection who receive adjuvant radiation therapy can have a single MRI Brain without and with contrast (CPT® 70553) approved at completion of radiotherapy and should then be imaged according to surveillance guidelines

      Members treated with chemotherapy can have MRI Brain without and with contrast (CPT® 70553) approved every 2 cycles during active treatment and at the end of planned chemotherapy

      MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate at the end of chemotherapy for members with localized intracranial tumors

        ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain
        ® Spinal imaging is appropriate every 2 cycles during chemotherapy for members with measurable spinal cord disease on MRI

      Following completion of chemotherapy some members will proceed to second-look surgery and/or radiotherapy
        ® MRI of all known sites of measurable disease can be performed prior to surgery and prior to radiotherapy, if necessary

      MRI Brain without and with contrast (CPT® 70553) and MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is appropriate at the end of all planned therapy
        ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

      Choroid Plexus Carcinoma Surveillance:

      MRI Brain without and with contrast (CPT® 70553) can be approved every 4 months for 3 years, then every 6 months for 2 years after completion of therapy

        ® For additional imaging guidelines for members in long term follow up after CNS tumor treatment that included radiation therapy, See PEDONC-19.3: Second Malignant Neoplasms (SMN)

      MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved at 12 and 24 months after completion of therapy for members with no history of spinal cord involvement

      For members with cord involvement at diagnosis, MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) can be approved every 4 months for 3 years, then every 6 months for 2 years after completion of therapy

        ® MRI Spine with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) can be approved if being performed immediately following a contrast-enhanced MRI Brain

      MR Spectroscopy is not indicated for routine surveillance

      References

      1. Parsons DW, Pollack IF, Hass-Kogan DA, et al. Gliomas, ependymomas, and other nonembryonal tumors of the central nervous system. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:628-670.
      2. Chintagumpala MM, Paulino A, Panigraphy A, et al. Embryonal and pineal region tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:671-699.
      3. Kieran MW, Chi SN, Manley PE, et al. Tumors of the Brain and Spinal Cord. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1779-1885.
      4. Nabors LB, Portnow J, Ahluwalia M, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 1.2019—March 5, 2019. Central Nervous System Cancers, available at: https://www.nccn.org/professionals/physician_gls/pdf/cns.pdf, referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Central Nervous System Cancers V1.2019 3/15/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org
      5. Menashe SJ, Iyer RS. Pediatric Spinal neoplasia: a practical imaging overview of intramedullary, intradural, and osseous tumors. Curr Probl Diagn Radiol. 2013;42(6):249-265. doi:10.1067/j.cpradiol.2013.05.003.
      6. Warmuth-Metz M. Imaging guidelines for pediatric brain tumor patients. In: Warmuth-Metz M, ed. Imaging and Diagnosis in Pediatric Brain Tumor Studies. Switzerland; Springer,Cham:2017:55-67.
      7. Warren KW, Poussaint TY, Vezina G, et al. Challenges with defining response to antitumor agents in pediatric neuro-oncology: a report from the response assessment in pediatric neuro-oncology (RAPNO) working group. Pediatr Blood Cancer. 2013;60(9):1397-1401. doi:10.1002/pbc.24562.
      8. Zukotynski K, Fahey F, Kocak M, et al. 18F-FDG PET and MR Imaging associations across a spectrum of pediatric brain tumors: a report from the Pediatric Brain Tumor Consortium. J Nucl Med. 2014;55(9):1473-1480. doi:10.2967/jnumed.114.139626.
      9. Gajjar A, Bowers DC, Karajannis MA, Leary S, Witt H, Gottardo NG. Pediatric brain tumors: innovative genomic information is transforming the diagnostic and clinical landscape. J Clin Oncol. 2015;33(27):2986-2998. doi:10.1200/JCO.2014.59.9217.
      10. Brandão LA, Poussaint TY. Pediatric brain tumors. Neuroimag Clin N Am. 2013 August;23(3):499-525. Accessed January 3, 2018. http://www.neuroimaging.theclinics.com/article/S1052-5149(13)00017-8/abstract.
      11. Brandão LA and Castillo M. Adult brain tumors: clinical applications of magnetic resonance spectroscopy. Neuroimag Clin N Am. 2013;23(3):527-555. doi:10.1016/j.nic.2013.03.003
      12. Zarifi M, Tzika AA. Proton MRS imaging in pediatric brain tumors. Pediatr Radiol. 2016;46(7):952-962. doi:10.1007/s00247-016-3547-5.
      13. Uslu L, Doing J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG-PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
      14. Soffietti R, Baumert BG, Bello L, et al. Guidelines on management of low-grade gliomas: report of an EFSN-EANO* Task Force. Eur J Neurol. 2010;17(9):1124-1133. doi:10.1111/j.1468-1331.2010.03151.x.
      15. Kruer MC, Kaplan AM, Etzl MM Jr, et al. The value of positron emission tomography and proliferation index in predicting progression in low-grade astrocytomas of childhood. J Neurooncol. 2009;95(2):239-245. doi:10.1007/s11060-009-9922-4.
      16. Chamdine O, Broniscer A, Wu S, Gajjar A, Qaddoumi I. Metastatic low-grade gliomas in children: 20 years’ experience at St. Jude Children’s Research Hospital. Pediatr Blood Cancer. 2016;63(1):62-70. doi:10.1002/pbc.25731.
      17. Chalil A, Ramaswamy V. Low grade gliomas in children. J Child Neurol. 2016;31(4):517-522. doi:10.1177/0883073815599259.
      18. Krishnatry R, Zhukova N, Stucklin ASG, et al. Clinical and treatment factors determining long-term outcomes for adult survivors of childhood low-grade glioma: a population-based study. Cancer. 2016;122(8):1261-1269. doi:10.1002/cncr.29907.
      19. Ullrich NJ. Neurocutaneous Syndromes and brain tumors. J Child Neurol. 2016;31(12):1399-1411. doi:10.1177/0883073815604220.
      20. Campian J, Gutmann DH. CNS tumors in neurofibromatosis. J Clin Oncol. 2017; 35(21):2378-2385. doi:10.1200/JCO.2016.71.7199.
      21. Morris EB, Gajjar A, Okuma JO, et al. Survival and late mortality in long-term survivors of pediatric CNS tumors. J Clin Oncol. 2007;25(12):1532-1538. doi:10.1200/JCO.2006.09.8194.
      22. Karthigeyan M, Gupta K, Salunke P. Pediatric central neurocytoma: a short series with literature review. J Child Neurol. 2016;32(1):53-59. Accessed January 3, 2018 doi:10.1177/0883073816668994.
      23. Tisnado J, Young R, Peck KK, et al. Conventional and advanced imaging of diffuse intrinsic pontine glioma. J Child Neurol. 2016;31(12):1386-1393. doi:10.1177/0883073816634855.
      24. Greenfield JP, Heredia AC, George E, Keiran MW, Morales La Madrid A. Gliomatosis cerebri: a consensus summary report from the First International Gliomatosis cerebri Group Meeting, March 26-27, 2015, Paris, France. Pediatr Blood Cancer. 2016;63(12):2072-2077. doi:10.1002/pbc.26169.
      25. Zaky W, Dhall G, Ji L, et al. Intensive induction chemotherapy followed by myeloablative chemotherapy with autologous hematopoietic progenitor cell rescue for young children newly-diagnosed with central nervous system atypical teratoid/rhabdoid tumors: the head start III experience. Pediatr Blood Cancer. 2014;61(1):95-101. doi:10.1002/pbc.24648.
      26. Gururangan S, Hwang E, Herndon JE II, et al. [18F]Fluorodeoxyglucose-Positron Emission Tomography in patients with medulloblastoma. Neurosurgery. 2004;55(6):1280-1289. doi:10.1227/01.NEU.0000143027.41632.2B.
      27. Millard NE and DeBraganca KC. Medulloblastoma. J Child Neurol. 2016;31(12):1341-1353. doi:10.1177/0883073815600866.
      28. Koschmann C, Bloom K, Upadhyaya S, Geyer JR. Survival after relapse of medulloblastoma. J Pediatr Hematol. Oncol. 2016;38(4):269-273. doi:10.1097/MPH.0000000000000547.
      29. Martinex S, Khakoo Y, Giheeney S, et al. Marker (+) CNS germ cell tumors in remission: are surveillance MRI scans necessary? Pediatr Blood Cancer. 2014;61(5):853-854. Accessed January 3, 2018. doi:10.1002/pbc.24888.
      30. Goldman S, Bouffet E, Fisher PG, et al. Phase II trial assessing the ability of neoadjuvant chemotherapy with or without second-look surgery to eliminate measurable disease for nongerminomatous germ cell tumors: a children’s oncology group study. J Clin Oncol. 2015;33(22):2464-2471. doi:10.1200/JCO.2014.59.5132.
      31. Abu Arja MH, Bouffet E, Finlay JL, AbdelBaki MS. Critical review of the management of primary central nervous system germ cell tumors. Pediatr Blood Cancer. 2019;66:e27658. doi:10.1002/pbc.27658.
      32. Vitanza NA, Partap S. Pediatric Ependymoma. J Child Neurol. 2016;31(12):1354-1366. doi:10.1177/0883073815610428
      33. Norris GA, Garcia J, Hankinson TC, et al. Diagnostic accuracy of neuroimaging in pediatric optic chiasm/suprasellar tumors. Pediatr Blood Cancer. 2019;66:e27860. doi:10.1002/pbc.27680.
      34. McCrea HJ, George E, Settler A, et al. Pediatric suprasellar tumors. J Child Neurol. 2016;31(12):1367-1376. doi:10.1177/0883073815620671.
      35. Grommes C, DeAngelis LM. Primary CNS lymphoma. J Clin Oncol. 2017;35(21):2410-2418. doi:10.1200/JCO.2017.72.7602.
      36. Larrew T, Eskandari R. Pediatric intraparenchymal meningioma, case report and comparative review. Pediatr Neurosurg. 2016;51(2):83-86. doi:10.1159/000441008.
      37. Li Z, Li H, Wang S, et al. Pediatric skull base meningiomas: clinical features and surgical outcomes. J Child Neurol. 2016;31(14):1523-1527. doi:10.1177/0883073816664669.
      38. Horská A, Barker PB. Imaging of brain tumors: MR Spectroscopy and metabolic imaging. Neuroimaging Clin N Am. 2010;20(3):293-310. doi:10.1016/j.nic.2010.04.003.
      39. Sundgren PC. MR Spectroscopy in radiation injury. AJNR Am J Neuroradiol. 2009;30(8):1469-1476. doi:10.3174/ajnr.A1580.
      40. Zaky W, Dhall G, Khatua S, et al. Choroid plexus carcinoma in children: the head start experience. Pediatr Blood Cancer. 2015;62(5):784-789. doi:10.1002/pbc.25436.
      41. Zaky W, Finlay JL, Pediatric choroid plexus carcinoma: Biologically and clinically in need of new perspectives, Pediatr Blood Cancer 2018;65:e27031. doi:10.1002/pbc.27031.
      42. Kramer K. Rare primary central nervous system tumors encountered in pediatrics. J Child Neurol. 2016;31(12):1394-1398. doi:10.1177/0883073815627878.


      PEDONC-5: Pediatric Lymphomas

      PEDONC-5.1: Pediatric Lymphoma – General Considerations
      PEDONC-5.2: Pediatric Hodgkin Lymphoma (HL)
      PEDONC-5.3: Pediatric Aggressive Mature B-Cell Non-Hodgkin Lymphomas (NHL)
      PEDONC-5.4: Anaplastic Large Cell Lymphoma (ALCL)

      PEDONC-5.1: Pediatric Lymphoma – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Lymphoma mostly commonly involves the lymph nodes (LNs). However, lymphoma can also arise from primary lymphoid tissues (bone marrow or thymus) or various secondary lymphoid tissues (spleen, mucosa-associated lymphoid tissue) or non-lymphoid organs (skin, bone, brain, lungs, liver, salivary glands, etc).

      Pediatric lymphomas are generally Hodgkin Lymphomas, Aggressive B-Cell Non-Hodgkin Lymphomas, Lymphoblastic Lymphomas, or Anaplastic Large Cell Lymphomas

      Members with Lymphoblastic Lymphoma (even those with bulky nodal disease) are treated using the leukemia treatment plan appropriate to the cell type (B or T cell). These members should be imaged using guidelines in PEDONC-3.2: Acute Lymphoblastic Leukemia.

      Other histologies are rare in pediatric members, and should be imaged according to the following guidelines:

        ® Follicular lymphoma: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-27.3: Follicular Lymphoma
        ® Marginal zone or MALT lymphomas: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-27.4: Marginal Zone Lymphomas
        ® Mantle cell lymphomas: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-27.5: Mantle Cell Lymphoma
        ® Cutaneous lymphomas: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-27.8: Cutaneous Lymphomas and T Cell Lymphomas
          ¡ Exception: Cutaneous B-Lymphoblastic Lymphoma should be imaged using guidelines in PEDONC-3.2: Acute Lymphoblastic Leukemia
        ® Castleman’s Disease: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-31.11: Castleman’s Disease (Unicentric and Multicentric)

      All CT imaging recommended in this section refers to CT with contrast only.
        ® Noncontrast CT imaging has not been shown to be beneficial in the management of pediatric lymphomas
        ® Given the limited utility of noncontrast CT imaging in pediatric lymphomas, MRI without or without and with contrast is recommended in place of CT for members who cannot tolerate CT contrast due to allergy or impaired renal function

      MRI without and with contrast of symptomatic or previously involved bony areas can be approved in known lymphoma members without prior plain x-ray or bone scan evaluation
        ® Bone scan is inferior to MRI for evaluation of known or suspected bone metastases in lymphoma

      MRI Brain without and with contrast (CPT® 70553) is the preferred study for evaluation of suspected Brain metastases in pediatric lymphoma
        ® CT Head with (CPT® 70460) or without and with contrast (CPT® 70470) can be approved when MRI is contraindicated
      PEDONC-5.2: Pediatric Hodgkin Lymphoma (HL)

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric Hodgkin Lymphoma Initial Staging:

      All members should undergo CT Neck (CPT® 70491), Chest (CPT® 71260), Abdomen/Pelvis (CPT® 74177), and CT with contrast or MRI without and with contrast of any other symptomatic body area is indicated for all members (See PEDONC-5.1: Pediatric Lymphoma General Considerations) as pediatric members have a high rate of neck and Waldeyer’s ring involvement with Hodgkin Lymphoma

      PET/CT (CPT® 78815) is indicated for initial staging of all members, and can be performed prior to biopsy if necessary for member scheduling

        ® Whole body PET/CT (CPT® 78816) may be approved if there is clinical suspicion of skull or distal lower extremity involvement

      CT or MRI of other body areas (See PEDONC-5.1: Pediatric Lymphoma General Considerations) may be indicated for rare members based on physical findings or PET/CT results.

      Pediatric Hodgkin Lymphoma Treatment Response:

      Restaging for treatment response can be performed as often as every 2 cycles of chemotherapy

      Both CT of Neck (CPT® 70491), Chest (CPT® 71260), and Abdomen/Pelvis (CPT® 74177) and other previously involved areas and PET/CT (CPT® 78815) can be approved during early treatment response evaluations as decisions about chemotherapy drug selection and radiation treatment are frequently made based on both anatomic (CT-based) and metabolic (PET/CT-based) responses.

        ® For members with low risk (stage IA or IIA) mixed cellularity Hodgkin lymphoma, PET/CT can be performed for treatment response after cycles 1 and 3 instead of cycles 2 and 4

      Once a particular member has a negative PET/CT (either Deauville or Lugano 1, 2 or 3 as reported in formal radiology interpretation), all subsequent treatment response evaluations should use CT only, including end of therapy evaluation.

      Pediatric Hodgkin Lymphoma Surveillance:

      Most members experiencing recurrence are detected based on physical findings, and frequent CT surveillance imaging of Hodgkin Lymphoma after completion of therapy does not improve post-recurrence overall survival.

      CT of the Neck (CPT® 70491), Chest (CPT® 71260), Abdomen/Pelvis (CPT® 74177) and other previously involved or currently symptomatic areas OR PET/CT (CPT® 78815 or 78816) should be approved for any member with clinical symptoms suggesting recurrence

      Members with stage I or II HL:

        ® CT of the Neck/Chest (CPT® 70491 and CPT® 71260) and other previously involved areas at 6 months and 12 months after completing therapy
        ® Surveillance at other time points from the end of therapy should use physical exam and CXR only

      Members with stage III or IV HL:
        ® CT of the Neck (CPT® 70491), Chest (CPT® 71260), and Abdomen/Pelvis (CPT® 74177) and other previously involved areas at 6 months and 12 months after completing therapy
        ® Surveillance at other time points from the end of therapy should use physical exam and CXR only

      Members with recurrent HL with no evidence of disease following successful treatment:
        ® CT of the Neck/Chest/Abdomen/Pelvis every 3 months for 1 year after completing therapy for recurrence

      PET/CT is not indicated for surveillance, but can be approved to clarify inconclusive findings on conventional imaging to evaluate the need for biopsy to establish recurrence. These requests should be forwarded for Medical Director review.


      PEDONC-5.3: Pediatric Aggressive Mature B-Cell Non-Hodgkin Lymphomas (NHL)

      For this condition imaging is medically necessary based on the following criteria:

      Aggressive mature B-Cell NHL includes all of the following diagnoses, all of which should be imaged according to this section:

        ® Burkitt’s lymphoma/leukemia (BL)
        ® Diffuse Large B-Cell Lymphoma (DLBCL)
        ® Primary Mediastinal Large B-Cell Lymphoma (PMBCL)
        ® Post-transplant Lymphoproliferative Disorder (PTLD)
          ¡ Most commonly occurs following solid organ or stem cell transplantation
        ® Viral-associated lymphoproliferative disorders
          ¡ Most commonly occurs following hematopoietic stem cell transplantation or in members with primary immunodeficiency
      Pediatric Aggressive Mature B-Cell NHL Initial Staging:

      CT of the Neck (CPT® 70491), Chest (CPT® 71260), and Abdomen/Pelvis (CPT® 74177) and CT with contrast or MRI without and with contrast any other symptomatic body area is indicated for all members (See PEDONC-5.1: Pediatric Lymphoma General Considerations)

      MRI Brain without and with contrast (CPT® 70553) is indicated if symptoms or extent of disease suggest intracranial extension (skull base involvement, for example) or metastasis

      PET/CT (CPT® 78815) is indicated for initial staging for all members

        ® Whole body PET/CT (CPT® 78816) may be approved if there is clinical suspicion of skull or distal lower extremity involvement.
        ® Due to the extremely aggressive nature of this group of tumors (the doubling time can be as short as 8 hours) it may not be possible to obtain PET/CT prior to therapy initiation. PET/CT should be approved for treatment response in these cases as these lymphomas are nearly universally FDG-avid.

      Pediatric Aggressive Mature B-Cell NHL Treatment Response:

      Initial treatment is usually 7 days of low intensity therapy, with early response evaluation determining next steps in therapy using CT with contrast or MRI without and with contrast of previously involved areas performed around day 6

        ® Members are customarily still inpatient for this evaluation so outpatient requests should be rare for this time point

      Following initial response evaluation, restaging for treatment response using CT with contrast or MRI without and with contrast (should be same modality as initial diagnosis if possible) of previously involved areas and PET/CT can be performed as often as every cycle of chemotherapy (~every 3 weeks)

      Once a particular member has a negative PET/CT (either Deauville or Lugano 1, 2 or 3 as reported in formal radiology interpretation), all subsequent treatment response evaluations should use CT imaging only, including end of therapy evaluation

        ® PET/CT may be indicated to assess disease activity in inconclusive residual masses seen on conventional imaging

      Pediatric Aggressive Mature B-Cell NHL Surveillance:

      Routine asymptomatic surveillance with advanced imaging has not been found to impact member outcomes as the majority of these members present clinically at relapse due to the highly aggressive nature of these lymphomas.

      CXR and Abdominal (CPT® 76700) and Pelvic (CPT® 76856) ultrasound are sufficient to follow asymptomatic members with residual masses in the chest or abdomen/pelvis. Surveillance imaging with CT or MRI has not been shown to improve member outcomes following recurrence and is not the standard of care.

      CT of the Neck (CPT® 70491), Chest (CPT® 71260), Abdomen/Pelvis (CPT® 74177) and other previously involved or currently symptomatic areas OR PET/CT (CPT® 78815 or 78816) should be approved for any member with clinical symptoms or laboratory findings suggesting recurrence.

        ® PET/CT (CPT® 78815) can be approved for suspected PTLD recurrence with documentation of new palpable nodes, rising LDH, or rising quantitative EBV PCR

      PET/CT is not indicated for surveillance, but can be approved to clarify inconclusive findings on conventional imaging to evaluate the need for biopsy to establish recurrence. These requests should be forwarded for Medical Director review.


      PEDONC-5.4: Anaplastic Large Cell Lymphoma (ALCL)

      For this condition imaging is medically necessary based on the following criteria:

      Similar in presentation to Hodgkin Lymphoma, and may be indistinguishable until immunocytology and molecular studies are complete.

      Anaplastic Large Cell Lymphoma Initial Staging:

      All members should undergo CT of the Neck/Chest/Abdomen/Pelvis (CPT® 70491, CPT® 71260, and CPT® 74177) and CT with contrast or MRI without and with contrast any other symptomatic body area is indicated for all members (See PEDONC-5.1: Pediatric Lymphoma General Considerations)

      PET/CT (CPT® 78815) is indicated for initial staging of all members and can be performed prior to biopsy if necessary for member scheduling.

        ® Whole body PET/CT (CPT® 78816) may be approved if there is clinical suspicion of skull or distal lower extremity involvement.

      CT or MRI of other body areas may be indicated for rare members based on physical findings or PET/CT results. Rarely members will have primary tumor sites outside the NeckPelvis region, and MRI without and with contrast may be substituted for soft tissue extremity or paraspinal primary masses as necessary.

      Bone scan (See PEDONC-1.3: Modality General Considerations) is indicated for members with bony primary tumors or metastatic disease

      Anaplastic Large Cell Lymphoma Treatment Response:

      Restaging for treatment response using CT with contrast or MRI without and with contrast of previously involved areas (should be same modality as initial diagnosis if possible) should be performed at the end of induction chemotherapy (commonly 4 to 6 weeks)

      For members treated with cytotoxic chemotherapy, either CT of previously involved areas or PET/CT may be approved for treatment response as often as every 2 cycles of chemotherapy as decisions about chemotherapy drug selection and radiation treatment can be made based on either anatomic or metabolic responses.

        ® If CT is performed for primary treatment response, PET/CT can be approved to clarify inconclusive findings detected on conventional imaging
        ® If PET/CT is performed for primary treatment response, CT or MRI can be approved to clarify inconclusive findings detected on PET imaging

      Once a particular member has a negative PET/CT (either Deauville or Lugano 1, 2 or 3 as reported in formal radiology interpretation), all subsequent treatment response evaluations should use CT imaging only, including end of therapy evaluation.

      Anaplastic Large Cell Lymphoma Surveillance:

      CT of the Neck (CPT® 70491), Chest (CPT® 71260), Abdomen/Pelvis (CPT® 74177) and other previously involved or currently symptomatic areas should be approved for any member with clinical symptoms suggesting recurrence.

      CT with contrast or MRI without and with contrast of all previously involved areas is indicated at 3, 6, 12, and 18 months after therapy is completed.

      Bone scan (See PEDONC-1.3: Modality General Considerations) is indicated at 3, 6, 12, and 18 months after therapy is completed for members with bony primary tumors or metastatic disease

      PET/CT is not indicated for surveillance, but can be approved to clarify inconclusive findings on conventional imaging to evaluate the need for biopsy to establish recurrence. These requests should be forwarded for Medical Director review.

      References

      1. Metzger ML, Krasin MJ, Choi JK, et al. Hodgkin lymphoma. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:568-586.
      2. Allen CE, Kamdar KY, Bollard CM, et al. Malignant Non-Hodgkin lymphomas in Children. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:587-603.
      3. Hayashi RJ, Wistinghausen B, Shiramizu B. Lymphoproliferative Disorders and Malignancies Related To Immunodeficiencies. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:604-616.
      4. Alexander S, Ferrando AA. Pediatric lymphoma. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1626-1671.
      5. Lee Chong A, Grant RM, Ahmed BA, Thomas KE, Connolly BL, Greenberg M. Imaging in pediatric patients: time to think again about surveillance. Pediatr Blood Cancer. 2010;55(3):407-413. doi:10.1002/pbc.22575.
      6. Uslu L, Doing J, Link M, Rosenberg J, Quon A, Daldrup-Link H. Value of 18F-FDG-PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
      7. Ceppi F, Pope E, Ngan B, Abla O. Primary cutaneous lymphomas in children and adolescents. Pediatr Blood Cancer. 2016;63(11):1886-1894. doi:10.1002/pbc.26076.
      8. Flerlage JE, Kelly KM, Beishuizen A, et al. Staging evaluation and response criteria harmonization (SEARCH) for childhood, adolescent, and young adult Hodgkin Lymphoma (CAYAHL): Methodology statement. Pediatr Blood Cancer. 2017;64(7):e26421. doi:10.1002/pbc.26421.
      9. Friedman DL, Chen L, Wolden S, et al. Dose-intensive response-based chemotherapy and radiation therapy for children and adolescents with newly diagnosed intermediate-risk Hodgkin lymphoma: a report from the Children’s Oncology Group Study AHOD0031. J Clin Oncol. 2014;32(32):3651-3658. doi:10.1200/JCO.2013.52.5410.
      10. Schwartz CL, Chen L, McCarten K, et al. Childhood Hodgkin International Prognostic Score (CHIPS) predicts event-free survival in Hodgkin lymphoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2017;64(4):e26278. doi:10.1002/pbc.26278.
      11. Friedmann AM, Wolfson JA, Hudson MM, et al. Relapse after treatment of Hodgkin lymphoma: outcome and role of surveillance after therapy. Pediatr Blood Cancer. 2013;60(9):1458-1463. doi:10.1002/pbc.24568.
      12. Mauz-Körholz C, Metzger ML, Kelly KM, et al. Pediatric Hodgkin lymphoma. J Clin Oncol. 2015;33(27):2975-2985. doi:10.1200/JCO.2014.59.4853.
      13. Voss SD, Chen L, Constine LS, et al. Surveillance computed tomography imaging and detection of relapse in intermediate- and advanced-stage pediatric Hodgkin’s lymphoma: A report from the Children’s Oncology Group. J Clin Oncol. 2012;30(21):2635-2640. doi:10.1200/JCO.2011.40.7841.
      14. Voss SD. Surveillance Imaging in Pediatric Hodgkin Lymphoma. Curr Hematol Malig Rep. 2013;8(3):218-225. doi:10.1007/s11899-013-0168-z.
      15. Appel BE, Chen L, Buxton AB, et al. Minimal treatment of low-risk, pediatric lymphocyte-predominant Hodgkin lymphoma: a report from the Children’s Oncology Group. J Clin Oncol. 2016;34(20):2372-2379. doi:10.1200/JCO.2015.65.3469.
      16. Rosolen A, Perkins SL, Pinkerton CR, et al. Revised international pediatric non-Hodgkin lymphoma staging system. J Clin Oncol. 2015;33:2112-2118. doi:10.1200/JCO.2014.59.7203.
      17. Sandlund JT, Guillerman RP, Perkins SL, et al. International pediatric Non-Hodgkin lymphoma response criteria. J Clin Oncol. 2015;33(18):2106-2111. doi:10.1200/JCO.2014.59.0745.
      18. Eissa HM, Allen CE, Kamdar K, et al. Pediatric Burkitt’s lymphoma and Diffuse B-cell lymphoma: are surveillance scans required? Pediatr Hematol Oncol. 2014;31(3):253-257. doi:10.3109/08880018.2013.834400.
      19. Minard-Colin V, Brugieres L, Reiter A, et al. Non-Hodgkin lymphoma in children and adolescents: progress through effective collaboration, current knowledge, and challenges ahead. J Clin Oncol. 2015;33(27):2963-2974. doi:10.1200/JCO.2014.59.5827.
      20. Dierickx D, Tousseym T, and Gheysens O. How I treat posttransplant lymphoproliferative disorders. Blood. 2015;126(20):2274-2283. doi:10.1182/blood-2015-05-615872.
      21. Vali R, Punnett A, Bajno L, Moineddin R, Shammas A. The value of 18F-FDG PET in pediatric patients with post-transplant lymphoproliferative disorder at initial diagnosis. Pediatr Transplant. 2015;19(8):932-939. doi:10.1111/petr.12611.
      22. Hapgood G and Savage KJ. The biology and management of systemic anaplastic large cell lymphoma. Blood. 2015;126(1):17-25. doi:10.1182/blood-2014-10-567461.
      23. Kempf W, Kazakov DV, Belousova IE, Mitteldorf C, Kerl K. Pediatric cutaneous lymphomas: a review and comparison with adult counterparts. J Eur Acad Dermatol Venereol. 2015;29(9):1696-1709. doi:10.1111/jdv.13044.
      24. Hochberg J, Flower A, Brugieres L, Cairo MS. NHL in adolescents and young adults: a unique population. Pediatr Blood Cancer. 2018;65:e27073. doi.org/10.1002/pbc.27073.
      25. Buhtoiarov IN. Pediatric lymphoma. Pediatr In Rev. 2017;38(9):410-423. doi:10.1542/pir.2016-0152.

      PEDONC-6: Neuroblastoma

      PEDONC-6.1: Neuroblastoma – General Considerations
      PEDONC-6.2: Staging and Risk Grouping – Neuroblastoma
      PEDONC-6.3: Neuroblastoma – Initial Staging
      PEDONC-6.4: Neuroblastoma – Treatment Response Imaging (Risk Group Dependent)
      PEDONC-6.5: Neuroblastoma – Surveillance Imaging (Risk Group Dependent)

      PEDONC-6.1: Neuroblastoma – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Note: Some payors consider PET to be investigational for the treatment of neuroblastoma, and those coverage policies may supersede the recommendations for PET in this section.

      Neuroblastoma is the most common extracranial solid tumor of childhood, and generally arises from the adrenal gland or along the sympathetic chain. Neuroblastoma is divided into very low, low, intermediate, and high risk disease based on International Neuroblastoma Risk Group (INRG) Staging System (see: PEDONC-6.5). The treatment approaches for each risk group vary widely and have distinct imaging strategies.

      90 to 95% of neuroblastomas secrete homovanillic acid (HVA) and vannilylmandelic acid (VMA) in the urine, and urine HVA/VMA should be performed at every disease evaluation for members with positive HVA or VMA at diagnosis.

      Esthesioneuroblastoma should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-3: Squamous Cell Carcinomas Of The Head And Neck

      PET imaging is rarely indicated in neuroblastoma, but can be approved in the following situations:

        ® Members with MIBG-negativity documented at initial diagnosis
          ¡ For these members, MIBG should not be repeated and whole body PET (CPT® 78816) may be performed rather than MIBG for metabolic tumor assessment.
          ¡ Members who are MIBG positive at diagnosis and then become MIBG negative in response to treatment should continue to use MIBG (CPT® 78800, 78801, 78802, 78803, or 78804) for metabolic imaging indications
        ® For all members, PET may be approved at major decision points such as hematopoietic stem cell transplantation or surgery if MIBG and CT/MRI findings are inconclusive
        ® Members currently receiving medications that may interfere with MIBG uptake that cannot safely be discontinued prior to imaging, including:
          ¡ Tricyclic antidepressants (amitriptyline, imipramine, etc.)
          ¡ Selective serotonin reuptake inhibitors (SSRI’s, sertraline, paroxetine, escitolapram, etc.)
          ¡ Neuroleptics (risperidone, haloperidol, etc.)
          ¡ Antihypertensive drugs (alpha or beta blockers, calcium channel blockers)
          ¡ Decongestants (phenylephrine, ephedrine, pseudoephedrine)
          ¡ Stimulants (methylphenidate, dextroamphetamine, etc.)
          ¡ PET should only be approved for this indication when specific documentation of the medication interaction is included with the current PET imaging request. These requests will be forwarded for Medical Director review.
      99mTc-MDP bone scan does not identify foci of disease that affect staging or clinical management and provides no advantage over MIBG scintigraphy and is not used for evaluation of most members with neuroblastoma

      PEDONC-6.2: Staging and Risk Grouping – Neuroblastoma

      For this condition imaging is medically necessary based on the following criteria:

      Most recent treatment protocols are using the recently validated International Neuroblastoma Risk Group (INRG) staging system, which is primarily defined by the complexity of local tumor extension and the presence or absence of distant metastases:

        ® L1: Localized tumor not involving vital structures as defined by the list of image-defined risk factors and confined to one body compartment
          ¡ Image-defined risk factors include a list of specific imaging findings defining members less likely to be candidates for complete surgical resection
          ¡ These risk factors involve the encasement of major blood vessels, airway, skull base, costovertebral junction, brachial plexus, spinal canal, or major organs or structures
        ® L2: Locoregional tumor with presence of one or more image-defined risk factors
        ® M: Distant metastatic disease (except stage MS)
        ® MS: Metastatic disease in children younger than 18 months with metastases confined to skin, liver, and/or bone marrow with < 10% involvement (MIBG must be negative in bone and bone marrow)
      INRG Neuroblastoma Risk Grouping
      Very low risk neuroblastoma (28% of members, event-free survival > 85%) includes:
      Stage L1 or L2 maturing ganglioneuroma or intermixed ganglioneuroblastoma

      Stage MS members meeting all of the following:

        ® Age < 18 months
        ® Without MYCN amplification
        ® Without 11q aberration
      Low Risk Neuroblastoma (27% of members, event-free survival > 75 to ≤ 85%) includes:
      Stage L2 members age < 18 months meeting all of the following:
        ® Any histology except maturing ganglioneuroma or intermixed ganglioneuroblastoma
        ® Without MYCN amplification
        ® Without 11q aberration

      Stage L2 members age ≥ 18 months meeting all of the following:
        ® Differentiating neuroblastoma or nodular ganglioneuroblastoma
        ® Without MYCN amplification
        ® Without 11q aberration

      Stage M members meeting all of the following:
        ® Age < 18 months
        ® Without MYCN amplification
        ® With hyperdiploidy (tumor DNA index > 1)
      Intermediate Risk Neuroblastoma (9% of members, event-free survival ≥ 50 to ≤ 75%) includes:
      Stage L2 members age < 18 months meeting all of the following:
        ® any histology except maturing ganglioneuroma or intermixed ganglioneuroblastoma
        ® With 11q aberration

      Stage L2 members age ≥ 18 months meeting all of the following:
        ® Neuroblastoma or nodular ganglioneuroblastoma
        ® Without MYCN amplification
        ® With 11q aberration

      Stage M members meeting all of the following:
        ® Age < 18 months
        ® Without MYCN amplification
        ® With diploidy (tumor DNA index = 1)
      High Risk Neuroblastoma (36% of members, event-free survival < 50%, includes the following)
      All members age ≥ 18 months with stage M disease regardless of other factors

      All members with neuroblastoma and MYCN amplification regardless of other factors

      All stage MS members with 11q aberration regardless of other factors

      PEDONC-6.3: Neuroblastoma – Initial Staging

      For this condition imaging is medically necessary based on the following criteria:

      One of the following imaging groups for all members:

        ® CT with contrast of the Neck/Chest/Abdomen/Pelvis (CPT® 70491, 71260, and 74177) OR
        ® MRI without and with contrast of the Neck/Chest/Abdomen/Pelvis (CPT® 70543, 71552, 74183, and 72197)

      MRI without and with contrast is preferred for evaluation of paraspinal tumors where cord compression is a possibility

      Metabolic imaging in neuroblastoma:

        ® Adrenal nuclear imaging (CPT® 78075) can be approved for evaluation of suspected adrenal neuroblastoma, ganglioneuroblastoma, or ganglioneuroma when CT or MRI is inconclusive
        ® 123I-metaiodobenzylguanidine (MIBG - CPT® 78800, 78801, 78802, 78803, or 78804) scintigraphy is the preferred metabolic imaging for neuroblastoma and is positive in 90 to 95% of neuroblastomas.
          ¡ MIBG provides superior sensitivity and sensitivity for detecting viable osseous disease compared with bone scintigraphy so technetium bone scan is not necessary when MIBG is utilized
        ® Most MIBG imaging studies are SPECT/CT studies using CT for localization only. Separate diagnostic CT codes should not be approved for this purpose.
        ® Occasionally MIBG cannot be performed prior to initiation of therapy. In this circumstance MIBG should be completed within 3 weeks of therapy initiation as the reduction in MIBG avidity in response to chemotherapy is not immediate. Inability to complete MIBG before starting therapy is not an indication to approve PET imaging.
        ® PET imaging is inferior to MIBG in neuroblastoma, and should not be used unless one of the exceptions stated in section PEDONC-6.1 is present

      Brain metastases are rare in neuroblastoma, but if clinical signs/symptoms suggest brain involvement, MRI Brain without and with contrast (CPT® 70553) is preferred for evaluation.
        ® MRI Brain of asymptomatic members with no history of brain metastases is not indicated for neuroblastoma.

      PEDONC-6.4: Neuroblastoma – Treatment Response Imaging (Risk Group Dependent)

      For this condition imaging is medically necessary based on the following criteria:

      Risk grouping will not be known at the time of initial staging, but is critical for all imaging decisions after initial staging is complete. The treating oncologist should always know the member’s risk grouping. It is not possible to establish the appropriate imaging plan for a neuroblastoma member without knowing his/her risk group.

      All Very Low Risk and Low Risk Neuroblastoma Not Receiving Chemotherapy:

      All members can have CT with contrast or MRI without and with contrast of the primary tumor site 6 to 8 weeks after diagnosis to determine if additional treatment is necessary.

        ® Ultrasound may be used in place of CT or MRI to avoid radiation and/or anesthesia exposure in low risk members

      Many members will be treated with surgical resection only without adjuvant therapy, and these members enter immediately into surveillance.

      All Intermediate Risk Neuroblastoma and Very Low Risk or Low Risk Neuroblastoma Receiving Chemotherapy:

      Members generally receive 2 to 12 cycles of moderate-intensity chemotherapy depending on response to treatment.

      Surgical resection may occur prior to or following chemotherapy depending on disease stage. Restaging prior to surgery is appropriate.

      Treatment response assessment can be approved as often as every 2 cycles of chemotherapy (~every 6 weeks and at the end of planned treatment) and includes:

        ® CT with contrast of the Chest/Abdomen/Pelvis (CPT® 71260, and CPT® 74177) or MRI without and with contrast, (CPT® 71552, CPT® 74183, and CPT® 72197) and other sites with prior measurable disease
        ® Urine HVA/VMA (if positive at diagnosis)
        ® Bone marrow aspiration/biopsy if positive at diagnosis

      MIBG scan (CPT® 78800, 78801, 78802, 78803, or 78804) can be approved every 4 cycles and at the end of planned treatment

      High Risk Neuroblastoma:

      This group of members receives highly aggressive therapy using sequential chemotherapy, surgery, high dose chemotherapy with stem cell rescue, radiotherapy, monoclonal antibody (mAb) immunotherapy, and biologic therapy.

      Treatment response assessment can be approved as often as every 2 cycles of chemotherapy, mAb, or biologic therapy (~every 6 weeks) and includes:

        ® CT with contrast, of the Chest/Abdomen/Pelvis (CPT® 71260, and CPT® 74177) or MRI without and with contrast, (CPT® 71552, CPT® 74183, and CPT® 72197) and other sites with prior measurable disease
        ® Urine HVA/VMA (if positive at diagnosis)
        ® Bone marrow aspiration/biopsy if positive at diagnosis
        ® MIBG scan (CPT® 78800, 78801, 78802, 78803, or 78804)
          ¡ 123I-MIBG scan is also indicated following 131I-MIBG therapy, but FDG-PET cannot be used after 131I-MIBG therapy
      Treatment response assessment is necessary at every change in modality (prior to surgery, HSCT, XRT, and mAb therapy) as well as at the end of therapy

      More frequent imaging can be approved around the time of surgery if needed for preoperative planning


      PEDONC-6.5: Neuroblastoma – Surveillance Imaging (Risk Group Dependent)

      For this condition imaging is medically necessary based on the following criteria:

      Very Low Risk and Low Risk Neuroblastoma:

      Urine HVA/VMA (if positive at diagnosis) at 1, 2, 3, 6, 9, 12, 18, 24, 36, 48, and 60 months after surgery

      CT with contrast or MRI without and with contrast of the primary tumor site 3, 6, 9, 12, 18, 24, and 36 months after surgery. If negative at 36 months, no further advanced imaging is necessary.

        ® Ultrasound may be sufficient to evaluate the primary tumor site for certain members and may be approved if requested to replace CT or MRI.

      MIBG is not indicated for surveillance of low risk neuroblastoma, but can be used to clarify findings suspicious for disease recurrence

      CT Chest is not indicated in asymptomatic surveillance imaging of neuroblastoma members with no prior history of thoracic disease

      Intermediate Risk Neuroblastoma:

      Urine HVA/VMA (if positive at diagnosis) every month until 12 months after completion of therapy, then at 14, 16, 18, 21, 24, 30, and 36 months after completion of therapy, then annually until 10 years after completion of therapy

      CT with contrast or MRI without and with contrast of the primary tumor and known metastatic sites at 3, 6, 9, 12, 18, 24, and 36 months after completion of therapy. If negative at 36 months, no further advanced imaging is necessary.

        ® Ultrasound may be sufficient to evaluate the primary tumor site for certain members and may be approved if requested to replace CT or MR

      For all members with stage 4 or M disease or members with stage 4S or MS disease AND positive MIBG at completion of therapy, MIBG scan (CPT® 78800, 78801, 78802, 78803, or 78804) at 3, 6, 9, 12, 24, and 36 months after completion of therapy.
        ® If negative at 36 months, no further MIBG imaging is necessary.
        ® For all other intermediate risk neuroblastoma members, MIBG (or PET, if MIBG-negative at initial diagnosis) during surveillance is not indicated.

      CT Chest is not indicated in asymptomatic surveillance imaging of neuroblastoma members with no prior history of thoracic disease.

      High Risk Neuroblastoma:

      Urine HVA/VMA (if positive at diagnosis) at 3, 6, 9, 12, 18, 24, 30, 36, 42, 48, 54, and 60 months after completion of therapy, then annually until 10 years after completion of therapy.

      CT with contrast or MRI without and with contrast of the primary tumor site at 3, 6, 9, 12, 18, 24, 30, 36, 42, 48, 54, and 60 months, then annually until 10 years after completion of therapy. If negative at 10 years, no further advanced imaging is necessary.

      MIBG scan (CPT® 78800, 78801, 78802, 78803, or 78804) at 3, 6, 9, 12, 18, 24, 30, and 36 months after completion of therapy. If negative at 36 months, no further MIBG or PET imaging is necessary.

        ® Early detection of recurrence with 123I-MIBG has been shown to improve post-relapse outcomes in high risk neuroblastoma

      CT Chest is not indicated in asymptomatic surveillance imaging of neuroblastoma members with no prior history of thoracic disease.

      For members with suspected recurrence:

        ® CT with contrast, of the Chest/Abdomen/Pelvis (CPT® 71260, and CPT® 74177) or MRI without and with contrast, (CPT® 71552, CPT® 74183, and CPT® 72197) and other sites of prior measurable disease or current symptoms
        ® MIBG scan (CPT® 78800, 78801, 78802, 78803, or 78804)
        ® Urine HVA/VMA

      References
      1. Brodeur GM, Hogarty MD, Bagatell R, et al. Neuroblastoma. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:772-797.
      2. Shusterman S and George RE. Neuroblastoma. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1675-1713.
      3. Sharp SE, Gelfand MJ, Shulkin BL. Pediatrics: diagnosis of neuroblastoma. Semin Nucl Med. 2011;41(5):345-353. doi:10.1053/j.semnuclmed.2011.05.001.
      4. Kushner BH, Kramer K, Modak S, et al. Sensitivity of surveillance studies for detecting asymptomatic and unsuspected relapse of high-risk neuroblastoma. J Clin Oncol. 2009;27(7):1041-1046. doi:10.1200/JCO.2008.17.6107.
      5. Uslu L, Doing J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
      6. Nuchtern JG, London WB, Barnewolt CE, et al. A prospective study of expectant observation as primary therapy for neuroblastoma in young infants: A Children’s Oncology Group Study. Ann Surg. 2012 October;256(4):573-580. doi:10.1097/SLA.0b013e31826cbbbd.
      7. Brisse HJ, McCarville MB, Granata C, et al. Guidelines for imaging and staging of neuroblastic tumors: Consensus report from the International Neuroblastoma Risk Group Project. Radiology. 2011;261(1):243-257. doi:10.1148/radiol.11101352.
      8. Monclair T, Brodeur GM, Ambros PF, et al. The International Neuroblastoma Risk Group (INRG) staging system: an INRG task force report. J Clin Oncol. 2009;27(2):298-303. doi:10.1200/JCO.2008.16.6876.
      9. Cohn SL, Pearson ADJ, London WB, et al. The International Neuroblastoma Risk Group (INRG) classification system: an INRG Task Force Report. J Clin Oncol. 2009;27(2):289-297. doi:10.1200/JCO.2008.16.6785.
      10. GE Healthcare. AdreView™ Iobenguane I 123 Injection prescribing information. Revised September 2008. https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/22290lbl.pdf
      11. Bombardieri E, Giammarile F, Aktolun C, et al. 131I/123I-Metaiodobenzylguanidine (MIBG) scintigraphy: procedure guidelines for tumor imaging. Eur J Nucl Med Mol Imaging. 2010;37(12):2436-2446. http://snmmi.files.cms-plus.com/docs/hpra/2010_published_OC_131I_123I_Metaiodobenzylguanidine_Scintigraphy.pdf.
      12. Mueller WP, Coppenrath E, Pfluger T. Nuclear medicine and multimodality imaging of pediatric neuroblastoma. Pediatr Radiol. 2013;43(4):418-427. doi:10.1007/s00247-012-2512-1.
      13. Sharp SE, Gelfand MJ, Shulkin BL. Pediatrics: Diagnosis of neuroblastoma. Semin Nucl Med. 2011;41(5):345-353. doi:10.1053/j.semnuclmed.2011.05.001.
      14. McCarville MB. What MRI can tell us about neurogenic tumors and rhabdomyosarcoma. Pediatr Radiol. 2016;46(6):881-890. doi:10.1007/s00247-016-3572-4.
      15. Pinto NR, Applebaum MA, Volchenboum SL, et al. Advances in risk classification and treatment strategies for neuroblastoma. J Clin Oncol. 2015;33(27):3008-3017. doi:10.1200/JCO.2014.59.4648.
      16. Park JR, Bagatell R, Cohn SL, et al. Revisions to the International Neuroblastoma Response Criteria: A consensus statement from the National Cancer Institute clinical trials planning meeting. J Clin Oncol. 2017;35(22):2580-2587. doi:10.1200/JCO.2016.72.0177.
      17. Federico SM, Brady SL, Pappo A, et al. The role of chest computed tomography (CT) as a surveillance tool in children with high risk neuroblastoma. Pediatr Blood Cancer. 2015;62(6):976-981. doi:10.1002/pbc.25400.
      18. Gauguet J-M, Pace-Emerson T, Grant FD, et al. Evaluation of the utility of 99mTC-MDP bone scintigraphy versus MIBG scintigraphy and cross-sectional imaging for staging members with neuroblastoma. Pediatr Blood Cancer. 2017;64:e26601. doi:10.1002/pbc.26601.
      19. Venkatramani R, Pan H, Furman WL, et al. Multimodality treatment of pediatric esthesioneuroblastoma. Pediatr Blood Cancer. 2016;63(3):465-470. doi:10.1002/pbc.25817.
      20. Owens C, Li BK, Thomas KE, et al. Surveillance imaging and radiation exposure, in the detection of relapsed neuroblastoma. Pediatr Blood Cancer. 2016:63(10);1786-1793. doi:10.1002/pbc.26099.
      21. Allen-Rhoades W, Whittle SB, Rainusso N. Pediatric solid tumors of infancy: an overview. Pediatr In Rev. 2018;39(2):57-67. doi:10.1542/pir.2017-0057.

      PEDONC-7: Pediatric Renal Tumors

      PEDONC-7.1: Pediatric Renal Tumors – General Considerations
      PEDONC-7.2: Unilateral Wilms Tumor (UWT)
      PEDONC-7.3: Bilateral Wilms Tumor (BWT)
      PEDONC-7.4: Pediatric Renal Cell Carcinoma (RCC)
      PEDONC-7.5: Clear Cell Sarcoma of the Kidney (CCSK)
      PEDONC-7.6: Malignant Rhabdoid Tumor of the Kidney (MRT) and Other Extracranial Sites
      PEDONC-7.7: Congenital Mesoblastic Nephroma (CMN)

      PEDONC-7.1: Pediatric Renal Tumors – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Note: Some payors consider PET imaging to be investigational for the treatment of Wilms tumor and other kidney tumors, and those coverage policies may supersede the recommendations for PET imaging in this section.

      A variety of tumors can occur in the pediatric kidney, and include the following:

        ® Wilms Tumor
          ¡ Favorable Histology (FHWT)
          ¡ Focal Anaplasia (FAWT)
          ¡ Diffuse Anaplasia (DAWT)
          ¡ Bilateral Wilms Tumor (BWT)
        ® Renal Cell Carcinoma (RCC)
        ® Clear Cell Sarcoma of the Kidney (CCSK)
        ® Malignant Rhabdoid Tumor of the Kidney (MRT)
        ® Congenital Mesoblastic Nephroma (CMN)
        ® Other cancers occurring in the kidney:
          ¡ Neuroblastoma
          ¡ Primitive Neuroectodermal Tumor
          ¡ Rhabdomyosarcoma
          ¡ Non-Rhabdomyosarcoma Soft Tissue Sarcomas
          ¡ These and other rare tumors have been reported occurring primarily in the kidney and should be imaged according to the guidelines for the specific histologic diagnosis.
      PEDONC-7.2: Unilateral Wilms Tumor (UWT)

      For this condition imaging is medically necessary based on the following criteria:

      Unilateral Wilms Tumor Initial Staging:

      Many members will present with an asymptomatic abdominal mass, and will undergo ultrasound as a primary evaluation. Doppler ultrasound to evaluate for tumor thrombus is no longer necessary unless CT findings are inconclusive, and should not be performed if CT is already completed.

      CT Abdomen/Pelvis with contrast (CPT® 74177) is indicated for all unilateral Wilms tumor members

        ® If bilateral renal lesions are noted on ultrasound or CT, MRI Abdomen (CPT® 74183) and Pelvis (CPT® 72197) without and with contrast should be strongly considered for better characterization

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) should be completed prior to anesthesia exposure if possible

      MRI Brain without and with contrast (CPT® 70553) is indicated for initial staging for any member with neurologic signs or symptoms raising suspicion of CNS metastases as only ~0.5% of Wilms tumor members will ever develop brain metastases

      Bone scan (See PEDONC-1.3) is indicated for any member with signs or symptoms raising suspicion of bony metastases

      PET is not indicated in the initial staging of any pediatric renal tumor

      Unilateral Wilms Tumor Treatment Response:

      A very low risk subset of stage I FHWT will be observed after nephrectomy, and enter directly into surveillance.

      The majority of members will receive chemotherapy with or without XRT, beginning within 14 days of initial surgery.

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned therapy

      CT Abdomen/Pelvis with contrast (CPT® 74177) or MRI Abdomen (CPT® 74183) and Pelvis (CPT® 72197) without and with contrast can be performed every 2 cycles during treatment and at the end of planned therapy

      PET is not routinely utilized to assess treatment response in Wilms tumor.

        ® However, since most Wilms tumors are FDG-avid, rare circumstances may occur where PET imaging should be approved to establish the presence of active disease only when a major therapeutic decision depends on PET avidity. These requests will be forwarded for Medical Director review.
      Unilateral Wilms Tumor Surveillance Imaging:
      There are no data to support the use of PET imaging for routine surveillance in any member with Wilms tumor.

      Very low risk FHWT treated with nephrectomy only:

        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) OR CXR at 3, 6, 12, and 18 months after nephrectomy
        ® CT Abdomen and Pelvis with contrast (CPT® 74177) OR Ultrasound (CPT® 76700 and 76506) of Abdomen and Pelvis at 3, 6, 12, and 18 months after nephrectomy
        ® Surveillance pelvic imaging is indicated in this member group due to higher risk of recurrence in surgery only treatment
        ® Other surveillance imaging should be by Abdominal US (CPT® 76700) and CXR

      FHWT treated with chemotherapy with or without XRT:
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250)every 6 months for 3 years after completion of all therapy
        ® CT Abdomen with contrast (CPT® 74160), MRI Abdomen without and with contrast (CPT® 74183), or Ultrasound (CPT® 76700) of the Abdomen every 6 months for 3 years after completion of all therapy
        ® Pelvic imaging is not indicated for surveillance unless prior pelvic involvement has been documented or there was tumor rupture at diagnosis
        ® Other surveillance imaging should be by Abdominal US and CXR

      FAWT or DAWT treated with chemotherapy with or without XRT:
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 2 years after completion of all therapy
        ® CT Abdomen and Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) every 3 months for 2 years after completion of all therapy
        ® Other surveillance imaging should be by Abdominal US and CXR

      Surveillance imaging with CT of the Chest/Abdomen/Pelvis (CPT® 71260 and CPT® 74177) following successful treatment for recurrent unilateral Wilms tumor can be approved at every 3 months for 1 year after completing therapy for recurrence.
        ® Surveillance imaging later than 12 months after completing therapy for recurrence should follow the standard timing listed in this surveillance section.
      PEDONC-7.3: Bilateral Wilms Tumor (BWT)

      For this condition imaging is medically necessary based on the following criteria:

      Bilateral Wilms Tumor Initial Staging:

      Many members will present with an asymptomatic abdominal mass, and will undergo ultrasound as a primary evaluation. Doppler ultrasound to evaluate for tumor thrombus is no longer necessary unless CT findings are inconclusive, and should not be performed if CT is already completed.

      Members with bilateral Wilms Tumor may begin therapy without a histologic diagnosis to preserve a localized disease stage and attempt to shrink the tumors to allow for renal-sparing surgical approaches.

      MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) is the preferred imaging modality for members with bilateral Wilms tumor

        ® CT Abdomen and Pelvis with contrast (CPT® 74177) is often performed prior to discovery of bilateral lesions and should not prevent MRI from being approved
        ® CT Abdomen and Pelvis with contrast (CPT® 74177) may be used for members with a contraindication to MRI
          ¡ Avoidance of anesthesia exposure is not a contraindication to MRI for these members
      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is indicated in the initial workup of all pediatric renal tumors and should be completed prior to anesthesia exposure if possible

      MRI Brain without and with contrast (CPT® 70553) is indicated for initial staging for any member with neurologic signs or symptoms raising suspicion of CNS metastases as only ~0.5% of Wilms tumor members will ever develop brain metastases

      Bone scan (See PEDONC-1.3: Modality General Considerations) is indicated for any member with signs or symptoms raising suspicion of bony metastases

      PET is not indicated in the initial staging of any pediatric renal tumor

      Bilateral Wilms Tumor Treatment Response:

      MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be performed every 2 cycles during treatment and at the end of planned therapy

        ® CT Abdomen and Pelvis with contrast (CPT® 74177) may be used for members with a contraindication to MRI
        ® If treating with chemotherapy without a biopsy, disease evaluation is indicated at week 6. If either tumor has not shrunk 50%, then open biopsy is indicated to confirm favorable histology.
        ® If partial nephrectomy still not feasible at week 6, the next disease evaluation is at week 12. Surgical resection should occur no later than week 12.

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned therapy

      PET is not routinely utilized to assess treatment response in Wilms tumor.

        ® However, since most Wilms tumors are FDG-avid, rare circumstances may occur where PET should be approved to establish the presence of active disease only when a major therapeutic decision depends on PET avidity. These requests will be forwarded for Medical Director review.

      Bilateral Wilms Tumor Surveillance Imaging:

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 6 months for 3 years after completion of all therapy

      CT Abdomen with contrast (CPT® 74160) or MRI Abdomen without and with contrast (CPT®74183) every 6 months for 3 years after completion of therapy

        ® “Extra” one-time imaging is supported at 3 months after completion of all therapy because close surgical margins occur frequently in members undergoing nephron-sparing surgical approaches, and the risk for early local recurrence is higher

      Pelvic imaging is not indicated for surveillance unless prior pelvic involvement has been documented or there was tumor rupture at diagnosis

      Other surveillance imaging should be by Abdominal US (CPT® 76700) and CXR

        ® When CT or MRI Abdomen no longer indicated, members with bilateral Wilms tumor should have screening Abdominal ultrasound every 3 months until age 8

      Surveillance imaging with CT of the Chest/Abdomen/Pelvis (CPT® 71260 and CPT® 74177) following successful treatment for recurrent bilateral Wilms tumor can be approved every 3 months for 1 year after completing therapy for recurrence.
        ® Surveillance imaging later than 12 months after completing therapy for recurrence should follow the standard timing listed in this surveillance section.
      PEDONC-7.4: Pediatric Renal Cell Carcinoma (RCC)

      For this condition imaging is medically necessary based on the following criteria:

      A majority of pediatric cases have a novel subtype involving TFE3 or TFEB translocations, which have a different natural history than “adult type” RCC. Members of any age with TFE3 or TFEB translocated RCC should be imaged according to this guideline section.

      40 to 45% of pediatric RCC cases have similar histologies to adult RCC (clear cell, papillary, chromophobe, etc.) and imaging decisions will be similar to adult oncology guidelines. Members with all other subtypes of RCC should be imaged according to Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-17: Renal Cell Cancer (RCC).

      Pediatric Renal Cell Carcinoma Initial Staging:

      Many members will present with an asymptomatic abdominal mass, and will undergo ultrasound as a primary evaluation. Doppler ultrasound to evaluate for tumor thrombus is no longer necessary unless CT findings are inconclusive, and should not be performed if CT is already completed.

      CT Abdomen and Pelvis with contrast (CPT® 74177) is indicated in all members

        ® If bilateral renal lesions are noted on ultrasound or CT, MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) should be strongly considered

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made, by complete nephrectomy for most unilateral renal tumors and biopsy for bilateral renal tumors or inoperable unilateral tumors

      MRI Brain without and with contrast (CPT® 70553) is indicated for any member with neurologic signs or symptoms raising suspicion of CNS metastases

      Bone scan (See PEDONC-1.3: Modality General Considerations) is indicated for any member with signs or symptoms raising suspicion of bony metastases

      PET scan is not indicated in the initial staging of any pediatric renal tumor

      Pediatric Renal Cell Carcinoma Treatment Response:

      Most members will have surgical resection of all disease at the time of diagnosis and will enter directly into surveillance.

      Members with residual measurable disease after initial surgery and receiving adjuvant medical therapy can have CT Chest with (CPT® 71260) or without contrast (CPT® 71250) and CT Abdomen with contrast (CPT® 74160) every 2 cycles during active treatment

      Pelvic imaging is not indicated unless prior pelvic involvement has been documented

      PET is not routinely utilized to assess treatment response in pediatric RCC.

        ® However, since some RCC tumors are FDG-avid, rare circumstances may occur where PET should be approved to establish the presence of active disease only when a major therapeutic decision depends on PET avidity these requests will be forwarded for Medical Director review.

      Pediatric Renal Cell Carcinoma Surveillance Imaging:

      All pediatric RCC members:

        ® MRI Brain without and with contrast (CPT® 70553) every 6 months for 2 years after completion of all therapy only for members with documented CNS metastases or new signs/symptoms suggestive of CNS recurrence.

      TFE3 or TFEB subtype:
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 2 years, then every 6 months for 2 years after completion of all therapy
        ® CT Abdomen with contrast (CPT® 74160) or MRI Abdomen without and with contrast (CPT® 74183) every 3 months for 2 years, then every 6 months for 2 years after completion of all therapy
        ® Pelvic imaging is not indicated for surveillance unless prior pelvic involvement has been documented

      All other histologies:
        ® Surveillance imaging is appropriate as listed in the adult Oncology Imaging Guidelines: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-17.4: Renal Cell Cancer (RCC) – Surveillance
      PEDONC-7.5: Clear Cell Sarcoma of the Kidney (CCSK)

      For this condition imaging is medically necessary based on the following criteria:

      Be careful not to confuse the diagnosis with clear cell RCC. See Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-17: Renal Cell Cancer (RCC) for imaging guidelines.


      Clear Cell Sarcoma Of The Kidney Initial Staging:

      Many members will present with an asymptomatic abdominal mass, and will undergo ultrasound as a primary evaluation. Doppler ultrasound to evaluate for tumor thrombus is no longer necessary unless CT findings are inconclusive, and should not be performed if CT is already completed.

      CT Abdomen and Pelvis with contrast (CPT® 74177) is indicated in all members

        ® If bilateral renal lesions are noted on ultrasound or CT, MRI Abdomen and Pelvis without and with contrast should be strongly considered

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made, by complete nephrectomy for most unilateral renal tumors and biopsy for bilateral renal tumors or inoperable unilateral tumors

      MRI Brain without and with contrast (CPT® 70553) is indicated for initial staging in all members with clear cell sarcoma of the kidney

      Bone scan (See PEDONC-1.3: Modality General Considerations) is indicated in all members with clear cell sarcoma of the kidney

      PET is not indicated in the initial staging of any pediatric renal tumor

      Clear Cell Sarcoma Of The Kidney Treatment Response:

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned therapy

      CT Abdomen and Pelvis with contrast (CPT® 4177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be performed every 6 weeks during treatment and at the end of planned therapy

      MRI Brain without and with contrast (CPT® 70553) can be performed:

        ® Every 2 cycles during treatment for members with CNS metastases at initial staging
        ® At the end of planned therapy for all members with CCSK

      Bone scan (See PEDONC-1.3: Modality General Considerations) at the end of planned therapy

      PET is not routinely utilized to assess treatment response in CCSK

        ® However, since clear cell sarcomas have been shown to be FDG-avid in other anatomic locations, rare circumstances may occur where PET should be approved to establish the presence of active disease only when a major therapeutic decision depends on PET avidity. These requests will be forwarded for Medical Director review.

      Clear Cell Sarcoma Of The Kidney Surveillance Imaging:

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 2 years after completion of all therapy

      CT Abdomen and Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) every 3 months for 2 years after completion of all therapy

      MRI Brain without and with contrast (CPT® 70553) every 6 months for 3 years after completion of all therapy

      Bone scan (See PEDONC-1.3: Modality General Considerations) every 3 months for 1 year, then every 6 months for 1 year after completion of all therapy

        ® If negative at 36 months, no further advanced imaging is necessary.

      Other surveillance imaging should be by Abdominal US (CPT® 76700) and CXR

      PEDONC-7.6: Malignant Rhabdoid Tumor of the Kidney (MRT) and Other Extracranial Sites

      For this condition imaging is medically necessary based on the following criteria:

      Be careful not to confuse the diagnosis with rhabdomyosarcoma. See PEDONC-8.2: Rhabdomyosarcoma (RMS) for Imaging Guidelines.

      A highly aggressive histologic variant that can also occur in other locations and all non-CNS sites should follow these guidelines.

      Primary CNS rhabdoid malignancies should be imaged according to PEDONC-4.5: Atypical Teratoid/Rhabdoid Tumors (ATRT).

      Malignant Rhabdoid Tumor Initial Staging:

      Many members will present with an asymptomatic abdominal mass, and will undergo ultrasound as a primary evaluation. Doppler ultrasound to evaluate for tumor thrombus is no longer necessary unless CT findings are inconclusive, and should not be performed if CT is already completed.

      CT Abdomen and Pelvis with contrast (CPT® 74177) is indicated in all members

        ® If bilateral renal lesions are noted on ultrasound or CT, MRI Abdomen and Pelvis without and with contrast should be strongly considered

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made, by complete nephrectomy for most unilateral renal tumors and biopsy for bilateral renal tumors or inoperable unilateral tumors

      MRI Brain without and with contrast (CPT® 70553) is indicated for all members with MRT of the kidney or other non-CNS site

      Bone scan (See PEDONC-1.3: Modality General Considerations) is indicated in all members with MRT of the kidney or other non-CNS site

      PET is not indicated in the initial staging of any pediatric renal tumor

      Malignant Rhabdoid Tumor Treatment Response:

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned therapy

      CT Abdomen and Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be performed every 2 cycles during treatment and at the end of planned therapy

        ® If primary site other than kidney, perform CT with contrast or MRI without and with contrast of primary site in place of abdominal and pelvic imaging

      MRI Brain without and with contrast (CPT® 70553) can be performed:
        ® Every 2 cycles during treatment for members with CNS metastases at initial staging
        ® At the end of planned therapy for all members with MRT

      Bone scan (See PEDONC-1.3: Modality General Considerations) at the end of planned therapy only if positive at initial diagnosis

      PET is not routinely utilized to assess treatment response in MRT.

        ® However, since malignant rhabdoid tumors have been shown to be FDG-avid, rare circumstances may occur where PET should be approved to establish the presence of active disease only when a major therapeutic decision depends on PET avidity. These requests will be forwarded for Medical Director review.

      Malignant Rhabdoid Tumor Surveillance Imaging:

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 2 years after completion of all therapy

      CT Abdomen and Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) every 3 months for 3 years after completion of all therapy

        ® If primary site other than kidney, perform CT with contrast or MRI without and with contrast of primary site in place of abdominal imaging

      MRI Brain without and with contrast (CPT® 70553) every 3 months for 1 year, then every 6 months for 1 year after completion of all therapy

      Bone scan (See PEDONC-1.3: Modality General Considerations) every 3 months for 1 year, then every 6 months for 1 year after completion of all therapy only if positive at initial diagnosis

        ® If negative at 36 months, no further advanced imaging is necessary

      Other surveillance imaging should be by Abdominal US (CPT® 76700) and CXR

      PEDONC-7.7: Congenital Mesoblastic Nephroma (CMN)

      For this condition imaging is medically necessary based on the following criteria:

      This is the most common primary renal tumor occurring in young infants, and the overall prognosis is very good.

      Complete surgical removal is curative in most cases, and histologically confirmed metastatic disease or bilateral disease has never been reported.

      Congenital Mesoblastic Nephroma Initial Staging

      Many members will present with an asymptomatic abdominal mass at the time of birth or abnormal prenatal ultrasound, and will undergo ultrasound as a primary evaluation.

      CT Abdomen and Pelvis with contrast (CPT® 74177) is indicated in all members

      CT Chest with (CPT® 71260) can be approved to evaluate inconclusive findings on Chest X-ray

      PET is not indicated in the initial staging of any pediatric renal tumor

      Congenital Mesoblastic Nephroma Treatment Response:

      Surgical resection is curative in most members. Children who have resection of the tumor can have a single CT Abdomen and Pelvis with contrast (CPT® 74177) approved following resection to establish baseline imaging and those with a complete resection should then be imaged according to surveillance guidelines

      Some members will receive preoperative chemotherapy to facilitate safer resection and can have CT Abdomen and Pelvis with contrast (CPT® 74177) approved every 2 cycles of therapy until surgery, and should then be imaged according to surveillance guidelines after their postoperative baseline imaging study

      Congenital Mesoblastic Nephroma Surveillance Imaging

      Recurrences are rare, but most occur within 12 months of diagnosis

      Given the young age of the members with CMN, ultrasound is the preferred surveillance imaging modality to avoid radiation and anesthesia exposures

        ® CT Abdomen and Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be approved every 3 months for 1 year after completion of all therapy for members with residual abnormalities present on post-operative imaging or inconclusive findings on ultrasound

      References

      1. Fernandez C, Geller JI, Ehrlich PF, et al. Renal tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:753-771.
      2. Dome JS, Mullen EA, and Argani P. Pediatric renal tumors. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:1714-1746.
      3. Kaste SC, Brady SL, Yee B, et al. Is routine pelvic surveillance imaging necessary in patients with Wilms tumor? Cancer. 2013;119(1):182-188. doi:10.1002/cncr.27687.
      4. Malkan AD, Loh A, Bahrami A, et al. An approach to renal masses in pediatrics. Pediatrics. 2015;135(1):142-158. doi:10.1542/peds.2014-1011.
      5. Khanna G, Rosen N, Anderson JR, et al. Evaluation of diagnostic performance of CT for detection of tumor thrombus in children with Wilms tumor: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2012;58(4):551-555. doi:10.1002/pbc.23222.
      6. Kieran K and Ehrlich PF. Current surgical standards of care in Wilms tumor. Urol Oncol. 2016;34(1):13-23. doi:10.1016/j.urolonc.2015.05.029.
      7. Moinul Hossain AK, Shulkin BL, Gelfand MJ, et al. FDG positron emission tomography/computed tomography studies of Wilms’ tumor. Eur J Nucl Med Mol Imaging. 2010;37(7):1300-1308. doi:10.1007/s00259-010-1396-2.
      8. Misch D, Steffen IG, Schönberger S, et al. Use of positron emission tomography for staging, preoperative response assessment and post therapeutic evaluation in children with Wilms tumor. Eur J Nucl Med Mol Imaging. 2008;35(9):1642-1650. doi:10.1007/s00259-008-0819-9.
      9. Servaes S, Khanna G, Naranjo A, et al. Comparison of diagnostic performance of CT and MRI for abdominal staging of pediatric renal tumors: a report from the Children’s Oncology Group. Pediatr Radiol. 2015;45(2):166-172. doi:10.1007/s00247-014-3138-2.
      10. Uslu L, Doing J, Link M, Rosenburg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for Evaluation of Pediatric Malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
      11. Dome JS, Graf N, Geller JI, et al. Advances in Wilms tumor treatment and biology: progress through international collaboration. J Clin Oncol. 2015;33(27):2999-3007. doi:10.1200/JCO.2015.62.1888.
      12. Venkatramani R, Chi Y-Y, Coppes MJ, et al. Outcome of patients with intracranial relapse enrolled on National Wilms Tumor Study Group clinical trials. Pediatr Blood Cancer. 2017;64(7):e26406. doi:10.1002/pbc.26406.
      13. Mullen EA, Chi Y-Y, Hibbitts E, et al. Impact of surveillance imaging modality on survival after recurrence in patients with favorable-histology Wilms tumor: a report from the Children’s Oncology Group. J Clin Oncol. 2018;36:3396-3403. doi:10.1200/JCO.18.00076.
      14. Ehrlich P, Chi Y-Y, Chintagumpala MM, et al. results of the first prospective multi-institutional treatment study in children with bilateral Wilms tumor (AREN0534): a report from the Children’s Oncology Group. Ann Surg. 2017;266(3):470-478. doi:10.1097/SLA.0000000000002356.
      15. Geller JI, Ehrlich PF, Cost NG, et al. Characterization of adolescent and pediatric renal cell carcinoma: a report from the Children’s Oncology Group study AREN03B2. Cancer. 2015 July; 121(14):2457-2464. doi:10.1002/cncr.29368.
      16. Rialon KL, Gulack BC, Englum BR, Routh JC, Rice HE. Factors impacting survival in children with renal cell carcinoma. J Pediatr Surg. 2015;50(6):1014-1018. doi:10.1016/j.jpedsurg.2015.03.027.
      17. Young EE, Brown CTG, Merguerian PA, Akhavan A. Pediatric and adolescent renal cell carcinoma. Urol Oncol. 2016;34(1):42-49. doi:10.1016/j.urolonc.2015.06.009.
      18. Gooskens SL, Houwing ME, Vujanic GM, et al. Congenital mesoblastic nephroma 50 years after its recognition: a narrative review. Pediatr Blood Cancer. 2017; 64(7):e26437. doi:10.1002/pbc.26437

      PEDONC-8: Pediatric Soft Tissue Sarcomas

      PEDONC-8.1: Pediatric Soft Tissue Sarcomas – General Considerations
      PEDONC-8.2: Rhabdomyosarcoma (RMS)
      PEDONC-8.3: Non-Rhabdomyosarcoma Soft Tissue Sarcomas (NRSTS)

      PEDONC-8.1: Pediatric Soft Tissue Sarcomas – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Note: Some payors consider PET to be investigational for the treatment of rhabdomyosarcoma and other soft tissue sarcomas, and those coverage policies may supersede the recommendations for PET in this section.

      Soft tissue sarcomas occur in both adult and pediatric members, but some histologic types are more common in one age group than the other. Unless specified below, members who are <18 years old should be imaged according to this guideline section. Exceptions include:

      Rhabdomyosarcoma members of all ages should be imaged according to guidelines in PEDONC-8.2: Rhabdomyosarcoma (RMS)

      Kaposi’s sarcoma members of all ages should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-31.10: Kaposi’s Sarcoma

      Pediatric soft tissue sarcomas are divided into two groups:


        1. Rhabdomyosarcoma (RMS) accounts for ~60% of soft tissue sarcomas in young members, but only ~25% of soft tissue sarcomas in adolescents

        2. Non-rhabdomyosarcoma soft tissue sarcomas (NRSTS) which encompasses all other histologic subtypes


      Evaluation of soft tissue masses of uncertain nature prior to biopsy should follow general imaging guidelines in Pediatric Musculoskeletal Imaging Policy (Policy # 164 in the Radiology Section); PEDMS-3: Soft Tissue and Bone Masses for members who are age 0 (newborn) through 17 years old, and Adult Musculoskeletal Imaging Policy (Policy #152 in the Radiology Section); MS-10: Soft Tissue Mass Or Lesion Of Bone for members who are ≥18 years old.


      PEDONC-8.2: Rhabdomyosarcoma (RMS)

      For this condition imaging is medically necessary based on the following criteria:

      Rhabdomyosarcoma Initial Staging:

      Because RMS can arise from any muscle tissue, the presenting symptoms and primary tumor sites vary widely and strongly influence the appropriate imaging decisions

        ® Either CT with contrast or MRI without and with contrast is acceptable for primary site imaging of RMS arising in the abdomen or pelvis at the discretion of the treating oncologist.
        ® CT with contrast is the preferred primary site imaging modality for RMS arising in the thoracic cavity (not the chest wall).
        ® MRI without and with contrast is the preferred primary site imaging modality for RMS occurring in all other anatomic locations, including the chest wall.

      Evaluation for lung metastases using CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is indicated in the initial workup of all pediatric soft tissue sarcomas and should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made

        ® PET/CT is superior to conventional imaging for detection of nodal and bony metastases in pediatric RMS and is indicated in the initial staging of all members after histologic diagnosis is established
          ¡ Whole body PET/CT (CPT® 78816) is the preferred study for initial staging of RMS
          ¡ Bone scan (See PEDONC-1.3: Modality General Considerations) may be substituted for PET imaging if PET not available
        ® CT Abdomen and Pelvis with contrast (CPT® 74177) is not routinely indicated in the initial metastatic staging of all members with pediatric RMS, but can be approved in the following situations:
          ¡ Evaluation of inconclusive PET findings
          ¡ Primary site of abdomen or pelvis
          ¡ Lower extremity primary sites
        ® MRI Brain (CPT® 70553) and Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) is indicated for initial staging in the following pediatric RMS:
          ¡ Primary site involving the paraspinal or paravertebral region
          ¡ PET or bone scan-avid lesions in skull, neck, vertebrae
          ¡ Any member with neurologic signs or symptoms raising suspicion of CNS metastases
      Rhabdomyosarcoma Treatment Response:

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned therapy for all members

      Primary site imaging:

        ® CT with contrast or MRI without and with contrast can be performed every 2 cycles during treatment and at the end of planned therapy
        ® Restaging imaging is appropriate after local control surgery (complete or partial resection) is completed

      Metastatic site imaging:
        ® Repeat imaging of all known metastatic sites using the same modality as during initial staging is appropriate whenever primary site imaging is necessary

      PET is not routinely utilized to assess treatment response in RMS, but is indicated in the following circumstances:
        ® Response assessment prior to local control surgery or radiation therapy
        ® Evaluation of residual mass visible on conventional imaging as part of end of therapy evaluation
        ® Response assessment of disease visible on PET but not conventional imaging
        ® Once PET has been documented to be negative for a given member’s cancer or all PET-avid disease has been surgically resected, PET should not be used for continued disease monitoring or surveillance unless one of the exceptions in section PEDONC-1: General Guidelines applies. These requests will be forwarded for Medical Director review.
        ® PET is generally not indicated during active treatment for recurrent pediatric cancer. In rare circumstances, PET may be appropriate when results are likely to result in a treatment change for the member, including a change from active treatment to surveillance. These requests will be forwarded for Medical Director review.

      Rhabdomyosarcoma Surveillance Imaging:

      All members with localized RMS:

        ® Primary tumor site should be imaged with either CT with contrast or MRI without and with contrast every 3 months for 1 year, then every 4 months for 2 years, then every 6 months for 1 year after completion of all therapy
        ® CXR every 3 months for 1 year, then every 4 months for 2 years, then every 6 months for 1 year after completion of all therapy
          ¡ CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is indicated for new or worsening clinical symptoms of chest disease or new findings on CXR
      All members with metastatic RMS:
        ® Primary tumor site should be imaged with either CT with contrast or MRI without and with contrast every 3 months for 1 year, then every 4 months for 2 years, then every 6 months for 1 year after completion of all therapy
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) and all known metastatic sites every 3 months for 1 year, then every 4 months for 2 years, then every 6 months for 1 year after completion of all therapy
        ® Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) should be used for surveillance of known bony metastases every 3 months for 1 year, then every 4 months for 2 years, then every 6 months for 1 year after completion of all therapy

      PET should not be used for surveillance imaging of RMS unless one of the following applies:
        ® Conventional imaging (CT, MRI, US, plain film) reveals findings that are inconclusive or suspicious for recurrence and PET avidity will determine whether biopsy or continued observation is appropriate
          ¡ Residual mass that has not changed in size since the last conventional imaging does not justify PET imaging
          ¡ PET avidity in a residual mass at the end of planned therapy is not an indication for PET imaging during surveillance.
        ® Rare circumstances where obvious clinical symptoms show strong evidence suggesting recurrence and PET would replace conventional imaging modalities. These requests will be forwarded for Medical Director review.
      PEDONC-8.3: Non-Rhabdomyosarcoma Soft Tissue Sarcomas (NRSTS)

      For this condition imaging is medically necessary based on the following criteria:

      All soft tissue sarcomas other than RMS fall into this category.

      NRSTS Initial Staging:

      Because soft tissue sarcomas can arise from any soft tissue, the presenting symptoms and primary tumor sites vary widely and strongly influence the appropriate imaging decisions.

        ® Either CT with contrast or MRI without and with contrast is acceptable for primary site imaging of NRSTS arising in the abdomen or pelvis at the discretion of the treating oncologist.
        ® CT with contrast is the preferred primary site imaging modality for NRSTS arising in the thoracic cavity (not the chest wall).
        ® MRI without and with contrast is the preferred primary site imaging modality for NRSTS occurring in all other anatomic locations, including the chest wall.

      In addition, evaluation for lung metastases using CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is indicated in the initial workup of all pediatric soft tissue sarcomas and should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made:

        ® PET/CT (CPT® 78815) may be considered in the following:
          ¡ Desmoplastic small round cell tumor
          ¡ Prior to neoadjuvant chemotherapy
          ¡ Evaluating inconclusive findings found on conventional imaging
          ¡ Whole body PET/CT (CPT® 78816) may be approved if there is clinical suspicion of skull or distal lower extremity involvement
        ® Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) is used to evaluate for bony metastases but should be omitted if PET is performed
        ® CT Abdomen and Pelvis with contrast (CPT® 74177) is not routinely indicated in the initial metastatic staging of pediatric NRSTS, but can be approved in the following situations:
          ¡ Evaluation of inconclusive PET findings
          ¡ Primary site of abdomen or pelvis
          ¡ Lower extremity primary sites
          ¡ Desmoplastic small round cell tumor
        ® MRI Brain (CPT® 70553) and Spine (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) without and with contrast is indicated for initial staging in the following pediatric NRSTS:
          ¡ Primary site of paraspinal or paravertebral region
          ¡ PET or nuclear bone scan-avid lesions in skull, neck, vertebrae
          ¡ Any member with neurologic signs or symptoms raising suspicion of CNS metastases
      NRSTS Treatment Response:

      Many members with NRSTS will be treated with surgical resection alone, and these members enter immediately into surveillance

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned therapy

      Primary site imaging:

        ® CT with contrast or MRI without and with contrast can be performed every 2 cycles during treatment and at the end of planned therapy
        ® Restaging imaging is appropriate after local control surgery (complete or partial resection) is completed

      Metastatic site imaging:
        ® Repeat imaging of all known metastatic sites using the same modality as during initial staging is appropriate whenever primary site imaging is necessary

      PET imaging is not routinely utilized to assess treatment response in NRSTS, but is indicated in the following circumstances if positive at initial diagnosis.
        ® Response assessment prior to local control surgery or radiation therapy
        ® Evaluation of residual mass visible on conventional imaging as part of end of therapy evaluation
        ® Response assessment of disease visible on PET but not conventional imaging
        ® Once PET has been documented to be negative for a given member’s cancer or all PET-avid disease has been surgically resected, PET should not be used for continued disease monitoring or surveillance unless one of the exceptions in section PEDONC-1: General Guidelines applies. These requests will be forwarded for Medical Director review.
        ® PET imaging is generally not indicated during active treatment for recurrent pediatric cancer. In rare circumstances, PET imaging may be appropriate when results are likely to result in a treatment change for the member, including a change from active treatment to surveillance. These requests will be forwarded for Medical Director review.

      Surveillance Imaging:

      All members with localized NRSTS:

        ® Primary site should be imaged with either CT with contrast or MRI without and with contrast every 6 months for 5 years after completion of all therapy
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 6 months for 5 years after completion of all therapy

      All members with metastatic NRSTS:
        ® Primary site should be imaged with either CT with contrast or MRI without and with contrast every 6 months for 5 years after completion of all therapy
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) and all known metastatic sites every 6 months for 5 years after completion of all therapy
        ® Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) should be used for surveillance of known bony metastases every 6 months for 5 years after completion of all therapy

      Surveillance after recurrence:
        ® Surveillance imaging using CT Chest (CPT® 71260) and CT with contrast or MRI without and with contrast of the primary site following successful treatment for recurrent NRSTS can be approved every 3 months for 1 year after completing therapy for recurrence.
          ¡ Surveillance imaging later than 12 months after completing therapy for recurrence should follow the standard timing listed in this surveillance section.
      PET should not be used for surveillance imaging of NRSTS unless one of the following applies:
        ® Conventional imaging (CT, MRI, US, plain film) reveals findings that are inconclusive or suspicious for recurrence and PET avidity will determine whether biopsy or continued observation is appropriate
          ¡ Residual mass that has not changed in size since the last conventional imaging does not justify PET
          ¡ PET avidity in a residual mass at the end of planned therapy is not an indication for PET imaging during surveillance.
        ® Rare circumstances where obvious clinical symptoms show strong evidence suggesting recurrence and PET would replace conventional imaging modalities
        ® These requests will be forwarded for Medical Director review.

      References

      1. Wexler LH, Skapek SX, Helman LJ. Rhabdomyosarcoma. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:798-826.
      2. Spunt SL, Million L, and Coffin C. The Nonrhabdomyosarcoma soft tissue sarcomas. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:827-854.
      3. Womer RB, Barr FG, Linardic CM. Rhabdomyosarcoma. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:1906-1945.
      4. Davis IJ, Perez-Atayde AR, Fisher DE. Nonrhabdomyosarcoma soft tissue sarcomas and other soft tissue tumors. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:1946-1982.
      5. McCarville MB. What MRI can tell us about neurogenic tumors and rhabdomyosarcoma. Pediatr Radiol. 2016;46(6):881-890. doi:10.1007/s00247-016-3572-4.
      6. Federico SM, Spunt SL, Krasin MJ, et al. Comparison of PET-CT and Conventional Imaging in staging pediatric rhabdomyosarcoma. Pediatr Blood Cancer. 2013;60(7):1128-1134. doi:10.1002/pbc.24430.
      7. Dharmarajan KV, Wexler LH, Gavane S, et al. Positron Emission Tomography (PET) evaluation after initial chemotherapy and radiation therapy predicts local control in rhabdomyosarcoma. Int J Radiat Biol Oncol Phys. 2012;84(4):996-1002. doi:10.1016/j.ijrobp.2012.01.077.
      8. Arnold MA, Anderson JA, Gastier-Foster JM, et al. Histology, fusion status, and outcome in alveolar rhabdomyosarcoma with low-risk clinical features: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2016;63(4):634-639. doi:10.1002/pbc.25862.
      9. Völker T, Denecke T, Steffen I, et al. Positron Emission Tomography for staging of pediatric sarcoma patients: results of a prospective multicenter trial. J Clin Oncol. 2007; 25(34):5435-5441. doi:10.1200/JCO.2007.12.2473.
      10. Ostermeier A, McCarville MB, Navid F, et al. FDG PET/CT imaging of desmoplastic small round cell tumor: findings at staging, during treatment and at follow-up. Pediatr Radiol. 2015;45(9):1308-1315. doi:10.1007/s00247-015-3315-y.
      11. Uslu L, Doing J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for evaluation of pediatric malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
      12. Roberts CC, Kransdorf MJ, Beaman FD, et al. Follow-Up of malignant or aggressive musculoskeletal tumors. ACR Appropriateness Criteria® 2015:1-15. https://acsearch.acr.org/docs/69428/Narrative/.
      13. Ferrari A, Chi Y-Y, De Salvo GL, et al. Surgery alone is sufficient therapy for children and adolescents with low-risk synovial sarcoma: a joint analysis from the European paediatric soft tissue sarcoma Study Group and the Children’s Oncology Group. Eur J Cancer. 2017;78:1-6. doi:10.1016/j.ejca.2017.03.003.

      PEDONC-9: Bone Tumors

      PEDONC-9.1: Bone Tumors – General Considerations
      PEDONC-9.2: Benign Bone Tumors
      PEDONC-9.3: Osteogenic Sarcoma (OS)
      PEDONC-9.4: Ewing Sarcoma Family of Tumors (ESFT), Including Primitive Neuroectodermal Tumors (PNET)

      PEDONC-9.1: Bone Tumors – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      These guidelines include both benign and malignant lesions.

      Bone tumors occur in both adult and pediatric members, but some are more common in one age group than the other. Unless specified below, members who are < 18 years old should be imaged according to this guideline section. Exceptions include:

        ® Osteogenic sarcoma members of all ages should be imaged according to guidelines in PEDONC-9.3: Osteogenic Sarcoma (OS)
        ® Ewing sarcoma and primitive neuroectodermal tumor members of all ages should be imaged according to guidelines in PEDONC-9.4: Ewing Sarcoma and Primitive Neuroectodermal Tumors (ESFT).
        ® Chondrosarcoma members of all ages should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-12.6: Bone Sarcomas – Initial Work-up/Staging
        ® Chordoma members of all ages should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-12.6: Bone Sarcomas – Initial Work-up/Staging
        ® Giant cell tumor of bone and enchondroma members of all ages should be imaged according to guidelines in Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-12.9: Benign Bone Tumors – General Considerations
        ® Other benign bone tumor members of all ages should be imaged according to guidelines in PEDONC-9.2: Benign Bone Tumors

      All bone tumors should be evaluated by plain x-ray prior to any advanced imaging.

      PET does not reliably distinguish between benign and malignant bone tumors and should not be performed prior to biopsy.

      PEDONC-9.2: Benign Bone Tumors

      For this condition imaging is medically necessary based on the following criteria:

      Osteochondroma

        ® Plain x-ray appearance is diagnostic for the majority of members and advanced imaging is generally unnecessary
        ® MRI without and with contrast can be approved after evaluation by the operating surgeon for preoperative planning
        ® MRI without contrast OR without and with contrast, as requested, is appropriate for members with osteochondroma when there is clinical concern for malignant transformation based on new or worsening pain symptoms or a change on a recent plain x-ray

      Osteoid osteoma
        ® CT without contrast is often the primary study when osteoid osteoma is suspected based on clinical history and plain film findings
        ® Bone scan SPECT (CPT® 78803) is indicated for suspected osteoid osteoma
        ® Some members will require both CT without contrast as well as MRI without and with contrast to make a definitive diagnosis

      Other benign tumors
        ® Variety of diagnoses, including osteoid osteoma, osteoblastoma, aneurysmal bone cysts, fibrous dysplasia, chondroblastoma and others,
        ® Plain x-ray appearance is diagnostic for many benign bone tumors and advanced imaging is generally unnecessary except for preoperative planning
        ® MRI without and with contrast is the primary modality for advanced imaging of bone tumors, and can be approved to help narrow differential diagnoses and determine whether biopsy is indicated
          ¡ For certain tumors, CT (contrast as requested) provides better visualization of specific bony details, and requests after evaluation by the operating surgeon for preoperative planning should generally be approved
      Surveillance imaging, when indicated, should utilize plain x-ray
        ® Some benign bone tumor types carry a risk of malignant degeneration over time, but routine advanced imaging surveillance has not been shown to improve outcomes for these members
        ® MRI without and with contrast can be approved to evaluate new findings on plain X-ray or new/worsening clinical symptoms not explained by a recent plain x-ray

      There are no data to support the use of PET in the evaluation of benign bone tumors, and PET requests should not be approved without biopsy confirmation of a malignancy.

      PEDONC-9.3: Osteogenic Sarcoma (OS)

      For this condition imaging is medically necessary based on the following criteria:

      Osteogenic Sarcoma Initial Staging:

      All bone tumors should be evaluated by plain x-ray prior to any advanced imaging

      MRI without and with contrast is the preferred primary site imaging

        ® CT, contrast as requested, can be approved if there is a contraindication to MRI or if requested after evaluation by the operating surgeon to clarify inconclusive MRI findings for preoperative planning
        ® MRA and/or CTA may rarely be indicated for complicated surgical resections, and can be approved after evaluation by the operating surgeon to clarify inconclusive MRI findings for preoperative planning
        ® Requests for CT, MRA, or CTA should be forwarded for Medical Director review

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is superior to PET/CT for the detection of pulmonary metastases, and is indicated in the initial workup of all suspected malignant bone tumors and should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made, initially by biopsy, as definitive resection is usually performed after neoadjuvant chemotherapy

        ® Distant bony metastases are rare in OS, but cause a significant change in treatment approach.
        ® Whole body PET/CT (CPT® 78816) is the preferred study for initial staging of OS after histologic diagnosis is established
          ¡ PET has superior sensitivity to bone scan (95% vs. 76%) but equivalent overall diagnostic accuracy (98% vs. 96%) for detection of bony metastases in pediatric OS
          ¡ Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) may be substituted for PET imaging if PET not available
          ¡ If PET/CT is negative at initial diagnosis, bone scan (See PEDONC-1.3: Modality General Considerations) is preferred for asymptomatic surveillance for bony metastases at time points after local control surgery
      CT Abdomen and Pelvis with contrast (CPT® 74177) is not routinely indicated in the initial metastatic staging of pediatric OS, but can be approved in the following situations:
          ¡ Evaluation of inconclusive PET findings
          ¡ Primary site of abdomen or pelvis
      Osteogenic Sarcoma Treatment Response:

      Most OS members undergo restaging after 10 to 12 weeks of neoadjuvant chemotherapy prior to local control surgery to confirm the absence of progressive disease prior to the extended break necessary for postoperative healing.

      Restaging at this time point should include:

        ® MRI without and with contrast of primary site
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250)
        ® Whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations)

      Following local control surgery, the following imaging guidelines should be used until the end of planned chemotherapy:
        ® MRI without and with contrast of primary site ~6 weeks after surgical procedure and at the end of planned chemotherapy
        ® Plain x-rays of the primary site and chest every 2 months
        ® CT Chest (with or without contrast, as requested):
          ¡ Measurable pulmonary metastases: every 6 weeks and at the end of planned chemotherapy
          ¡ No measurable pulmonary metastases: every 4 months and at the end of planned chemotherapy
        ® Bone scan (See PEDONC-1.3: Modality General Considerations) every 4 months and at the end of planned chemotherapy
          ¡ Whole body PET/CT can be used in place of bone scan, if positive for distant bone metastases at initial diagnosis
      Members with metastatic disease do not routinely undergo local control surgery unless metastatic disease has resolved with chemotherapy.
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned chemotherapy
        ® MRI without and with contrast of primary site can be performed every 2 cycles during treatment and at the end of planned chemotherapy
        ® If previously positive for bony metastases, whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations) every 2 cycles during treatment and at the end of planned chemotherapy
        ® Imaging may be indicated more frequently around the time of surgical resection of primary or metastatic lesions to assess for resectability

      PET is generally not indicated during active treatment for recurrent pediatric cancer. In rare circumstances, PET imaging may be appropriate when results are likely to result in a treatment change for the member, including a change from active treatment to surveillance. These requests will be forwarded for Medical Director review.

      Osteogenic Sarcoma Surveillance Imaging:

      Appendicular bone primary tumor site:

        ® Plain x-rays of the primary tumor site should be completed every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy
        ® MRI is not routinely indicated for surveillance imaging of appendicular primary sites but should be approved for the following:
          ¡ The member does not have an endoprosthesis that will cause MRI or CT artifact
          ¡ To clarify inconclusive findings on plain x-ray
          ¡ To evaluate significant pain symptoms suggestive of primary site recurrence
      Axial bone primary tumor site:
        ® MRI without and with contrast of the primary tumor site can be approved every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy

      Metastatic disease surveillance:
        ® Members with localized OS:
          ¡ CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 1 year then every 4 months for 1 year after completion of all therapy
            § Chest X-ray (CXR) should be used for pulmonary recurrence surveillance after 24 months, and CT Chest can be approved to clarify inconclusive CXR findings
        ® Members with metastatic or recurrent OS:
          ¡ CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy
        ® Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) should be used for evaluation of distant bony metastases every 3 months for 1 year, then every 6 months for 2 years, then annually for 2 years after completion of all therapy
        ® PET/CT has no established role for asymptomatic surveillance of OS, but can be approved in the following circumstances:
          ¡ Conventional imaging reveals findings that are inconclusive or suspicious for recurrence and PET avidity will determine whether biopsy or continued observation is appropriate
          ¡ Rare circumstances where obvious clinical symptoms show strong evidence suggesting recurrence and PET would replace conventional imaging modalities
          ¡ Restaging after biopsy-confirmed recurrence
          ¡ These requests will be forwarded for Medical Director review.
      PEDONC-9.4: Ewing Sarcoma Family of Tumors (ESFT), Including Primitive Neuroectodermal Tumors (PNET)

      For this condition imaging is medically necessary based on the following criteria:

      ESFT Initial Staging:

      All bone tumors should be evaluated by plainx-ray prior to any advanced imaging

      ESFT can also occur in the soft tissues, soft tissue masses without bony involvement that are ill-defined or non-discrete should be evaluated by limited ultrasound prior to any advanced imaging.

      MRI without and with contrast is the preferred primary site imaging

        ® CT, contrast as requested, can be approved if there is a contraindication to MRI or if requested after evaluation by the operating surgeon to clarify inconclusive MRI findings for preoperative planning
        ® MRI Chest without and with contrast is indicated for chest wall primary tumors, in addition to the CT Chest for pulmonary metastasis detection
        ® MRA and/or CTA may rarely be indicated for complicated surgical resections, and can be approved after evaluation by the operating surgeon to clarify inconclusive MRI findings for preoperative planning
        ® Requests for CT, MRA, or CTA should be forwarded for Medical Director review

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is superior to PET/CT for the detection of pulmonary metastases, and is indicated in the initial workup of all suspected malignant bone tumors and should be completed prior to anesthesia exposure if possible

      Other staging imaging should be deferred until a histologic diagnosis is made, initially by biopsy, as definitive resection is performed after neoadjuvant chemotherapy

        ® Bone and bone marrow metastases can occur in ESFT, and cause a significant change in treatment approach. PET/CT can replace bone scan and bone marrow biopsy in ESFT members and is indicated in the initial staging of all ESFT members after histologic diagnosis is established
          ¡ Whole body PET/CT (CPT® 78816) is the preferred study for initial staging of ESFT
          ¡ Bone scan (See PEDONC-1.3: Modality General Considerations) may be substituted for PET imaging if PET not available
          ¡ If PET/CT is negative for bony metastases at initial diagnosis, bone scan (See PEDONC-1.3: Modality General Considerations) is preferred for asymptomatic surveillance at all-time points after completion of therapy
        ® CT Abdomen and Pelvis with contrast (CPT® 74177) is not routinely indicated in the initial metastatic staging of pediatric ESFT, but can be approved in the following situations:
          ¡ Evaluation of inconclusive PET findings
          ¡ Primary site involving the abdomen or pelvis
      ESFT Treatment Response:

      All ESFT members undergo restaging after ~12 weeks of neoadjuvant chemotherapy prior to local control surgery to confirm the absence of progressive disease prior to the extended break necessary for postoperative healing.

      Restaging at this time point should include:

        ® MRI without and with contrast of primary site
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250)
        ® Whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations)

      Following local control surgery, the following imaging guidelines should be used until the end of planned chemotherapy:
        ® MRI without and with contrast of primary site 3 months after surgical procedure and at the end of planned chemotherapy
        ® Plain x-rays of the primary site and chest immediately after local control then every 3 months
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250):
          ¡ Measurable pulmonary metastases: every 6 weeks and at the end of planned chemotherapy
          ¡ No measurable pulmonary metastases: every 3 months and at the end of planned chemotherapy
        ® Whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations) at the end of planned chemotherapy

      Members with metastatic disease do not routinely undergo local control surgery unless metastatic disease has resolved with chemotherapy.
        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be performed every 2 cycles during treatment and at the end of planned chemotherapy
        ® MRI without and with contrast of primary site can be performed every 2 cycles during treatment and at the end of planned chemotherapy
        ® If previously positive for bony metastases, whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations) every 2 cycles during treatment and at the end of planned chemotherapy
        ® Imaging may be indicated more frequently around the time of surgical resection of primary or metastatic lesions to assess for resectability

      PET is generally not indicated during active treatment for recurrent pediatric cancer. In rare circumstances, PET may be appropriate when conventional imaging is inconclusive and results are likely to result in a treatment change for the member, including a change from active treatment to surveillance. These requests will be forwarded for Medical Director review.

      ESFT Surveillance Imaging:
      Appendicular bone primary tumor site:

        ® Plain x-rays of the primary tumor site should be completed every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy
          ¡ MRI is not routinely indicated for surveillance imaging of these primary sites after completion of chemotherapy but should be approved for the following:
            § The member does not have an endoprosthesis that causes MRI or CT artifact
            § To clarify inconclusive findings on plain x-ray
            § To evaluate significant pain symptoms suggestive of primary site recurrence
      Axial bone or any soft tissue primary site:
        ® CT with contrast or MRI without and with contrast of the primary tumor site can be approved every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy

      Metastatic disease surveillance:
        ® Members with localized ESFT:
          ¡ CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 1 year then every 4 months for 1 year after completion of all therapy
            § Chest X-ray (CXR) should be used for pulmonary recurrence surveillance after 24 months, and CT Chest can be approved to clarify inconclusive CXR findings
        ® Members with metastatic or recurrent ESFT:
          ¡ CT Chest with (CPT® 71260) or without contrast (CPT® 71250) every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy
        ® Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) should be used for evaluation of distant bony metastases every 3 months for 1 year, then every 4 months for 1 year, then every 6 months for 1 year, then annually for 2 years after completion of all therapy
        ® PET/CT has no established role for asymptomatic surveillance of ESFT, but can be approved in the following circumstances:
          ¡ Conventional imaging reveals findings that are inconclusive or suspicious for recurrence and PET avidity will determine whether biopsy or continued observation is appropriate
          ¡ Rare circumstances where obvious clinical symptoms show strong evidence suggesting recurrence and PET would replace conventional imaging modalities
          ¡ Restaging after biopsy-confirmed recurrence
          ¡ These requests will be forwarded for Medical Director review.
      References

      1. Hawkins DS, Brennan BMD, Bölling T, et al. Ewing Sarcoma. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:855-876.
      2. Gorlick R, Janeway K, and Marina N. Osteosarcoma. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:877-898.
      3. DuBois SG, Grier HE, and Lessnick SL. Ewing Sarcoma. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:1983-2017.
      4. Janeway KA. Osteosarcoma. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:2018-2055.
      5. Azouz EM. Magnetic resonance imaging of benign bone lesions: cysts and tumors. Top Magn Reson Imaging. 2002 August;13(4):219-230.
      6. Wolf M. Knee pain in children, part III: stress injuries, benign bone tumors, growing pains. Pediatr Rev. 2016;37(3):114-119. doi:10.1542/pir.2015-0042.
      7. Uslu L, Doing J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for Evaluation of Pediatric Malignancies. J Nucl Med. 2015;56(2):274-286. doi:10.2967/jnumed.114.146290.
      8. Reed DR, Hayashi M, Wagner L, et al. Treatment pathway of bone sarcoma in children, adolescents, and young adults. Cancer. 2017 March;123(12):2206-2218.
      9. Hurley C, McCarville MB, Shulkin BL, et al. Comparison of 18F-FDG-PET-CT and Bone Scintigraphy for evaluation of osseous metastases in newly diagnosed and recurrent osteosarcoma. Pediatr Blood Cancer. 2016;63(8):1381-1386. Accessed January 4, 2018. doi:10.1002/pbc.26014.
      10. Byun BH, Kong CB, Lim I, et al. Comparison of (18)F-FDG PET/CT and (99m)TC-MDP bone scintigraphy for detection of bone metastasis in osteosarcoma. Skeletal Radiol. 2013;42(12):1673-1681.
      11. Quartuccio N, Treglia G, Saisano M, et al. The role of Fluorine-18-Fluorodeoxyglucose positron emission tomography in staging and restaging of patients with osteosarcoma. Radiol Oncol. 2013;47(2):97-102. doi:10.2478/raon-2013-0017.
      12. Isakoff MS, Bielack SS, Meltzer P, Gorlick R. Osteosarcoma: current treatment and a collaborative pathway to success. J Clin Oncol. 2015;33(27):3029-3035. doi:10.1200/JCO.2014.59.4895.
      13. Newman EN, Jones RL, Hawkins DS. An evaluation of [F-18]-Fluorodeoxy-D-Glucose positron emission tomography, bone scan, and bone marrow aspiration/biopsy as staging investigations in Ewing sarcoma. Pediatr Blood Cancer. 2013;60(7):1113-1117. doi:10.1002/pbc.24406.
      14. Roberts CC, Kransdorf MJ, Beaman FD, et al. Follow-up of malignant or aggressive musculoskeletal tumors. ACR Appropriateness Criteria® 2015:1-15. https://acsearch.acr.org/docs/69428/Narrative/.
      15. Meyer JS, Nadel HR, Marina N, et al. Imaging guidelines for children with Ewing sarcoma and osteosarcoma: a report from the Children’s Oncology Group Bone Tumor Committee. Pediatr Blood Cancer. 2008;51(2):163-170. doi:10.1002/pbc.21596.
      16. Gaspar N, Hawkins DS, Dirksen U, et al. Ewing sarcoma: current management and future approaches through collaboration. J Clin Oncol. 2015;33(27):3036-3046. doi:10.1200/JCO.2014.59.5256.
      17. Mascarenhas L, Felgenhauer JL, Bond MC, et al. Pilot study of adding vincristine, topotecan, and cyclophosphamide to interval-compressed chemotherapy in newly diagnosed patients with localized Ewing sarcoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2016;63(3):493-498. doi:10.1002/pbc.25837.
      18. Weiser DA, Kaste SC, Siegel MJ, et al. Imaging in childhood cancer: a society for pediatric radiology and children’s oncology group joint task force report. Pediatr Blood Cancer. 2013;60(8):1253-1260. doi:10.1002/pbc.24533.
      19. Dimitrakopoulou-Strauss A, Strauss LG, Heichel T, et al. The role of quantitative 18F-FDG PET studies for the differentiation of malignant and benign bone lesions. J Nucl Med. 2002;43(4):510-518.
      20. Morrison WB, Weissman BN, Kransdorf MJ, et al. Primary bone tumors. ACR Appropriateness Criteria®, 2013;1-13. Date of origin: 1995 Last review date: 2013 https://acsearch.acr.org/docs/69421/Narrative/.

      PEDONC-10: Pediatric Germ Cell Tumors

      For this condition imaging is medically necessary based on the following criteria:

      Malignant pediatric germ cell tumors commonly include one of four histologic subtypes (yolk sac tumor, choriocarcinoma, embryonal carcinoma, or mixed histology), but the overall treatment strategies are similar for all malignant germ cell tumors. Tumors can occur in testicular, ovarian or extragonadal primary locations.

      This section applies to primary germ cell tumors occurring outside the central nervous system in children who are ≤15 years old at the time of initial diagnosis.

        For members who are >15 years old at diagnosis, the overall prognosis is inferior and these members should be imaged according to adult guidelines in: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-20: Testicular, Ovarian and Extragonadal Germ Cell Tumors in the Oncolocy Imaging Guidelines.

      Sex cord stromal tumors (granulosa cell, theca, sertoli, and leydig tumors) are rare in pediatrics and should be imaged according to adult guidelines in: Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-20: Testicular, Ovarian and Extragonadal Germ Cell Tumors in the Oncolocy Imaging Guidelines.

      For CNS germ cell tumors, use the imaging guidelines in: PEDONC-4.7: CNS Germinomas and Non-Germinomatous Germ Cell Tumors (NGGCT).

      Pediatric GCT Initial Staging:

      Ovarian, testicular, and abdominal extragonadal GCT should have ultrasound and tumor markers (AFP, -hCG) as initial evaluation

        ® Mediastinal primary tumors should be evaluated by CT Chest with contrast
        ® Ovarian masses that are <10 cm in size, have minimal or no visible solid component on ultrasound, and have normal tumor markers are almost universally benign teratomas or functional cysts and advanced imaging is not necessary unless ultrasound is insufficient for immediate preoperative planning.

      Once a primary mass suspected to be GCT is discovered, initial staging with CT Abdomen/Pelvis with contrast (CPT® 74177) is indicated prior to histologic confirmation
        ® The degree of abdominal exploration and node sampling necessary for adequate staging is determined in part by imaging findings and is required for preoperative planning
        ® Testicular primary tumors can defer abdominal imaging until after histologic confirmation at the discretion of the operating surgeon
        ® MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be approved to clarify inconclusive CT findings or for members with a known contraindication to CT contrast

      CT Chest with contrast (CPT® 71260) is indicated in the initial workup of all pediatric GCT and should be completed prior to anesthesia exposure if possible

      MRI Brain without and with contrast (CPT® 70553) can be approved for members with symptoms suggesting CNS metastases

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) should be used for initial evaluation of bony metastases in members with systemic symptoms or bone pain

      There has been no published evidence to date supporting the routine use of PET/CT in the evaluation of pediatric GCT

        ® Additionally, PET has been found to have similar efficacy to CT imaging in initial staging of adults with non-seminomatous GCT (the majority of pediatric GCT are non-seminomatous)

      Pediatric GCT Treatment Response:

      Members with localized GCT are often cured with surgery alone and do not receive adjuvant therapy. These members should be imaged using surveillance guidelines after surgery is completed.

      Members receiving adjuvant chemotherapy are usually treated with 4 to 6 cycles of combination chemotherapy.

      The primary method of response assessment is by tumor marker decrease

        ® For members with disease not completely resected at initial diagnosis, repeat imaging with CT Chest/Abdomen/Pelvis (CPT® 71260 and CPT® 74177) with contrast can be approved every 2 cycles (~every 6 weeks)
        ® CT imaging may be indicated more frequently to assess for surgical resectability in members who have received more than 4 cycles of chemotherapy

      CT Chest/Abdomen/Pelvis with contrast (CPT® 71260 and CPT® 74177) is indicated at the end of planned chemotherapy or following neoadjuvant chemotherapy for initially unresectable tumors

      Imaging of any metastatic sites should be approved every 2 cycles and at the end of planned therapy with the same modality used during initial staging

      PET as a marker of treatment response has been shown not to be predictive of member outcomes in GCT and should not be approved

        ® Suspicious lesions seen on conventional imaging should be biopsied to confirm active disease
        ® Alternatively, a short-interval CT study can be approved if the relapse risk is determined to be low by the treating physician and biopsy would cause unnecessary morbidity for the member

      Pediatric GCT Surveillance Imaging:

      The primary method of surveillance in pediatric GCT is frequent assessment of serum tumor markers, unless tumor markers were not elevated at diagnosis.

      CT Chest/Abdomen/Pelvis with contrast (CPT® 71260 and CPT® 74177) should be approved for any clinically significant rise in tumor markers or symptoms suggesting recurrent disease

      CT Abdomen/Pelvis with contrast (CPT® 74177) can be approved every 6 months for 2 years then every 12 months for 3 years after completion of all therapy for members with normal tumor markers at the time of diagnosis

      For stage I members age 0-10 years treated with surgery only:

        ® Chest X-ray (CXR) should be completed every 3 months for 1 year after completion of all therapy
          ¡ CT Chest is indicated to evaluate abnormal CXR findings or if the primary tumor site was in the thoracic cavity
        ® CT Abdomen/Pelvis with contrast (CPT® 74177) can be approved every 3 months for 1 year after completion of all therapy

      For stage I members ages 11+ years treated with surgery only:
        ® Chest X-Ray (CXR) should be completed every 4 months for 2 years, then every 6 months for 1 year, then every 12 months for 1 year after completion of all therapy
          ¡ CT Chest is indicated to evaluate abnormal CXR findings or in lieu of CXR if the primary tumor site was in the thoracic cavity
        ® CT Abdomen/Pelvis with contrast (CPT® 74177) can be approved every 4 months for 2 years, then every 6 months for 1 year, then every 12 months for 1 year after completion of all therapy

      For stage II-IV members:
        ® Chest X-ray (CXR) should be completed every 6 months for 2 years then every 12 months for 1 year after completion of all therapy
          ¡ CT Chest is indicated to evaluate abnormal CXR findings or in lieu of CXR if a primary or metastatic tumor site was in the thoracic cavity
        ® CT Abdomen/Pelvis with contrast (CPT® 74177) can be approved every 6 months for 2 years, then every 12 months for 1 year completion of all therapy

      Members with brain or bone metastases should have surveillance imaging on the same schedule as the primary site imaging with the same modality used during initial staging.

      References

      1. Frazier AL, Olson TA, Schneider DT, et al. Germ cell tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:899-918.
      2. Frazier AL, Billmire D, Amatruda J. Pediatric germ cell tumors. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:2056-2099.
      3. Gilligan T, Beard C, Carneiro B, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 2.2017—December 8, 2016, Testicular Cancer, available at: https://www.nccn.org/professionals/physician_gls/pdf/testicular.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Testicular Cancer V2.2017 12/8/16. ©2016 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org
      4. Armstrong DK, Plaxe SC, Alvarez RD, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 4.2017—November 9, 2017 Ovarian Cancer, available at: https://www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Ovarian Cancer V4.2017 11/9/17. ©2017 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org
      5. de Wit M, Brenner W, Hartmann M, et al. [18F]-FDG-PET in clinical stage I/II non-seminomatous germ cell tumors: results of the German multicentre trial. Ann Oncol. 2008;19(9):1619-1623. doi:10.1093/annonc/mdn170.
      6. Huddart RA, O’Doherty MJ, Padhani A, et al. 18Flourodeoxyglucose positron emission tomography in the prediction of relapse in patients with high-risk, clinical stage I nonseminomatous germ cell tumors: preliminary report of MRC trial TE22--the NCRI testis tumor clinical study group. J Clin Oncol. 2007;25(21):3090-3095. doi:10.1200/JCO.2006.09.3831.
      7. Papic JC, Finnell MA, Slaven JE, et al. Predictors of ovarian malignancy in children: overcoming clinical barriers of ovarian preservation. J Pediatr Surg. 2014;49(1):144-148. doi:10.1016/j.jpedsurg.2013.09.068.
      8. Olson TA, Murray MJ, Rodriguez-Galindo C, et al. Pediatric and adolescent extracranial germ cell tumors: the road to collaboration. J Clin Oncol. 2015;33(27):3018-3028. doi:10.1200/JCO.2014.60.5337.
      9. Fresneau B, Orbach D, Faure-Conter C, et al. Sex-cord stromal tumors in children and teenagers: results of the TGM-95 study. Pediatr Blood Cancer. 2015;62(12):2114-2119. doi:10.1002/pbc.25614.
      10. Fonseca A, Xia C, Lorenzo AJ et al, Detection of Relapse by Tumor Markers Versus Imaging in Children and Adolescents With Nongerminomatous Malignant Germ Cell Tumors: A Report From the Children’s Oncology Group, J Clin Oncol 2019;37:396-402. doi:10.1200/JCO.18.00790.

      PEDONC-11: Pediatric Liver Tumors
      PEDONC-11.1: Pediatric Liver Tumors – General Considerations
      PEDONC-11.2: Hepatoblastoma
      PEDONC-11.3: Pediatric Hepatocellular Carcinoma (HCC)
      PEDONC-11.1: Pediatric Liver Tumors – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Note: Some payors consider PET imaging to be investigational for the treatment of hepatobiliary tumors, and those coverage policies may supersede the recommendations for PET imaging in this section.

      Pediatric liver tumors primarily include hepatoblastoma and hepatocellular carcinoma, but hepatic germ cell tumors and primary hepatic sarcomas occur with some frequency. Tumor markers are useful for initial evaluation as well as treatment response, particularly in hepatoblastoma. Early consideration of liver transplant may be undertaken in children and adolescents with unrespectable localized disease, provided that the disease remains confined to the liver.

      Primary hepatic germ cell tumors should follow imaging guidelines in: PEDONC-10: Pediatric Germ Cell Tumors.

      Primary hepatic sarcomas should follow imaging guidelines in: PEDONC-8.3: Non-Rhabdomyosarcoma Soft Tissue Sarcomas (NRSTS).

      Imaging requests relating to liver transplant surgery and surveillance should follow guidelines in section Adult Abdomen Imaging Policy (Policy #148 in the Radiology Section); AB-42: Transplant in the abdomen imaging guidelines.



      PEDONC-11.2: Hepatoblastoma

      For this condition imaging is medically necessary based on the following criteria:

      Hepatoblastoma Initial Staging:

      Hepatoblastoma occurs most commonly in very young children (median diagnosis age of 19 months). Most cases of hepatoblastoma are sporadic, but some are associated with genetic abnormalities, including Beckwith-Wiedemann syndrome, familial adenomatous polyposis, and trisomy 18. Most suspected liver tumors will have ultrasound and tumor markers (AFP, -HCG, CEA) as part of the initial evaluation.

      Ultrasound may be approved even after MRI or CT imaging in order to allow evaluation for tumor thrombus

      Once a primary liver mass is discovered, definitive imaging is indicated prior to histologic diagnosis, and may involve any of the following:

        ® MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) is preferred for evaluating tumor margins and vascular anatomy
        ® CT Abdomen/Pelvis with contrast (CPT® 74177)
          ¡ Noncontrast imaging is not indicated due to the increased radiation exposure and limited additive benefit
        ® Some tumors may require both MRI and CT during initial evaluation
        ® MRA (CPT® 74185) or CTA (CPT® 74175) Abdomen are often indicated to evaluate vascular invasion

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is indicated in the initial work-up of all pediatric liver tumors and should be completed prior to anesthesia exposure if possible

      MRI Brain without and with contrast (CPT® 70553) can be approved only for members with symptoms suggesting CNS metastases

      Bone scan (See PEDONC-1.3: Modality General Considerations) should be used for initial evaluation of bony metastases only in members with systemic symptoms or bone pain

      There has been no published evidence to date supporting the routine use of PET/CT imaging in the evaluation of pediatric hepatoblastoma

        ® PET/CT should only be considered in very rare circumstances for preoperative planning when MRI and CT scans are insufficient for surgical decision making.
        ® PET/CT should not be approved in lieu of biopsy of suspicious lesions
        ® These requests will be forwarded for Medical Director review.

      Hepatoblastoma Treatment Response:

      Members with localized hepatoblastoma of pure fetal histology are often cured with surgery alone and do not receive adjuvant therapy. These members should be imaged using surveillance guidelines after surgery is completed.

      Members receiving adjuvant chemotherapy are usually treated with 2 to 8 cycles of combination chemotherapy. Tumor marker decrease is important in response assessment but does not eliminate the need for advanced imaging in members with unresected hepatoblastoma.

      For members with disease not completely resected at initial diagnosis, the following can be approved every 2 cycles (~6 weeks) and at the end of planned therapy for all members:

        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250)
        ® CT Abdomen/Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197)
          ¡ While the majority of members will require abdomen and pelvis imaging at all time points, the pelvis imaging may be omitted at the discretion of the ordering physician based on the member’s specific clinical situation
          ¡ MRA (CPT® 74185) or CTA (CPT® 74175) Abdomen are often indicated to evaluate vascular invasion
        ® Imaging of any metastatic sites with the same modality used during initial staging

      Imaging may be indicated more frequently to assess for surgical resectability in members who have received more than 4 cycles of chemotherapy.

      Abdominal ultrasound is indicated if tumor thrombus was detected at initial diagnosis

        ® If no tumor thrombus was present, continued ultrasound evaluations are not indicated without a specific reason documented in the clinical records

      PET/CT should only be considered in very rare circumstances for preoperative planning when MRI and CT scans are insufficient for surgical decision making.
        ® PET/CT should not be approved in lieu of biopsy of suspicious lesions.
        ® These requests will be forwarded for Medical Director review.

      Hepatoblastoma Surveillance Imaging:

      The primary method of surveillance in hepatoblastoma is frequent assessment of serum tumor markers (primarily AFP).

      No specific imaging is indicated for surveillance in members with an AFP of >100 ng/ml at diagnosis or recurrence.

        ® CT Chest and Abdomen with contrast (CPT® 71260 and CPT® 74160) can be approved for any clinically significant rise in tumor markers or symptoms suggesting recurrent disease

      For members with AFP ≤100 ng/ml at diagnosis or recurrence, the following imaging is appropriate:
        ® CT Abdomen with contrast (CPT® 74160) should be completed every 3 months for 2 years then every 4 months for 2 years after completion of all therapy
        ® Chest X-ray or CT Chest with contrast (CPT® 71260) should be completed every 3 months for 2 years then every 4 months for 2 years after completion of all therapy
        ® Members with brain or bone metastases should have surveillance imaging on the same schedule as the primary site imaging with the same modality used during initial staging.

      PET/CT has no documented role in the surveillance evaluation of pediatric hepatoblastoma


      PEDONC-11.3: Pediatric Hepatocellular Carcinoma (HCC)

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric HCC Initial Staging:

      HCC, including its rare histologically distinct variant fibrolamellar hepatocellular carcinoma (FL-HCC), occurs mostly in older children and adolescents. Despite recent advances in treatment, overall survival of pediatric HCC diagnosed in advanced stages remains exceedingly poor, with five-year survival of only 17% to 22% for all stages of pediatric HCC (and FL-HCC). Most suspected liver tumors will have ultrasound and tumor markers (AFP, β-HCG, CEA) as initial evaluation.

      Ultrasound may be approved even after MRI or CT imaging in order to allow evaluation for tumor thrombus.

      Once a primary liver mass is discovered, definitive imaging is indicated prior to histologic diagnosis, and may involve any of the following:

        ® CT Abdomen/Pelvis with contrast (CPT® 74177)
        ® MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197)
        ® Some tumors may require both MRI and CT during initial evaluation
        ® MRA (CPT® 74185) or CTA (CPT® 74175) Abdomen are often indicated to evaluate vascular invasion

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) is indicated in the initial work-up of all pediatric liver tumors and should be completed prior to anesthesia exposure if possible

      MRI Brain without and with contrast (CPT® 70553) can be approved only for members with symptoms suggesting CNS metastases

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) should be used for initial evaluation of bony metastases only in members with systemic symptoms or bone pain

      PET/CT should only be considered in very rare circumstances for preoperative planning when MRI and CT are insufficient for surgical decision making.

        ® PET/CT should not be approved in lieu of biopsy of suspicious lesions
        ® These requests require Medical Director review.

      Pediatric HCC Treatment Response:

      The majority of hepatocellular carcinoma members are treated with surgery alone and do not receive adjuvant therapy. Members with successful upfront gross total resection should be imaged using surveillance guidelines after surgery is completed.

      For members with disease not completely resected at initial diagnosis, the following can be approved every 2 cycles (~6 weeks) and at the end of planned therapy for all members:

        ® CT Chest with (CPT® 71260) or without contrast (CPT® 71250)
        ® CT Abdomen/Pelvis with contrast (CPT® 74177) or MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197)
          ¡ While the majority of members will require abdomen and pelvis imaging at all time points, the pelvis imaging may be omitted at the discretion of the ordering physician based on the member’s specific clinical situation
          ¡ MRA (CPT® 74185) or CTA (CPT® 74175) Abdomen are often indicated to evaluate vascular invasion
        ® Imaging of any metastatic sites with the same modality used during initial staging

      Abdominal ultrasound is indicated if tumor thrombus was detected at initial diagnosis
        ® If no tumor thrombus was present, continued ultrasound evaluations are not indicated without a specific reason documented in the clinical records

      PET/CT should only be considered in very rare circumstances for preoperative planning when MRI and CT scans are insufficient for surgical decision making.
        ® PET/CT should not be approved in lieu of biopsy of suspicious lesions
        ® These requests will be forwarded for Medical Director review.

      Pediatric HCC Surveillance Imaging:

      CT Abdomen/Pelvis with contrast (CPT® 74177) can be completed every 3 months for 1 year then every 6 months for 1 year, then annually for 3 years after completion of all therapy

      Chest X-ray or CT Chest with contrast (CPT® 71260) should be every 3 months for 1 year then every 6 months for 1 year, then annually for 3 years after completion of all therapy

      Members with brain or bone metastases should have surveillance imaging on the same schedule as the primary site imaging with the same modality used during initial staging.

      PET/CT has no documented role in the surveillance evaluation of pediatric hepatocellular carcinoma

      References

      1. Meyers RL, Trobaugh-Lotario AD, Malogolowkin MH, et al. Pediatric liver tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:726-752.
      2. Tomlinson GE. Hepatoblastoma and other liver tumors in children. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:1886-1905.
      3. Pugmire BS, Towbin AJ. Magnetic resonance imaging of primary pediatric liver tumors. Pediatr Radiol. 2016;46:764-777. doi:10.1007/s00247-016-3612-0.
      4. Chavhan GB, Shelmerdine S, Jhaveri K, Babyn PS. Liver MR Imaging in children: current concepts and technique. RadioGraphics. 2016;36(5):1517-1532. doi:10.1148/rg.2016160017.
      5. Rai P, Feusner H. Cerebral metastasis of hepatoblastoma: a review. J Pediatr Hematol Oncol. 2016;38(4):279-282. doi:10.1097/MPH.0000000000000554.
      6. Allen-Rhoades W, Whittle SB, and Rainusso N, Pediatric Solid Tumors of Infancy: An Overview, Pediatr Rev 2018;39:57-67. doi:10.1542/pir.2017-0057.
      7. Allan BJ, Wang B, Davis JS, et al. A review of 218 pediatric cases of hepatocellular carcinoma. J Pediatr Surg. 2014;49(1):166-171. doi:10.1016/j.jpedsurg.2013.09.050.
      8. Pappo AS, Furman WL, Schulz KA, Ferrari A, Helman L, Krailo MD. Rare tumors in children: progress through collaboration. J Clin Oncol. 2015;33(27):3047-3054. doi:10.1200/JCO.2014.59.3632.
      9. Benson AB, D’Angelica MI, Abbott DE, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 3.2019—August 1, 2019, Hepatobiliary Cancers, available at: https://www.nccn.org/professionals/physician_gls/pdf/hepatobiliary.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Hepatobiliary Cancers V3.2019 8/1/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org.
      10. D’Souza AM, Shah R, Gupta A, et al, Surgical management of children and adolescents with upfront completely resected hepatocellular carcinoma, Pediatr Blood Cancer 2019;65:e27293. doi:10.1002/pbc.27293.
      11. Weeda VB, Aronson DC, Verheij J, and Lamers WH, Is hepatocellular carcinoma the same disease in children and adults? Comparison of histology, molecular backgroung, and treatment in pediatric and adult patients, Pediatr Blood Cancer 2019;66:e27475. doi:10.1002/pbc.27475.

      PEDONC-12: Retinoblastoma

      PEDONC-12.1: Retinoblastoma – General Considerations
      PEDONC-12.2: Retinoblastoma Imaging

      PEDONC-12.1: Retinoblastoma – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Retinoblastoma (RB) is primarily a disease of the infant and young child, and presents with leukocoria (loss of red reflex). About 75% of members are diagnosed before the age of two years (bilateral RB presents at 12 months of age). Retinoblastoma can occur as heritable (25% of cases) or nonheritable (75%) disease. Heritable RB is associated with a germline mutation in the RB1 gene often resulting typically in bilateral disease. Individuals who carry the RB1 mutation also have increased risk of developing other cancers, such as osteosarcoma, soft tissue sarcomas, or melanoma. For more information on heritable retinoblastoma, see PEDONC-2.12: Familial Retinoblastoma Syndrome.

      Detailed evaluation by a physician with significant training and/or experience in retinoblastoma (most commonly a pediatric ophthalmologist or pediatric oncologist) is indicated prior to considering advanced imaging.

      Retinoblastoma can be unilateral, bilateral, or trilateral (involving the pineal gland). Extraocular spread of retinoblastoma is rare and generally confined to the brain.


      PEDONC-12.2: Retinoblastoma Imaging

      For this condition imaging is medically necessary based on the following criteria:

      Retinoblastoma Initial Staging

      Tumor biopsy is NOT required prior to imaging

      MRI Orbits (CPT® 70543) and Brain (CPT® 70553) without and with contrast can be approved in the initial work-up of all members with retinoblastoma

        ® Brain imaging may be omitted or deferred at the discretion of the treating ophthalmologist or oncologist

      MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) may be approved if there is evidence of CNS metastasis on:
        ® Ophthalmologic exam
        ® MRI Brain
        ® Lumbar CSF cytology

      CT should generally be avoided in retinoblastoma members under one year of age or with family history of retinoblastoma (heritable) due to substantially increased risks for secondary malignancy
        ® CT Chest (CPT® 71260) and MRI Abdomen and Pelvis without and with contrast (CPT® 74183 and CPT® 72197) can be approved for members with clinical symptoms to suggest metastatic disease

      CT Orbital (contrast as requested) and orbital ultrasound can be approved if ordered by the treating ophthalmologist for a specified indication

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) is the preferred imaging modality for members with systemic bone pain suggestive of bony metastases

      PET has no documented role in the evaluation of retinoblastoma

      Retinoblastoma Treatment Response:

      MRI Orbits (CPT® 70543) and/or Brain (CPT® 70553) can be approved every 2 cycles (~ every 6 weeks) and at the end of planned therapy

      For members with metastatic disease, imaging of known positive areas using the same modality at initial staging can be approved every 2 cycles (~6 to 8 weeks) and at the end of planned therapy

      Retinoblastoma Surveillance:

      The primary method of surveillance in retinoblastoma is examination under anesthesia (EUA), although some older children can be sufficiently evaluated by exam without anesthesia (EWA).

        ® Surveillance using advanced imaging is generally not indicated for unilateral retinoblastoma after enucleation or exenteration, but can be approved for evaluation of specific clinical concerns.
        ® Members undergoing ocular salvage treatment approaches can have MRI Orbits (CPT® 70543) and Brain (CPT® 70553) approved every 6 months for 2 years following completion of therapy.

      Members with bilateral retinoblastoma or germline mutation in RB1 are at increased risk for subsequent pineoblastoma, so MRI Brain without and with contrast (CPT® 70553) can be approved every 6 months for 5 years for the time of diagnosis with retinoblastoma
        ® Routine MRI follow up for pineal disease is not currently supported by evidence in unilateral retinoblastoma members without germline RB1 mutations

      References

      1. Hurwitz RL, Shields CL, Shields JA, et al. Retinoblastoma. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:700-725.
      2. Rodriguez-Galindo C, Wilson MW, and Dyer M. Retinoblastoma. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:1747-1778.
      3. de Jong MC, Kors WA, de Graaf P, Castelijns JA, Kivelä T, Moll AC. Trilateral retinoblastoma: a systematic review and meta-analysis. Lancet Oncol. 2014;15(10):1157-1167. doi:10.1016/S1470-2045(14)70336-5.
      4. De Graaf P, Göricke S, Rodjan F, et al. Guidelines for imaging retinoblastoma: imaging principles and MRI standardization, Pediatr Radiol. 2012;42(1):2-14. doi:10.1007/s00247-011-2201-5.
      5. Allen-Rhoades W, Whittle SB, and Rainusso N, Pediatric Solid Tumors of Infancy: An Overview, Pediatr Rev 2018;39:57-67. doi:10.1542/pir.2017-0057.
      6. Lohmann DR, Gallie BL. Retinoblastoma. In: Pagon RA, Adam MP, Ardinger HH et al, eds. GeneReviews™ [Internet]. Seattle, WA: University of Washington, Seattle; 1993-2019. version November 21, 2018. https://www.ncbi.nlm.nih.gov/books/NBK1452/.

      PEDONC-13: Pediatric Nasopharyngeal Carcinoma

      PEDONC-13.1: Pediatric Nasopharyngeal Carcinoma –General Considerations
      PEDONC-13.2: Pediatric – NPC Imaging

      PEDONC-13.1: Pediatric Nasopharyngeal Carcinoma – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric nasopharyngeal carcinoma (NPC) is rare in comparison to adult NPC but is responsible for up to 50% of nasopharyngeal cancers in children and has higher rates of aggressive type III EBV-associated histology than adult NPC.

      Metastasis frequently occurs in cervical lymph nodes and retropharyngeal space. Distal metastasis usually appears in bones, lungs, mediastinum, and rarely, in the liver. In many members, the initial presentation is a cervical adenopathy, and diagnosis is made with a lymph node biopsy.

      Standard upfront treatment in pediatric NPC consists of 3 to 4 cycles of neoadjuvant chemotherapy followed by definitive chemoradiotherapy. Rare members with lower stage disease may be treated with radiotherapy alone.

      PEDONC-13.2: Pediatric NPC – Imaging

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric NPC Initial Staging:

      Quantitative EBV DNA PCR should be measured at initial diagnosis, as it can serve as an effective tumor marker if elevated at initial diagnosis.

      MRI Brain without and with contrast (CPT® 70553) and MRI Neck without and with contrast (CPT® 70543) is indicated in the initial staging of all pediatric NPC members

        ® CT Head without and with contrast (CPT® 70470), CT Maxillofacial without and with contrast (CPT® 70488) and/or CT Neck with contrast (CPT® 70491) can be approved for members with documented contraindication to MRI imaging (avoidance of sedation should not be the sole reason)
        ® Skull base invasion is common in pediatric NPC and has a dramatic impact on prognosis, and is more easily recognized on MRI imaging

      CT Chest with contrast (CPT® 71260) is indicated in initial staging of all members

      Whole body PET/CT (CPT® 78816) is approvable after histologic confirmation of NPC to evaluate for distant bony metastases

        ® Bone scan (See PEDONC-1.3: Modality General Considerations) can be used for members when PET/CT is unavailable

      Pediatric NPC Treatment Response:

      MRI Brain without and with contrast (CPT® 70553) and MRI Neck without and with contrast (CPT® 70543) are indicated for response assessment at the following time points:

        ® Following completion of neoadjuvant chemotherapy
        ® Following completion of chemoradiotherapy

      CT Chest with contrast (CPT® 71260) and whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations) are indicated at the following time points:
        ® Following completion of neoadjuvant chemotherapy only if positive at initial diagnosis
        ® Following completion of chemoradiotherapy

      PET is generally not indicated during active treatment for recurrent pediatric cancer. In rare circumstances, PET may be appropriate when results are likely to result in a treatment change for the member, including a change from active treatment to surveillance. These requests will be forwarded for Medical Director review.

      Pediatric NPC Surveillance:

      MRI Brain without and with contrast (CPT® 70553) and MRI Neck without and with contrast (CPT® 70543) are indicated every 3 months for 1 year, then every 6 months for 2 years after completion of all planned therapy

      CT Chest with contrast (CPT® 71260) is indicated every 3 months for 1 year, then every 6 months for 2 years after completion of all planned therapy

      Whole body PET/CT (CPT® 78816) or bone scan (See PEDONC-1.3: Modality General Considerations) are not indicated for routine surveillance in asymptomatic members but can be approved in the following situations:

        ® Clarification of specified inconclusive findings seen on conventional imaging (should not replace biopsy)
        ® Restaging to identify sites of disease when EBV PCR levels are abnormally high and conventional imaging is negative
        ® Restaging after histologically confirmed recurrence of NPC
        ® These requests will be forwarded for Medical Director review.

      References

      1. Pappo AS, Rodriguez-Galindo C, and Furman WL. Management of infrequent cancers of childhood. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:1098-1123.
      2. Goldberg JM, Pappo AS, and Bishop M. Rare tumors of childhood. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:2123-2145.
      3. Cheuk DKL, Sabin ND, Hossain M, et al. Positron emission tomography-computed tomography for staging and follow-up of pediatric nasopharyngeal carcinoma. Eur J Nucl Med Mol Imaging. 2012;39(7):1097-1106. doi:10.1007/s00259-012-2091-2.
      4. Stambuk HE, Patel SG, Mosier KM, Wolder SL, Holodny AI. Nasopharyngeal carcinoma: recognizing the radiographic features in children. Am J Neuroradiol. 2005;26(6):1575-1579.
      5. Sahai P, Mohanti BK, Sharma A, et al. Clinical outcome and morbidity in pediatric patients with nasopharyngeal cancer treated with chemotherapy. Pediatr Blood Cancer. 2017;64(2):259-266. doi:10.1002/pbc.26240.

      PEDONC-14: Pediatric Adrenocortical Carcinoma

      PEDONC-14.1: Pediatric Adrenocortical Carcinoma – General Considerations
      PEDONC-14.2: Pediatric ACC – Imaging

      PEDONC-14.1: Pediatric Adrenocortical Carcinoma – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric Adrenocortical Carcinoma (ACC) is a rare but aggressive tumor, with fewer than 25 cases diagnosed each year. Most members are diagnosed because of virilizing symptoms, Cushing syndrome, and rarely with feminization and hyperaldosteronism or detection on screening imaging recommended for specified cancer predisposition syndromes. The mainstay of treatment is surgery. Chemotherapy, adrenal suppression, and radiotherapy typically follow resection. See: PEDONC-2: Cancer Predisposition Syndromes & Screening Strategies

      PEDONC-14.2: Pediatric ACC – Imaging

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric ACC Initial Staging:

      CT Abdomen without and with contrast (CPT® 74170) or MRI Abdomen without and with contrast (CPT® 74183) is indicated in the initial staging of all pediatric ACC members

      CT Chest with contrast (CPT® 71260) is indicated in initial staging of all members

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) is indicated to evaluate for bony metastases in all members at initial diagnosis

      PET has no documented role in the evaluation and treatment of pediatric ACC.

      Pediatric ACC Treatment Response:

      Many ACC members are treated with surgery alone and do not receive adjuvant therapy. These members should be imaged using surveillance guidelines after surgery is completed.

      For members treated with chemotherapy, CT Abdomen without and with contrast (CPT® 74170) or MRI Abdomen without and with contrast (CPT®74183) is indicated for response assessment every 2 cycles (~6 weeks) during chemotherapy and following completion of all planned chemotherapy

      CT Chest with contrast (CPT® 71260) is indicated every 2 cycles (~6 weeks) during chemotherapy and following completion of all planned chemotherapy

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) is indicated every 2 cycles (~6 weeks) during chemotherapy only if positive for distant metastases at initial diagnosis, and following completion of chemotherapy

      For members treated with radiotherapy, CT Abdomen without and with contrast (CPT® 74170) or MRI Abdomen without and with contrast (CPT®74183) is indicated for response assessment at the completion of radiotherapy

      Pediatric ACC Surveillance:

      CT Abdomen without and with contrast (CPT® 74170) or MRI Abdomen without and with contrast (CPT® 74183) is indicated every 3 months for 2 years, then every 6 months for 3 years after completion of all planned therapy

      Surveillance CT Chest is not indicated for members with localized disease at diagnosis

      For members with metastatic ACC, CT Chest with contrast (CPT® 71260) is indicated every 3 months for 2 years, then every 6 months for 3 years after completion of all planned therapy

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations) is indicated in all members with suspected bone recurrence

      References

      1. Waguespack SG, Huh WW, and Bauer AJ. Endocrine tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:919-945.
      2. Michalkiewicz E, Sandrini R, Figueiredo B, et al. Clinical and outcome characteristics of children with adrenocortical tumors: a report from The International Pediatric Adrenocortical Tumor Registry. J Clin Oncol. 2004:22(5):838-845. doi:10.1200/JCO.2004.08.085.
      3. Flynt KA, Dillman JR, Davenport MS, et al. Pediatric adrenocortical neoplasms: can imaging reliably discriminate adenomas from carcinomas? Pediatr Radiol. 2015;45(8):1160-1168. doi:10.1007/s00247-015-3308-x.
      4. Gupta N, Rivera M, Novotny P, et al, Adrenocortical Carcinoma in Children: A Clinicopathological Analysis of 41 Patients at the Mayo Clinical from 1950 to 2017, Horm Res Paediatr 2018;90:8-18. doi: 10.1159/000488855.

      PEDONC-15: Pediatric Melanoma and Other Skin Cancers

      For this condition imaging is medically necessary based on the following criteria:

      Pediatric melanoma is historically rare, but has a steadily rising incidence, especially in adolescents and young adults (AYAs). Staging is assigned using the American Joint Committee on Cancer (AJCC) staging for adult melanoma. Most cases of melanoma arising in children and AYAs (~75%) are localized at diagnosis, and approximately 90% of members with pediatric melanoma are amenable to radical excision. The clinical management of adolescents and young adults with melanoma is still challenging and evolving because it is difficult to diagnose, and there is no standard treatment.

      Non-melanoma skin cancers (mostly basal cell carcinoma and squamous cell carcinoma) are extremely rare in pediatric members. In many cases, predisposing factors such as prolonged immunosuppression, radiation therapy, chemotherapy, voriconazole use, or a combination of the factors are present, and established age-specific guidelines for management of these skin tumors do not exist.

      Imaging guidelines and treatment approaches are consistent with those used for adults with melanoma and other skin cancers, and these members should follow the imaging guidelines in section Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-5: Melanomas and Other Skin Cancers.

      References

      1. Rodriguez-Galindo C, Furman WL, Pappo AS. Rare pediatric tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:946-966.
      2. Goldberg JM, Pappo AS, Bishop M. Rare tumors of childhood. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:2123-2145.
      3. Coit DG, Thompson JA, Albertini MR, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 1.2018—October 11, 2017, Melanoma, available at: https://www.nccn.org/professionals/physician_gls/pdf/melanoma.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Melanoma V1.2018 10/11/17. ©2017 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org
      4. Senerchia AA, Ribiero KB, Rodriguez-Galindo C. Trends in incidence of primary cutaneous malignancies in children, adolescents, and young adults: a population-based study. Pediatr Blood Cancer. 2014;61(2):211-216. doi:10.1002/pbc.24639.
      5. Kolandijan NA, Wei C, Burke A, Bedikian AY. Malignant melanoma in teenagers and young adults. J Pediatr Hematol Oncol. 2014;36(7):552-558. doi:10.1097/MPH.0000000000000231.
      6. Brecht IB, De Paoli A, Bisogno G, et al, Pediatric patients with cutaneous melanoma: A European study, Pediatr Blood Cancer 2018;65:e26974. doi:10.1002/pbc.26974.
      7. Indini A, Brecht I, Del Vecchio M, et al, Cutaneous melanoma in adolescents and young adults, Pediatr Blood Cancer 2018;65:e27292. doi:10.1002/pbc.27292.
      8. Kohsravi H, Schmidt B, and Huang JT, Characteristics and outcomes of nonmelanoma skin cancer (NMSC) in children and young adults, J Am Acad Dermatol 2015;73:785-790. doi:10.1016/j.aad.2015.08.007.

      PEDONC-16: Pediatric Salivary Gland Tumors

      For this condition imaging is medically necessary based on the following criteria:

      The majority of pediatric salivary gland tumors arise in the parotid gland. Approximately 10 to 15% of tumors arise in the submandibular, sublingual, or minor salivary glands.

      Roughly 75% of pediatric salivary gland tumors are benign, most commonly pleomorphic adenoma.

      The most common malignant tumors occurring in the salivary glands are mucoepidermoid carcinoma, adenoid cystic carcinoma, acinic cell carcinoma, undifferentiated carcinoma, and rarely adenocarcinoma.

      American Joint Committee on Cancer (AJCC) staging is used for pediatric as well as adult salivary gland tumors.

      Imaging guidelines for malignant pediatric salivary gland tumors are consistent with those used for adults with salivary gland tumors, and these members should follow the imaging guidelines in section Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-4: Salivary Gland Cancers.


      References

      1. Rodriguez-Galindo C, Furman WL, and Pappo AS. Rare pediatric tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:946-966.
      2. Goldberg JM, Pappo AS, and Bishop M. Rare tumors of childhood. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:2123-2145.
      3. Pfister DG, Spencer S, Adelstein D, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 2.2017—May 8, 2017, Head and Neck Cancers, available at: https://www.nccn.org/professionals/physician_gls/pdf/head-and-neck.pdf, referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Head and Neck Cancers V2.2017 5/8/17. ©2017 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org
      4. Chiaravalli S, Guzzo M, Bisogno G, et al. Salivary gland carcinomas in children and adolescents: The Italian TREP project experience. Pediatr Blood Cancer. 2014;61(11):1961-1968.
      5. Rebours C, Couloigner V, Galmiche L, et al. Pediatric salivary gland carcinomas: diagnostic and therapeutic management. Laryngoscope. 2017;127:140-147.
      6. Zamani M, Gronhoj C, Jensen JS, et al. Survival and characteristrics of pediatric salivary gland cancer: A systematic review and meta-analysis. Pediatr Blood Cancer. 2019;66:e27543. doi:10.1002/pbc.27543.

      PEDONC-17: Pediatric Breast Masses

      For this condition imaging is medically necessary based on the following criteria:

      Less than 1% of pediatric breast lesions are malignant, and advanced imaging is generally not recommended without histological confirmation of malignancy.

      Ultrasound (CPT® 76641 and CPT® 76642) is the primary and preferred modality used for evaluation of pediatric breast masses

      Mammography has limited utility in pediatric breast mass evaluation due to the high mammographic breast density in this age group, and the risk of the radiation exposure outweighs the benefit of this modality. As a result, mammography is NOT recommended for evaluation of pediatric or adolescent breast masses.

        ® BI-RADS classification may overstate the risk of malignancy or need for biopsy in pediatric members.

      MRI has very limited utility in evaluation of pediatric breast masses prior to biopsy, but may be indicated in rare cases for surgical planning when ultrasound is non-diagnostic.
        ® All advanced imaging requests for pediatric breast masses should be forwarded for Medical Director review.

      Pediatric members with confirmed breast cancer should be imaged according to section Adult Oncology Imaging Policy (Policy #155 in the Radiology Section); ONC-11: Breast Cancer.

      References

      1. Rodriguez-Galindo C, Furman WL, Pappo AS. Rare pediatric tumors. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th edition. Philadelphia, PA: Wolters Kluwer; 2016:946-966.
      2. Goldberg JM, Pappo AS, Bishop M. Rare Tumors of Childhood. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th edition. Philadelphia, PA: Elsevier Saunders; 2015:2123-2145.
      3. Kaneda HJ, Mack J, Kasales CJ, Schetter S. Pediatric and adolescent breast masses: a review of pathophysiology, imaging, diagnosis, and treatment. AJR Am J Roentgenol. 2013;200(2):W204-W212. doi:10.2214/AJR.12.9560.
      4. Koning JL, Davenport KP, Poole PS, Kruk PG, Grabowski JE. Breast Imaging-Reporting and Data System (BI-RADS) classification in 51 excised palpable pediatric breast masses. J Pediatr Surg. 2015;50(10):1746-1750. doi:10.1016/j.jpedsurg.2015.02.062.
      5. Siegel MJ, Chung EM. Breast. In: Seigel MJ, ed. Pediatric Sonography. 5th ed. Philadelphia, PA: Wolters Kluwer; 2019:196-210.
      6. Siegel MJ, Chung EM. Breast masses in children and adolescents. Applied Radiology. 2017;46(9):12-17.
      7. Johnson RH, Anders CK, Litton JK, Ruddy KJ, Bleyer A. Breast cancer in adolescents and young adults. Pediatr Blood Cancer. 2018;65:e27397. doi:10.1002/pmb.27397.
      8. Sanders LM, Sharma P, El Madany M, et al. Clinical breast concerns in low-risk pediatric patients: practice review with proposed recommendations. Pediatr Radiol. 2018;48:186-195. doi:10.1007/s00247-017-4007-6.

      PEDONC-18: Histiocytic Disorders

      PEDONC-18.1: Histiocytic Disorders – General Considerations
      PEDONC-18.2: Langerhans Cell Histiocytosis (LCH)
      PEDONC-18.3: Hemophagocytic Lymphohistiocytosis (HLH)
      PEDONC-18.4: Non-Langerhans Cell Histiocytoses

      PEDONC-18.1: Histiocytic Disorders – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      The majority of histiocytic disorders occurring in the pediatric population are either Langerhans Cell Histiocytosis (LCH) or Hemophagocytic Lymphohistiocytosis (HLH).

      The Non-Langerhans cell histiocytoses encompass a variety of diseases, and have limited imaging considerations except as specified later in this section.

      PEDONC-18.2: Langerhans Cell Histiocytosis (LCH)

      For this condition imaging is medically necessary based on the following criteria:

      Includes a heterogeneous group of disorders formerly known by other names, including histiocytosis X, eosinophilic granuloma, Letterer-Siwe Disease, Hand-Schuller-Christian Disease, and diffuse reticuloendotheliosis. LCH has a widely variable clinical presentation, ranging from single indolent lesions to disseminated multisystem disease.

      Most common sites of involvement are skin, bones, liver, lung, and pituitary, though other sites are possible.

      LCH Initial Imaging Studies:

      For all members:

        ® Chest X-ray (CXR)
        ® Abdominal ultrasound (CPT® 76700)
        ® Skeletal survey
          ¡ PET should not be used to replace skeletal survey in LCH
      MRI Brain without and with contrast (CPT® 70553) for any of the following:
        ® Headaches or visual or neurologic disturbances
        ® Polyuria/polydipsia or other endocrine abnormalities
        ® Skull or craniofacial (including jaw) bone involvement
        ® Otorrhea or hearing loss (CT Temporal Bone may be substituted if requested)
        ® Other signs or symptoms suggesting intracranial involvement, including neurodegeneration syndrome

      CT Chest with (CPT® 71260) or without contrast (CPT® 71250) for any of the following:
        ® Abnormal CXR
        ® Symptoms of pulmonary involvement and normal CXR

      MRI Abdomen without and with contrast (CPT® 74183) for any of the following:
        ® Elevated liver function tests (usually > 5x upper limit of normal)
        ® Abnormalities seen on abdominal ultrasound
        ® CT Abdomen with contrast (CPT® 74160) can be substituted if requested by ordering physician to avoid general anesthesia

      MRI Spine without and with contrast (Cervical-CPT® 72156, Thoracic-CPT® 72157, Lumbar-CPT® 72158) for any of the following:
        ® Vertebral lesions seen on skeletal survey
        ® Clinical symptoms (including back pain) suggesting spinal involvement and negative skeletal survey

      Whole body PET/CT (CPT® 78816) for any of the following:
        ® Multifocal bone involvement seen on skeletal survey
        ® Bone pain and negative skeletal survey
        ® Other clinical symptoms suggesting multisite disease

      Whole body Tc-99m bone scan (CPT®78306) can be approved in lieu of PET for the same indications if PET is unavailable

      LCH Treatment Response:

      Members with localized or single site disease are often treated only with local therapies or observed, and should be imaged according to surveillance guidelines

      Members receiving systemic therapy will usually undergo treatment for ~12 months. Treatment response is assessed using any modalities showing disease at initial diagnosis after ~6 weeks of treatment.

        ® Those with persistent measurable disease will usually be evaluated again after week 12 of therapy
          ¡ Once PET/CT shows no remaining FDG-avid lesions, additional PET imaging is not indicated
          ¡ As a general rule, both PET/CT and CT with contrast or MRI without and with contrast should not be approved for simultaneous treatment response evaluation without specific documentation showing that both are necessary
      Following the initial phase, members can have treatment response evaluation every ~3 months while receiving active treatment.
        ® Shorter interval imaging can be approved for documented signs or symptoms concerning for disease progression

      All members should have the following studies at the end of planned therapy:
        ® Chest X-ray (CXR)
        ® Abdominal ultrasound (CPT® 76700)
        ® Skeletal survey
        ® Repeat of all additional imaging studies positive at initial workup (except PET)

      PET is generally not indicated during active treatment for recurrent pediatric cancer. In rare circumstances, PET may be appropriate when results are likely to result in a treatment change for the member, including a change from active treatment to surveillance. These requests will be forwarded for Medical Director review.

      LCH Surveillance Imaging:

      Surveillance imaging is determined by areas of disease involvement.

      Bone involvement

        ® Plain x-ray of involved bony areas at 6 weeks, then at 3 and 6 months after completion of therapy
        ® Additional films are not necessary unless symptoms suggest new or recurrent disease
        ® PET is not indicated for surveillance, but can be considered to evaluate members with recurrent disease
        ® Skull or craniofacial (including jaw) bone involvement at diagnosis are at higher risk for CNS recurrence, and should be imaged according to CNS involvement section below

      Pulmonary involvement
        ® CXR every 6 months after completion of therapy
          ¡ CT Chest with (CPT® 71260) or without contrast (CPT® 71250) can be approved for new abnormalities on CXR or new pulmonary symptoms with a negative CXR
      CNS involvement
        ® CNS LCH has a particularly high rate of refractory and recurrent disease, and requires longer imaging surveillance
        ® MRI Brain without and with contrast (CPT® 70553) is indicated for members with previously documented measurable intracranial lesions at 6 weeks, 3 months, and 6 months after completion of all therapy.
          ¡ If negative at that time, continued surveillance is indicated at 1, 2, 4, 7, and 10 years after completion of all planned therapy
          ¡ If residual measurable intracranial lesions are present at 6 months, imaging can be repeated every 3 months until negative or unchanged on two consecutive studies, at which time the schedule in the previous bullet should begin
        ® MRI Brain without and with contrast (CPT® 70553) is indicated for members with documented hypothalamic-pituitary dysfunction at 1, 2, 4, 7, and 10 years after completion of all planned therapy.
          ¡ MRI can be approved at any time for worsening neurologic symptoms
        ® Intraspinal lesions are rare, but should be imaged according to the same guidelines as brain imaging using MRI without and with contrast of all involved spine levels

      Liver involvement
        ® Persistent liver involvement is rare, and imaging after completion of LCH therapy will be highly individualized depending on degree of liver dysfunction and plans for supportive therapy or liver transplant
        ® Most members with liver involvement will receive surveillance Abdominal ultrasound (CPT® 76700) every 6 months
      PEDONC-18.3: Hemophagocytic Lymphohistiocytosis (HLH)

      For this condition imaging is medically necessary based on the following criteria:

      Advanced imaging requests for HLH should be forwarded for Medical Director review.

      There are no standard imaging studies required for the diagnosis and initial evaluation of HLH. Most cases are diagnosed with a combination of physical findings, laboratory testing, and bone marrow evaluation. Advanced imaging studies may be necessary to assess organ dysfunction as HLH commonly affects the liver, spleen, and bone marrow, and less commonly the kidneys, lungs, and brain

      Common studies that may be indicated in the initial evaluation of HLH include:

        ® Abdominal ultrasound (CPT® 76700)
        ® CT Abdomen and/or Pelvis (contrast as requested)
        ® MRI Abdomen (CPT® 74183) and/or Pelvis (CPT® 72197) without and with contrast
        ® CXR
        ® CT Chest with contrast (CPT® 71260)
        ® MRI Brain without and with contrast (CPT® 70553)

      It is NOT required to perform ultrasound or plain film in a stepwise fashion if CT or MRI is planned as members with HLH can deteriorate rapidly.

      There is no established standard role for PET in the diagnosis or treatment response evaluation of HLH

        ® Secondary HLH is very difficult to treat if the primary cause is not concurrently treated
        ® In these cases, if conventional imaging has been completed and is unrevealing, whole body PET/CT (CPT® 78816) can be considered for the purpose of identifying a site for tissue diagnosis of a primary source of infection or malignancy
        ® If a malignancy is identified as the inciting factor for HLH, additional imaging decisions for that malignancy should be based on the appropriate diagnosis-specific guidelines
      PEDONC-18.4: Non-Langerhans Cell Histiocytoses

      For this condition imaging is medically necessary based on the following criteria:

      Includes diagnoses such as juvenile xanthogranuloma (JXG), sinus histiocytosis with lymphadenopathy (Rosai-Dorfman Disease, RDD), and Erdheim-Chester Disease (ECD).

      In general, these are localized cutaneous or nodal disease without need for regular advanced imaging, but important exceptions are listed in this section.

      Juvenile Xanthogranuloma (JXG):

      Generally involves only skin or cervical nodes, and involutes spontaneously, imaging of involved nodal areas may be appropriate using CT with contrast of appropriate area

      Systemic JXG is associated with multi-organ involvement and imaging studies may include:

        ® MRI Brain (CPT® 70553) and/or Orbits (CPT® 70543) without and with contrast
        ® CT Neck (CPT® 70491), Chest (CPT® 71260), and/or Abdomen (CPT® 74160) with contrast

      There is no established role for PET in the diagnosis or treatment of JXG

      Rosai-Dorfman Disease (RDD):

      Characterized by bulky adenopathy (usually cervical) with frequent systemic involvement

      Appropriate imaging studies may include:

      MRI Brain (CPT® 70553) and/or Orbits (CPT® 70543) without and with contrast

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations)

      CT Neck (CPT® 70491), Chest (CPT® 71260) and/or Abdomen/Pelvis (CPT® 74177) with contrast

      There is no established role for PET in the diagnosis or treatment of RDD, but whole body PET/CT (CPT® 78816) may be approved if PET/CT will provide critical information for major treatment decision making that cannot be obtained using conventional imaging or biopsy.

        ® Because of the paucity of evidence for PET in RDD, PET/CT should not be used to replace tissue confirmation for any clinical scenario in RDD.
        ® These requests will be forwarded for Medical Director review.

      There is no established role for routine surveillance imaging of asymptomatic members after treatment for RDD, but CT with contrast can be approved for evaluation of new or worsening clinical symptoms suggesting recurrent disease

      Erdheim-Chester Disease (ECD):

      An aggressive histiocytic disorder with overall poor prognosis that is characterized by long bone involvement with frequent spread to multiple organs

      ECD Initial Imaging Studies:

      Appropriate imaging studies at initial diagnosis may include:

      MRI Brain (CPT® 70553) and/or Orbits (CPT® 70543) without and with contrast

      Nuclear bone scan (See PEDONC-1.3: Modality General Considerations)

      Whole body PET/CT (CPT® 78816)

      CT Neck (CPT® 70491), Chest (CPT® 71260) and/or Abdomen/Pelvis (CPT® 74177) with contrast

      CTA or MRA of Chest (CPT® 72175 or CPT® 71555) or Abdomen (CPT® 74175 or CPT® 74185) to evaluate vascular tree involvement

      Cardiac MRI without and with contrast (CPT® 75561)

      ECD Treatment Response:

      Most members will receive systemic therapy. Treatment response imaging can be approved every 3 months during active treatment using any modalities showing disease at initial diagnosis, including PET/CT.

        ® Once PET/CT shows no remaining FDG-avid lesions, additional PET imaging is not indicated unless conventional imaging studies are inconclusive and acute treatment decisions will be made based on PET results. These requests will be forwarded for Medical Director review.

      ECD Surveillance Imaging:

      Surveillance imaging can be approved every 3 months for the first year after completion of treatment, then every 6 months using any modalities showing disease at initial diagnosis.

      PET/CT is not supported for routine surveillance of ECD, but can be approved if conventional imaging is inconclusive for suspected recurrence. These requests will be forwarded for Medical Director review.

      References – PEDONC-18

      1. McClain KL, Allen CE, Hicks MJ. Histiocytic diseases. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:617-627.
      2. Degar BA, Fleming MD, Rollins BJ, et al. Histiocytoses. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:2100-2122.
      3. Haupt R, Minkov M, Astigarraga I, et al. Langerhans cell histiocytosis (LCH): guidelines for diagnosis, clinical work-up, and treatment for patients till the age of 18 years. Pediatr Blood Cancer. 2013;60(2):175-184. doi:10.1002/pbc.24367.
      4. Chandrakasan S, Filipovich AH. Hemophagocytic Lymphohistiocytosis: advances in pathophysiology, diagnosis, and treatment. J Pediatr. 2013;163(5):1253-1259. doi:10.1016/j.jpeds.2013.06.053.
      5. Phillips M, Allen C, Gerson P, et al. Comparison of FDG-PET scans to conventional radiography and bone scans in management of Langerhans cell histiocytosis. Pediatr Blood Cancer. 2009;52(1):97-101. doi:10.1002/pbc.21782.
      6. Chellapandian D, Shaiki F, van den Bos C, et al. Management and outcome of patients with langerhans cell histiocytosis and single-bone CNS-risk lesions: a multi-institutional retrospective study. Pediatr Blood Cancer. 2015;62(12):2162-2166. doi:10.1002/pbc.25645.
      7. Allen CE, Ladisch S, McClain KL. How I treat Langerhans cell histiocytosis. Blood. 2015;126(1):26-35. doi:10.1182/blood-2014-12-569301.
      8. Schram AM, Berliner N. How I treat Hemophagocytic lymphohistiocytosis in the adult patient. Blood. 2015;125(19):2908-2914. doi:10.1182/blood-2015-01-551622.
      9. Weitzman S, Jaffe R. Uncommon histiocytic disorders: the non-Langerhans cell histiocytoses. Pediatr Blood Cancer. 2005;45(3):256-264. doi:10.1002/pbc.20246.
      10. Diamond EL, Dagna L, Hyman DM, et al. Consensus guidelines for the diagnosis and clinical management of Erdheim-Chester disease. Blood. 2014;124(4):483-492. doi:10.1182/blood-2014-03-561381.

      PEDONC-19: Long Term Pediatric Cancer Survivors

      PEDONC-19.1: Long Term Pediatric Cancer Survivors – General Considerations
      PEDONC-19.2: Cardiotoxicity and Echocardiography
      PEDONC-19.3: Second Malignant Neoplasms (SMN)
      PEDONC-19.4: Osteonecrosis in Long Term Cancer Survivors

      PEDONC-19.1: Long Term Pediatric Cancer Survivors – General Considerations

      For this condition imaging is medically necessary based on the following criteria:

      This section applies to members who have passed the end of the surveillance imaging period for their specific cancer, or 5 years after completion of therapy, whichever occurs first.

      As these are long term survivors, many members falling under this guideline section will have reached adult age. However, these guidelines relate specifically to late effects of childhood cancer treatment and should be applied to all long term childhood cancer survivors regardless of current age.

      The Children’s Oncology Group has published comprehensive guidelines for the management of long-term childhood cancer survivors, and these are available at: http://www.survivorshipguidelines.org.

      A summary of cancer treatment should be available for all members in this category and should generally include, at minimum:

      Type of cancer and stage

      Dates of diagnosis, recurrence, cancer-related surgeries, beginning and end dates of chemotherapy, radiotherapy, and/or stem cell transplant

      Protocol number used for treatment and cumulative chemotherapy drug dose exposures

      Cumulative radiation dose, fraction number, modality, and field exposure
      Annual detailed history and complete physical examination is a critical component of cancer survivorship care and along with laboratory testing serves as the primary method of screening for the majority of late effects.

      Advanced imaging for asymptomatic screening is not routinely indicated except as specified in this section.

      Imaging requests related to new clinical signs or symptoms in a long term cancer survivor not explicitly covered in this section should be reviewed according to the guideline for the member’s cancer type or the relevant non-malignant clinical problem.



      PEDONC-19.2: Cardiotoxicity and Echocardiography

      For this condition imaging is medically necessary based on the following criteria:

      Exposure to cardiotoxic anthracycline chemotherapy agents is common in pediatric Oncology due to the high success rate of this drug class in the treatment of pediatric cancers. Screening echocardiography (CPT® 93306, CPT® 93307, or CPT® 93308) for life is indicated after exposure to anthracycline chemotherapy or cardiac exposure to radiotherapy.

      Cardiotoxic drugs include the following:

      Doxorubicin
      Daunorubicin
      Idarubicin
      Epirubicin
      Mitoxantrone

      Cardiac risk is assessed based on the age of the member at the time of treatment initiation, the cumulative drug exposure expressed as doxorubicin equivalent mg/m2, and the presence or absence of radiotherapy exposure to cardiac muscle.
      SCREENING ECHOCARDIOGRAM INDICATIONS
      Age at time of Exposure
      Cumulative Doxorubicin Equivalent Dose
      Cumulative radiation dose to cardiac muscle
      Echocardiogram frequency
      All ages
      None
      None
      None
      0-0.99 years
      ≥250 mg/m2
      None
      Annual
      0-249 mg/m2
      Any dose
      Annual
      0-249 mg/m2
      None
      Every 2 years
      1-4.99 years
      ≥250 mg/m2
      Any dose
      Annual
      0-249 mg/m2
      15+ Gy
      Annual
      0-14.99 Gy
      Every 2 years
      None
      35+ Gy
      Annual
      15-34.99 Gy
      Every 2 years
      0-14.99 Gy
      Every 5 years
      5+ years
      ≥250 mg/m2
      Any dose
      Every 2 years
      0-249 mg/m2
      15+ Gy
      Every 2 years
      0-14.99 Gy
      Every 5 years
      None
      35+ Gy
      Every 2 years
      15-34.99 Gy
      Every 5 years
      0-14.99 Gy
      None
      All ages with known ventricular dysfunction
      Annual

      Stress echocardiography is not indicated as a screening study for anthracyclines cardiotoxicity in the absence of coronary artery disease symptoms. See Adult Cardiac Imaging Policy (Policy #149 in the Radiology Section); CD-1.4: Stress Testing with Imaging – Indications for imaging guidelines.

      Female cancer survivors who are pregnant or planning to become pregnant:

        ® If any of the following are present, echocardiogram is recommended as a baseline exam, repeated as needed during and immediately following pregnancy:
          ¡ ≥250 mg/m2 cumulative doxorubicin equivalent exposure
          ¡ ≥35 Gy chest radiotherapy
          ¡ Any cardiotoxic drug exposure from the list above AND ≥15 Gy chest radiotherapy
      PEDONC-19.3: Second Malignant Neoplasms (SMN)

      For this condition imaging is medically necessary based on the following criteria:

      SMN—Breast Cancer

      Clinical breast exam every 6 months supplemented with:

      Annual Breast MRI (CPT® 77049) and annual mammogram is recommended beginning at age 25 or 8 years after completion of radiotherapy (whichever occurs later) for members receiving a cumulative radiation exposure of ≥ 20 Gy in the following fields for any pediatric cancer type except Wilms tumor:

        ® Chest (thorax)
        ® Whole lung
        ® Mediastinal
        ® Axilla
        ® Mini-mantle, mantle, or extended mantle
        ® Total (TLI) or subtotal (SLTI) lymphoid irradiation
        ® Total body irradiation (TBI)

      Annual breast MRI (CPT® 77049) and annual mammogram is recommended beginning at age 25 or 8 years after completion of radiotherapy (whichever occurs later) for members receiving ≥ 12 Gy of whole lung radiation for treatment of Wilms tumor

      SMN – CNS Tumors

      These are associated with radiation exposure to the brain and with neurofibromatosis.

      Routine surveillance of most completely asymptomatic members with normal neurologic exams is not supported by evidence

        ® MRI Brain without and with contrast (CPT® 70553) can be approved every 2 years after completion of radiotherapy for members with NF1 or NF2

      MRI Brain without and with contrast (CPT® 70553) should be approved if requested for any member with history of brain radiotherapy and new neurologic symptoms including simple headache

      MRI Cervical (CPT® 72156), Thoracic (CPT® 72157), and Lumbar spine (CPT® 72158) without and with contrast should be approved if requested for any member with history of spine radiotherapy and new neurologic symptoms including change in quality of pain

        ® MRI Spine can be performed with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) if being performed immediately following a contrast-enhanced MRI Brain

      For members with history of brain radiotherapy and persistent neurologic symptoms, annual MRI Brain without and with contrast (CPT® 70553) can be approved

      For members with history of spine radiotherapy and persistent neurologic symptoms, annual MRI Cervical (CPT® 72156), Thoracic (CPT® 72157), and Lumbar spine (CPT® 72158) without and with contrast can be approved

        ® MRI Spine can be performed with contrast only (Cervical-CPT® 72142, Thoracic-CPT® 72147, Lumbar-CPT® 72149) if being performed immediately following a contrast-enhanced MRI Brain

      SMN—Colorectal Cancer

      Colonoscopy is recommended every 5 years beginning at age 30 or 5 years after radiation exposure (whichever is later) for members with ≥ 30 Gy radiation exposure to the following fields:


        ® Thoracic, Lumbar, Sacral, or Whole Spine
        ® Abdomen
        ® Pelvis
        ® Total Body Irradiation (TBI)

      Colonoscopy is also recommended every 5 years beginning at age 30 or 5 years after radiation exposure (whichever is later) for members with:


        ® Personal history of ulcerative colitis, GI malignancy, adenomatous polyps, or hepatoblastoma
        ® Familial polyposis
        ® Family history of colorectal cancer or polyps in a first degree (parent or sibling) relative
        While the American Cancer Society recently added computed tomographic colonography (CTC) (AKA “Virtual Colonoscopy”) as an acceptable option for colorectal cancer screening of average-risk adults, the National Comprehensive Cancer Network and United States Preventive Services Task Force concluded that data was too premature to warrant its use in screening. Colonoscopy remains the preferred screening modality for survivors at highest risk of colorectal cancer.

        PEDONC-19.4: Osteonecrosis in Long Term Cancer Survivors

        For this condition imaging is medically necessary based on the following criteria:

        Osteonecrosis is associated with corticosteroid, chemotherapy, and radiation exposure during treatment for ALL, NHL, and allogeneic HSCT in pediatrics. Osteonecrosis occurs primarily in hips, knees, and ankles and is frequently multifocal.

        Osteoradionecrosis of the jaw can occur in members receiving radiotherapy to the mandible or maxilla; those receiving ≥ 40 Gy are at highest risk. Although unusual, it can also occur in any bone without symptoms. It is rare in other disease types.

        Plain films of symptomatic areas are indicated prior to advanced imaging.

        Routine bone density screening using DEXA or Quantitative CT screening has not been well normalized in the pediatric population, but imaging can be approved for those with symptoms to suggest bone density issues

          ® DEXA or Quantitative CT screening is generally not recommended until age 18 unless a specific intervention will be planned based on the imaging results.

        Serial advanced imaging is not indicated in osteonecrosis without specific documentation regarding how the advanced imaging will change current member management
          ® When advanced imaging is necessary for acute management decisions, MRI without contrast of the affected joint(s) can be approved.
          ® Surveillance imaging of asymptomatic members to detect osteonecrosis has not been shown to impact member outcomes, and it is not standard to alter treatment based on imaging findings alone without symptoms.
            ¡ Follow up MRI of incidentally discovered osteonecrosis findings in asymptomatic members has not been shown to impact member outcomes and is not necessary
        See PEDONC-3.2: Acute Lymphoblastic Leukemia (ALL) for information on imaging osteonecrosis in ALL members during active treatment.

        References

        1. Landier W, Armenian SH, Meadows AT, et al. Late effects of childhood cancer and its treatment. In: Pizzo PA, Poplack DG, eds. Principles and Practice of Pediatric Oncology. 7th ed. Philadelphia, PA: Wolters Kluwer; 2016:1173-1196.
        2. Vrooman L, Diller L, and Kenney LB. Childhood cancer survivorship. In: Orkin SH, Fisher DE, Ginsburg D, Look AT, Lux SE, Nathan DG, eds. Nathan and Oski’s Hematology and Oncology of Infancy and Childhood. 8th ed. Philadelphia, PA: Elsevier Saunders; 2015:2397-2434.
        3. Children’s Oncology Group. Long-term follow up guidelines for survivors of childhood, adolescent and young adult cancers, version 5.0. Monrovia, CA: Children’s Oncology Group; October 2018; Available on-line: www.survivorshipguidelines.org
        4. Ryerson AB, Border WL, Wasilewski-Masker K, et al. Assessing anthracycline-treated childhood cancer survivors with advanced stress echocardiography. Pediatr Blood Cancer. 2015;62(3):502-508. doi:10.1002/pbc.25328.
        5. Tieu MT, Cigsar C, Ahmed S, et al. Breast cancer detection among young survivors of pediatric Hodgkin lymphoma with screening magnetic resonance imaging. Cancer. 2014;120(16):2507-2513. doi:10.1002/cncr.28747.
        6. Lange JM, Takashima JR, Peterson SM, Kalapurakal JA, Green DM, Breslow NE. Breast cancer in female survivors of Wilms tumor: a report from the national Wilms tumor late effects study. Cancer. 2014;120(23):3722-3730. doi:10.1002/cncr.28908.
        7. Bevers TB, Helvie M, Bonaccio E, et al. National Comprehensive Cancer Network (NCCN) Guidelines Version 1.2019—May 17, 2019, Breast Cancer Screening and Diagnosis, available at: https://www.nccn.org/professionals/physician_gls/pdf/breast-screening.pdf, Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines™) for Breast Cancer Screening and Diagnosis V1.2019 5/17/19. ©2019 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines™ and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines™, go online to NCCN.org.
        8. Recht M, Mostoufi-Moab S, McFadden J, et al. Bone health in pediatric hematology-oncology. In: American Society of Pediatric Hematology/Oncology Annual Meeting. Chicago; 2014.
        9. Bhatia S, Armenian SH, Armstrong GT, et al. Collaborative research in childhood cancer survivorship: the current landscape. J Clin Oncol. 2015;33(27):3055-3064. doi:10.1200/JCO.2014.59.8052.
        10. Chavhan GB, Babyn PS, Nathan PC, Kaste SC. Imaging of acute and subacute toxicities of cancer therapy in children. Pediatr Radiol. 2016;46(1):9-20. doi:10.1007/s00247-015-3454-1.
        11. Karol SE, Mattano LA, Yang W, et al. Genetic risk factors for the development of osteonecrosis in children under age 10 treated for acute lymphoblastic leukemia. Blood Journal. 2016;127(5):558-564. doi:10.1182/blood-2015-10-673848.
        12. Marcucci G, Beltrami G, Tamburini A, et al. Bone health in childhood cancer: review of the literature and recommendations for the management of bone health in childhood cancer survivors. Ann Oncol. 2019;30:908-920. doi:10.1093/annonc/mdz120.
        13. Sanders LM, Sharma S, El Madany M, King AB, Goodman KS, Sanders AE. Clinical breast concerns in low-risk pediatric patients: practice review with proposed recommendations. Pediatr Radiol. 2018(48):186–195. doi:10.1007/s00247-017-4007-6.

        Medicaid Coverage:

        For members enrolled in Medicaid and NJ FamilyCare plans, Horizon BCBSNJ applies the above medical policy.

        FIDE SNP: For members enrolled in a Fully Integrated Dual Eligible Special Needs Plan (FIDE-SNP): (1) to the extent the service is covered under the Medicare portion of the member’s benefit package, the above Medicare Coverage statement applies; and (2) to the extent the service is not covered under the Medicare portion of the member’s benefit package, the above Medicaid Coverage statement applies

        ________________________________________________________________________________________

        Horizon BCBSNJ Medical Policy Development Process:

        This Horizon BCBSNJ Medical Policy (the “Medical Policy”) has been developed by Horizon BCBSNJ’s Medical Policy Committee (the “Committee”) consistent with generally accepted standards of medical practice, and reflects Horizon BCBSNJ’s view of the subject health care services, supplies or procedures, and in what circumstances they are deemed to be medically necessary or experimental/ investigational in nature. This Medical Policy also considers whether and to what degree the subject health care services, supplies or procedures are clinically appropriate, in terms of type, frequency, extent, site and duration and if they are considered effective for the illnesses, injuries or diseases discussed. Where relevant, this Medical Policy considers whether the subject health care services, supplies or procedures are being requested primarily for the convenience of the covered person or the health care provider. It may also consider whether the services, supplies or procedures are more costly than an alternative service or sequence of services, supplies or procedures that are at least as likely to produce equivalent therapeutic or diagnostic results as to the diagnosis or treatment of the relevant illness, injury or disease. In reaching its conclusion regarding what it considers to be the generally accepted standards of medical practice, the Committee reviews and considers the following: all credible scientific evidence published in peer-reviewed medical literature generally recognized by the relevant medical community, physician and health care provider specialty society recommendations, the views of physicians and health care providers practicing in relevant clinical areas (including, but not limited to, the prevailing opinion within the appropriate specialty) and any other relevant factor as determined by applicable State and Federal laws and regulations.

        ___________________________________________________________________________________________________________________________

        Index:
        Pediatric Oncology Imaging Policy
        Oncology Imaging Policy, Pediatric
        Computed Tomography, Oncology, Pediatric
        CT, Oncology, Pediatric
        Computed Tomography Angiography, Oncology, Pediatric
        CTA, Oncology, Pediatric
        Magnetic Resonance Imaging, Oncology, Pediatric
        MRI, Oncology, Pediatric
        Magnetic Resoance Angiography, Oncology, Pediatric
        MRA, Oncology, Pediatric
        Positron Emission Tomography, Oncology, Pediatric
        PET, Oncology, Pediatric
        Nuclear Medicine Imaging, Oncology, Pediatric
        Ultrasound, Oncology, Pediatric
        Gallium-68 Dotatate PET, Pediatric Oncology Imaging
        Fluorine-18 Fluorodeoxyglucose PET, Pediatric Oncology Imaging
        18F-FDG PET, Pediatric Oncology Imaging
        68Ga-DOTATATE PET, Pediatric Oncology Imaging

        References:


        Codes:
        (The list of codes is not intended to be all-inclusive and is included below for informational purposes only. Inclusion or exclusion of a procedure, diagnosis, drug or device code(s) does not constitute or imply authorization, certification, approval, offer of coverage or guarantee of payment.)

        CPT*

          HCPCS

          * CPT only copyright 2020 American Medical Association. All rights reserved. CPT is a registered trademark of the American Medical Association.

          _________________________________________________________________________________________

          Medical policies can be highly technical and are designed for use by the Horizon BCBSNJ professional staff in making coverage determinations. Members referring to this policy should discuss it with their treating physician, and should refer to their specific benefit plan for the terms, conditions, limitations and exclusions of their coverage.

          The Horizon BCBSNJ Medical Policy Manual is proprietary. It is to be used only as authorized by Horizon BCBSNJ and its affiliates. The contents of this Medical Policy are not to be copied, reproduced or circulated to other parties without the express written consent of Horizon BCBSNJ. The contents of this Medical Policy may be updated or changed without notice, unless otherwise required by law and/or regulation. However, benefit determinations are made in the context of medical policies existing at the time of the decision and are not subject to later revision as the result of a change in medical policy

          ____________________________________________________________________________________________________________________________