Medulloblastoma and other central nervous system (CNS) embryonal tumors may begin in embryonic (fetal) cells that remain in the brain after birth.
There are different types of CNS embryonal tumors.
Pineoblastoma forms in cells of the pineal gland.
Certain genetic conditions increase the risk of childhood medulloblastoma.
Genetic counseling may be done for children with medulloblastoma or pineoblastoma.
Symptoms of medulloblastoma, other CNS embryonal tumors, and pineoblastoma depend on the child’s age and where the tumor is.
Tests that examine the brain and spinal cord are used to diagnose childhood medulloblastoma, other CNS embryonal tumors, and pineoblastoma.
A biopsy may be done to be sure of the diagnosis.
Certain factors affect prognosis (chance of recovery) and treatment options.
You may want to get a second opinion.
Medulloblastoma and other central nervous system (CNS) embryonal tumors may begin in embryonic (fetal) cells that remain in the brain after birth.
Medulloblastoma is a fast-growing tumor that forms in the cerebellum (the lower, back part of the brain). Medulloblastoma is the most common type of CNSembryonal tumor. CNS embryonal tumors are uncontrolled growths of cells in the brain. These tumors form in cells that are left over from fetal development, called embryonal cells. Pineoblastoma is a fast-growing type of brain tumor that forms in or around a tiny organ near the center of the brain called the pineal gland.
These tumors may be benign (not cancer) or malignant (cancer). Benign brain tumors grow and press on nearby areas of the brain but rarely spread to other parts of the brain. Malignant brain tumors are likely to grow quickly and spread into other parts of the brain. They may also spread to other parts of the body, but this is rare. When a tumor grows into and presses on an area of the brain or spreads to other parts of the brain, it may stop that part of the brain from working the way it should. Both benign and malignant brain tumors can cause serious signs or symptoms and need treatment.
Most medulloblastomas, other CNS embryonal tumors, and pineoblastomas in children are malignant. These tumors tend to spread through the cerebrospinal fluid to other parts of the brain and spinal cord.
Although cancer is rare in children, brain tumors are the second most common type of childhood cancer, after leukemia. This summary is about the treatment of primary brain tumors (tumors that begin in the brain).
EnlargeAnatomy of the inside of the brain, showing the pineal and pituitary glands, optic nerve, ventricles (with cerebrospinal fluid shown in blue), and other parts of the brain.
There are different types of CNS embryonal tumors.
The different types of CNS embryonal tumors include:
Medulloblastomas
Most CNS embryonal tumors are medulloblastomas. Medulloblastomas are fast-growing tumors that form in brain cells in the cerebellum. The cerebellum is at the lower back part of the brain between the cerebrum and the brain stem. The cerebellum controls movement, balance, and posture. It is rare for medulloblastomas to spread to the bone, bone marrow, lung, or other parts of the body.
Other types of CNS embryonal tumors (nonmedulloblastoma)
Other types of CNS embryonal tumors are fast-growing tumors and may form in brain cells anywhere in the brain, including the cerebrum, brain stem, or spinal cord. The cerebrum is at the top of the head and is the largest part of the brain. The cerebrum controls thinking, learning, problem-solving, emotions, speech, reading, writing, and voluntary movement. It is rare for these tumors to spread to the bone, bone marrow, lung, or other parts of the body.
There are many types of CNS embryonal (nonmedulloblastoma) tumors:
Cribriform neuroepithelial tumors
Cribriformneuroepithelial tumor forms in the ventricles in the brain. This tumor most often occurs in infants and young children. Cribriform neuroepithelial tumor occurs more often in boys.
Embryonal tumors with multilayered rosettes
Embryonal tumors with multilayered rosettes (ETMR) are rare tumors that form in the brain and spinal cord. ETMR most commonly occur in young children and are fast-growing tumors.
CNS neuroblastomas
CNS neuroblastomas are a very rare type of neuroblastoma that form in the nerve tissue of the cerebrum or the layers of tissue that cover the brain and spinal cord. CNS neuroblastomas may be large and spread to other parts of the brain or spinal cord.
CNS high-grade neuroepithelial tumor with a change in the BCOR gene
CNS high-grade neuroepithelial tumor is a very rare tumor that forms in the brain. This tumor occurs most often in children younger than 10 years, but can occur in older children and adolescents.
CNS Ewing sarcoma with a change in the CIC gene
CNS Ewing sarcoma is a very rare tumor found in the brain or spine. This tumor most often occurs in children younger than 10 years.
CNS high-grade neuroepithelial tumor with a change in the MN1 gene
CNS high-grade neuroepithelial tumor is a very rare tumor that forms in the brain or spinal cord. This tumor most often occurs in adolescents and females.
Medulloepitheliomas
Medulloepithelioma is a fast-growing tumor that usually forms in the brain, spinal cord, or nerves just outside the spinal column. It occurs most often in infants and young children.
CNS embryonal tumor with changes in the PLAGL gene
CNS embryonal tumor with changes in the PLAGL gene is a very rare tumor that forms in the brain. It affects both children and adults.
The pineal gland is a tiny organ in the center of the brain. The gland makes melatonin, a substance that helps control our sleep cycle. Pineoblastoma are usually malignant fast-growing tumors with cells that look very different from normal pineal gland cells. Pineoblastomas are not a type of CNS embryonal tumor but treatment for them is similar to treatment for CNS embryonal tumors.
Pineoblastoma is linked with inherited changes in the retinoblastoma (RB1) gene. A child with the inherited form of retinoblastoma (cancer that forms in the tissues of the retina) has an increased risk of pineoblastoma. When retinoblastoma forms at the same time as a tumor in or near the pineal gland, it is called trilateral retinoblastoma. MRI (magnetic resonance imaging) testing in children with retinoblastoma may detect pineoblastoma at an early stage when it can be treated successfully. It is rare for pineoblastoma to spread to the bone, bone marrow, lung, or other parts of the body.
Certain genetic conditions increase the risk of childhood medulloblastoma.
Childhood medulloblastoma is caused by certain changes to the way brain cells function, especially how they grow and divide into new cells. Often, the exact cause of the cell changes is unknown. Learn more about how cancer develops at What Is Cancer?
A risk factor is anything that increases the chance of getting a disease. Not every child with one or more of these risk factors will develop medulloblastoma. And it will develop in some children who don’t have a known risk factor.
The risk for medulloblastoma is increased in people who have any of the following inherited diseases:
Talk with your child’s doctor if you think your child may be at risk.
Genetic counseling may be done for children with medulloblastoma or pineoblastoma.
It may not be clear from the family medical history whether a child with a brain tumor has an inherited condition that increased their risk. Genetic counselors and other specially trained health professionals can discuss your child’s diagnosis and the family’s medical history to understand:
your options for ELP1, APC, SUFU, PTCH1, TP53, PALB2, or BRCA2 gene testing if your child has medulloblastoma
your options for RB1 or DICER1 gene testing if your child has pineoblastoma
the risk of other cancers for your child
the risk of cancer for your child’s siblings
the risks and benefits of learning genetic information
Genetic counselors can also help you cope with your child’s genetic testing results, including how to discuss the results with family members.
Symptoms of medulloblastoma, other CNS embryonal tumors, and pineoblastoma depend on the child’s age and where the tumor is.
Children may not have symptoms of medulloblastoma, other CNS embryonal tumors, or pineoblastoma until the tumor has grown bigger. It’s important to check with your child’s doctor if your child has:
loss of balance, trouble walking, lack of coordination, or slow speech
a headache, especially in the morning, or headache that goes away after vomiting
Infants and young children with these tumors may be irritable or grow slowly. Also they may not eat well or meet developmental milestones such as sitting, walking, and talking in sentences. These tumors may also cause an increase in the size of an infant’s head.
These symptoms may be caused by problems other than medulloblastoma, other CNS embryonal tumors, or pineoblastoma. The only way to know is to see your child’s doctor.
Tests that examine the brain and spinal cord are used to diagnose childhood medulloblastoma, other CNS embryonal tumors, and pineoblastoma.
If your child has symptoms that suggest medulloblastoma, another type of CNS embryonal tumor, or pineoblastoma, the doctor will need to find out if these are due to cancer or another problem. They will ask about your child’s personal and family health history and do a physical exam. Depending on the results, they may recommend other tests. If your child is diagnosed with medulloblastoma, another type of CNS embryonal tumor, or pineoblastoma, the results of these tests will help you and your child’s doctor plan treatment.
The tests used to diagnose medulloblastoma, other CNS embryonal tumors, and pineoblastoma may include:
MRI (magnetic resonance imaging) of the brain and spinal cord with gadolinium is a procedure that uses a magnet, radio waves, and a computer to make a series of detailed pictures of areas inside the brain and spinal cord. A substance called gadolinium is injected into a vein. The gadolinium collects around the cancer cells so they show up brighter in the picture. This procedure is also called nuclear magnetic resonance imaging (NMRI). Sometimes magnetic resonance spectroscopy (MRS) is done during the MRI scan to look at the chemicals in brain tissue.
CT scan (CAT scan) uses a computer linked to an x-ray machine to make a series of detailed pictures inside the body from different angles. A dye may be injected into a vein or swallowed to help the organs or tissues show up more clearly. This procedure is also called computed tomography, computerized tomography, or computerized axial tomography. Learn more about Computed Tomography (CT) Scans and Cancer.
Lumbar puncture is a procedure used to collect cerebrospinal fluid (CSF) from the spinal column. This is done by placing a needle between two bones in the spine and into the lining around the spinal cord to remove a sample of CSF. The sample of CSF is checked under a microscope for signs of tumor cells. The sample may also be checked for the amounts of protein and glucose. A higher-than-normal amount of protein or lower-than-normal amount of glucose may be a sign of a tumor. This procedure is also called an LP or spinal tap. EnlargeLumbar puncture. A patient lies in a curled position on a table. After a small area on the lower back is numbed, a spinal needle (a long, thin needle) is inserted into the lower part of the spinal column to remove cerebrospinal fluid (CSF, shown in blue). The fluid may be sent to a laboratory for testing.
A biopsy may be done to be sure of the diagnosis.
If doctors think your child may have medulloblastoma, another type of CNS embryonal tumor, or pineoblastoma, a biopsy may be done. The biopsy is done by removing part of the skull and using a needle to remove a sample of tissue. Sometimes, a computer-guided needle is used to remove the tissue sample. A pathologist views the tissue under a microscope to look for cancer cells. If cancer cells are found, the doctor may remove as much tumor as safely possible during the same surgery. The piece of skull is usually put back in place after the procedure.
EnlargeCraniotomy. An opening is made in the skull and a piece of the skull is removed to show part of the brain.
The following tests may be done on the sample of tissue that is removed:
Immunohistochemistry is a laboratory test that uses antibodies to check for certain antigens (markers) in a sample of a patient’s tissue. The antibodies are usually linked to an enzyme or a fluorescent dye. After the antibodies bind to a specific antigen in the tissue sample, the enzyme or dye is activated, and the antigen can then be seen under a microscope. This type of test is used to help diagnose cancer and to help tell one type of cancer from another type of cancer.
Molecular testing checks for certain genes, proteins, or other molecules in a sample of tissue, blood, or bone marrow. Molecular tests also check for certain changes in a gene or chromosome that may cause or affect the chance of developing medulloblastoma, another type of embryonal tumor, or pineoblastoma. A molecular test may be used to help plan treatment, find out how well treatment is working, or make a prognosis. Children with medulloblastoma, another type of embryonal tumor, or pineoblastoma may be eligible for molecular testing through the Molecular Characterization Initiative.
The Molecular Characterization Initiative offers free molecular testing to children, adolescents, and young adults with certain types of newly diagnosed cancer. The program is offered through NCI’s Childhood Cancer Data Initiative. To learn more, visit About the Molecular Characterization Initiative.
Certain factors affect prognosis (chance of recovery) and treatment options.
If your child has been diagnosed with medulloblastoma, other CNS embryonal tumor, or pineoblastoma, you likely have questions about how serious the cancer is and your child’s chances of survival. The likely outcome or course of a disease is called prognosis.
The prognosis and treatment options depend on:
the type of tumor and where it is in the brain
whether the cancer has spread within the brain and spinal cord when the tumor is found
the age of the child when the tumor is found
how much of the tumor remains after surgery
whether there are certain changes in the chromosomes, genes, or brain cells
whether the tumor has just been diagnosed or has recurred (come back)
No two people are alike, and responses to treatment can vary greatly. Your child’s cancer care team is in the best position to talk with you about your child’s prognosis.
You may want to get a second opinion.
You may want to get a second opinion to confirm your child’s diagnosis and treatment plan. If you seek a second opinion, you will need to get important medical test results and reports from the first doctor to share with the second doctor. The second doctor will review the genetic test results, pathology report, slides, and scans. This doctor may agree with the first doctor, suggest changes to the treatment plan, or provide more information about your child’s tumor.
To learn more about choosing a doctor and getting a second opinion, see Finding Cancer Care. You can contact NCI’s Cancer Information Service via chat, email, or phone (both in English and Spanish) for help finding a doctor or hospital that can provide a second opinion. For questions you might want to ask at your child’s appointments, see Questions to Ask Your Doctor About Cancer.
Staging Childhood Medulloblastoma, Other Central Nervous System Embryonal Tumors, and Pineoblastoma
Key Points
Medulloblastoma, other CNS embryonal tumors, and pineoblastoma in children are treated based on the tumor type and the child’s age.
Treatment of medulloblastoma in children older than 3 years also depends on whether the tumor is average risk or high risk.
Average risk
High risk
The results of the tests and procedures done to diagnose medulloblastoma, other CNS embryonal tumors, and pineoblastoma in children are used to plan cancer treatment.
Sometimes childhood medulloblastoma and other central nervous system embryonal tumors come back after treatment.
Medulloblastoma, other CNS embryonal tumors, and pineoblastoma in children are treated based on the tumor type and the child’s age.
Cancer stage describes the extent of cancer in the body, such as the size of the tumor, whether it has spread, and how far it has spread from where it first formed. There is no staging system used for childhood medulloblastoma, other central nervous system (CNS) embryonal tumors, or pineoblastoma, but the tests and procedures done to diagnose the cancer are also used to help plan treatment.
Treatment of other CNS embryonal tumors and pineoblastoma in children is based on the child’s age. Children aged 3 years and younger may be given different treatment than children older than 3 years.
Treatment of medulloblastoma in children older than 3 years also depends on whether the tumor is average risk or high risk.
Average risk
Medulloblastomas are called average risk when all of the following are true:
The tumor was completely removed by surgery or there was only a very small amount remaining.
The cancer has not spread to other parts of the body.
High risk
Medulloblastomas are called high risk if any of the following are true:
Some of the tumor was not removed by surgery.
The cancer has spread to other parts of the brain or spinal cord or to other parts of the body.
In general, cancer is more likely to recur (come back) after treatment in patients with a high-risk tumor.
The results of the tests and procedures done to diagnose medulloblastoma, other CNS embryonal tumors, and pineoblastoma in children are used to plan cancer treatment.
If your child is diagnosed with medulloblastoma, another type of CNS embryonal tumor, or pineoblastoma, they will be referred to a pediatric oncologist/neuro-oncologist. This is a doctor who specializes in staging and treating childhood cancers. They will recommend tests to determine the extent (stage) of cancer. Some of the tests used to diagnose the cancer are repeated after surgery. This is to find out how much tumor remains after surgery and to see if the cancer has spread from the brain to the spine or other parts of the body. It is important to know if the cancer has spread in order to plan the best treatment. Learn more about diagnostic tests in the General Information section.
The following tests may be used to find out if the cancer has spread beyond the brain and spinal cord:
Bone marrow aspiration and biopsy are procedures in which a sample of bone marrow and bone is removed from the hipbone or breastbone using a special needle. A pathologist views the sample under a microscope to look for signs of cancer. A bone marrow aspiration and biopsy are only done when there are signs the cancer has spread to the bone marrow. EnlargeBone marrow aspiration and biopsy. After a small area of skin is numbed, a bone marrow needle is inserted into the child’s hip bone. Samples of blood, bone, and bone marrow are removed for examination under a microscope.
Bone scan is a procedure to check if there are rapidly dividing cells, such as cancer cells, in the bone. A very small amount of radioactive material is injected into a vein and travels through the bloodstream. The radioactive material collects in the bones with cancer and is detected by a scanner. A bone scan is only done when there are signs or symptoms that the cancer has spread to the bone.
Sometimes childhood medulloblastoma and other central nervous system embryonal tumors come back after treatment.
Childhood medulloblastoma and other types of CNS embryonal tumors most often recur (come back) within 3 years after treatment but may come back many years later. Recurrent childhood medulloblastoma and other CNS embryonal tumors may come back in the same place as the original tumor and/or in a different place in the brain or spinal cord.
Treatment Option Overview
Key Points
There are different types of treatment for children who have medulloblastoma and other central nervous system (CNS) embryonal tumors.
Children who have medulloblastoma, other CNS embryonal tumors, and pineoblastoma should have their treatment planned by a team of health care providers who are experts in treating brain tumors in children.
The following types of treatment may be used:
Surgery
Radiation therapy
Chemotherapy
High-dose chemotherapy with autologous stem cell rescue
Targeted therapy
New types of treatment are being tested in clinical trials.
There are different types of treatment for children who have medulloblastoma and other central nervous system (CNS) embryonal tumors.
There are different types of treatment for children and adolescents with medulloblastoma, other types of CNSembryonal tumors, or pineoblastoma. You and your child’s cancer care team will work together to decide treatment. Many factors will be considered, such as your child’s overall health and whether the tumor is newly diagnosed or has come back.
Children who have medulloblastoma, other CNS embryonal tumors, and pineoblastoma should have their treatment planned by a team of health care providers who are experts in treating brain tumors in children.
A pediatric oncologist, a doctor who specializes in treating children with cancer, oversees treatment of medulloblastoma, other CNS embryonal tumors, and pineoblastoma. The pediatric oncologist works with other pediatric health care providers who are experts in treating children with brain tumors and who specialize in certain areas of medicine. Other specialists may include:
Your child’s treatment plan will include information about the cancer, the goals of treatment, treatment options, and the possible side effects. It will be helpful to talk with your child’s cancer care team before treatment begins about what to expect. For help every step of the way, see our downloadable booklet, Children with Cancer: A Guide for Parents.
The following types of treatment may be used:
Surgery
Surgery is used to diagnose and treat childhood medulloblastoma, other CNS embryonal tumors, and pineoblastoma as described in the General Information section of this summary.
After the doctor removes all the cancer that can be seen at the time of the surgery, some patients may be given chemotherapy, radiation therapy, or both to kill any cancer cells that are left. Treatment given after the surgery, to lower the risk that the cancer will come back, is called adjuvant therapy.
Radiation therapy
Radiation therapy uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. Medulloblastoma, other CNS embryonal tumors, or pineoblastoma in children may be treated with external beam radiation therapy. External beam radiation therapy uses a machine outside the body to send radiation toward the area of the body with cancer.
Certain ways of giving external radiation therapy can help keep radiation from damaging nearby healthy tissue. These types of radiation therapy include:
Conformal radiation therapy uses a computer to make a 3-dimensional (3-D) picture of the tumor and shapes the radiation beams to fit the tumor. This allows a high dose of radiation to reach the tumor and causes less damage to nearby healthy tissue.
Stereotactic radiation therapy uses a machine that aims radiation directly at the tumor, causing less damage to nearby healthy tissue. The total dose of radiation is divided into several smaller doses given over several days. A rigid head frame is attached to the skull to keep the head still during this radiation treatment. This procedure is also called stereotactic radiosurgery and stereotaxic radiation therapy.
Because radiation therapy can affect growth and brain development in young children, especially children who are 3 years or younger, chemotherapy may be given to delay or reduce the need for radiation therapy.
Radiation therapy to the brain can also affect growth and development in children older than 3 years. For this reason, clinical trials are studying new ways of giving radiation that may have fewer side effects than standard methods.
Chemotherapy
Chemotherapy (also called chemo) uses drugs to stop the growth of cancer cells, either by killing the cells or by stopping them from dividing. Chemotherapy may be given alone or with other types of treatment, such as radiation therapy.
To treat medulloblastoma, other CNS embryonal tumors, and pineoblastoma, chemotherapy is taken by mouth or injected into a vein. When given this way, the drugs enter the bloodstream and can reach cancer cells throughout the body. Chemotherapy that may be used alone or in combination includes:
High-dose chemotherapy with autologous stem cell rescue
High doses of chemotherapy are given to kill cancer cells. This cancer treatment destroys healthy cells, including blood-forming cells. Stem cell transplant is a treatment to replace the blood-forming cells. Stem cells (immature blood cells) are removed from the blood or bone marrow of the patient and are frozen and stored. After the patient completes chemotherapy, the stored stem cells are thawed and given back to the patient through an infusion. These reinfused stem cells grow into (and restore) the body’s blood cells.
Targeted therapy
Targeted therapy uses drugs or other substances to block the action of specific enzymes, proteins, or other molecules involved in the growth and spread of cancer cells.
Vismodegib may be used to treat recurrent medulloblastoma in children who have finished growing.
Targeted therapy is also being studied for the treatment of childhood medulloblastoma and other CNS embryonal tumors that have recurred (come back) after treatment.
New types of treatment are being tested in clinical trials.
A treatment clinical trial is a research study meant to help improve current treatments or obtain information on new treatments for patients with cancer. For some patients, taking part in a clinical trial may be the best treatment choice.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. Clinical trials supported by other organizations can be found on the ClinicalTrials.gov website.
Learn more at Clinical Trials Information for Patients and Caregivers. Because cancer in children is rare, taking part in a clinical trial should be considered. Some clinical trials are open only to patients who have not started treatment.
Chemotherapy with or without radiation therapy to the area where the tumor was removed.
Children older than 3 years with average-risk medulloblastoma
Treatment of newly diagnosed average-risk medulloblastoma in children older than 3 years includes:
Surgery to remove as much of the tumor as possible. This is followed by radiation therapy to the brain and spinal cord. Chemotherapy may also be given during and after radiation therapy.
Surgery to remove the tumor, radiation therapy, and high-dose chemotherapy with stem cell rescue.
Children older than 3 years with high-risk medulloblastoma
Treatment of newly diagnosed high-risk medulloblastoma in children older than 3 years includes:
Surgery to remove as much of the tumor as possible. This is followed by a larger dose of radiation therapy to the brain and spinal cord than the dose given for average-risk medulloblastoma. Chemotherapy is also given during and after radiation therapy.
Surgery to remove the tumor, radiation therapy, and high-dose chemotherapy with stem cell rescue.
Treatment of Other CNS Embryonal (nonmedulloblastoma) Tumors in Children
Children aged 3 years and younger with nonmedulloblastoma, nonmedulloepithelioma embryonal tumors
Treatment of newly diagnosed nonmedulloblastoma, nonmedulloepithelioma embryonal tumors in children 3 years or younger includes:
Surgery to remove as much of the tumor as possible, followed by chemotherapy.
Children older than 3 years with nonmedulloblastoma, nonmedulloepithelioma embryonal tumors
Treatment of newly diagnosed nonmedulloblastoma, nonmedulloepithelioma embryonal tumors in children older than 3 years includes:
Surgery to remove as much of the tumor as possible. This is followed by radiation therapy to the brain and spinal cord. Chemotherapy is also given during and after radiation therapy.
Children with embryonal tumors with multilayered rosettes or medulloepithelioma
Treatment of newly diagnosed embryonal tumor with multilayered rosettes (ETMR) or medulloepithelioma may include:
Surgery to remove as much of the tumor as possible. This is followed by chemotherapy. Radiation therapy may also be given.
Treatment of newly diagnosed CNS neuroblastoma may include:
Surgery to remove as much of the tumor as possible. This is followed by radiation therapy to the brain and spinal cord. Chemotherapy may also be given.
Biopsy to diagnose medulloblastoma and other CNS embryonal tumors. Surgery to remove as much of the tumor as possible may be done.
For children who previously received radiation therapy and chemotherapy, treatment may include repeat radiation at the site where the cancer started and where the tumor has spread. Stereotactic radiation therapy and/or chemotherapy may also be used.
For infants and young children who previously received chemotherapy only and have a local recurrence, treatment may be chemotherapy with radiation therapy to the tumor and the area close to it. Surgery to remove the tumor may also be done.
For patients who previously received radiation therapy, high-dose chemotherapy and stem cell rescue may be used. It is not known whether this treatment improves survival.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Side Effects
Key Points
The tumor and the treatment may cause symptoms that continue after treatment ends.
The tumor and the treatment may cause symptoms that continue after treatment ends.
Signs or symptoms caused by the tumor may begin before the cancer is diagnosed and continue for months or years. It is important to talk with your child’s doctors about signs or symptoms caused by the tumor that may continue after treatment.
Cancer treatments can cause side effects. Which side effects your child might have depends on the type of treatment they receive, the dose, and how their body reacts. Talk with your child’s treatment team about which side effects to look for and ways to manage them.
Problems from cancer treatment that begin 6 months or later after treatment and continue for months or years are called late effects. Late effects of cancer treatment may include:
Children diagnosed with medulloblastoma may have certain problems after surgery or radiation therapy, such as changes in the ability to think, learn, and pay attention. Also, cerebellar mutism syndrome may occur after surgery. Signs of this syndrome include:
delayed ability to speak
trouble swallowing and eating
loss of balance, trouble walking, and worsening handwriting
loss of muscle tone
mood swings and changes in personality
Some late effects may be treated or controlled. It is important to talk with your child’s doctors about the effects cancer treatment can have on your child and the types of symptoms to expect after cancer treatment has ended. Learn more about Late Effects of Treatment for Childhood Cancer.
Follow-Up Care
Some of the tests that were done to diagnose the cancer or to find out the stage of the cancer may be repeated to see how well the treatment is working. Decisions about whether to continue, change, or stop treatment may be based on the results of these tests. This is sometimes called re-staging. Learn more about these tests in the General Information section.
Some of the imaging tests will continue to be done from time to time after treatment has ended. The results of these tests can show if your child’s condition has changed or if the brain tumor has recurred (come back). If the imaging tests show abnormal tissue in the brain, a biopsy may also be done to find out if the tissue is made up of dead tumor cells or if new cancer cells are growing. These tests are sometimes called follow-up tests or check-ups.
Coping With Cancer
When a child has cancer, every member of the family needs support. Taking care of yourself during this difficult time is also important. Reach out to your child’s treatment team and to people in your family and community for support. To learn more, see Support for Families: Childhood Cancer and the booklet Children with Cancer: A Guide for Parents.
Related Resources
For more information about childhood medulloblastoma and other central nervous system embryonal tumor, see:
Physician Data Query (PDQ) is the National Cancer Institute’s (NCI’s) comprehensive cancer information database. The PDQ database contains summaries of the latest published information on cancer prevention, detection, genetics, treatment, supportive care, and complementary and alternative medicine. Most summaries come in two versions. The health professional versions have detailed information written in technical language. The patient versions are written in easy-to-understand, nontechnical language. Both versions have cancer information that is accurate and up to date and most versions are also available in Spanish.
PDQ is a service of the NCI. The NCI is part of the National Institutes of Health (NIH). NIH is the federal government’s center of biomedical research. The PDQ summaries are based on an independent review of the medical literature. They are not policy statements of the NCI or the NIH.
Purpose of This Summary
This PDQ cancer information summary has current information about the treatment of childhood medulloblastoma and other central nervous system embryonal tumors. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.
Reviewers and Updates
Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.
The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Pediatric Treatment Editorial Board.
Clinical Trial Information
A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).
Permission to Use This Summary
PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”
The best way to cite this PDQ summary is:
PDQ® Pediatric Treatment Editorial Board. PDQ Childhood Medulloblastoma and Other Central Nervous System Embryonal Tumors Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/brain/patient/child-cns-embryonal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389401]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use in the PDQ summaries only. If you want to use an image from a PDQ summary and you are not using the whole summary, you must get permission from the owner. It cannot be given by the National Cancer Institute. Information about using the images in this summary, along with many other images related to cancer can be found in Visuals Online. Visuals Online is a collection of more than 3,000 scientific images.
Disclaimer
The information in these summaries should not be used to make decisions about insurance reimbursement. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s E-mail Us.
An adult central nervous system (CNS) tumor is a disease in which abnormal cells form in the tissues of the brain and/or spinal cord.
A tumor that starts in another part of the body and spreads to the brain is called a metastatic brain tumor.
The brain controls many important body functions.
The spinal cord connects the brain to nerves in most parts of the body.
There are different types of brain and spinal cord tumors.
Astrocytic Tumors
Oligodendroglial Tumors
Mixed Gliomas
Ependymal Tumors
Medulloblastomas
Pineal Parenchymal Tumors
Meningeal Tumors
Germ Cell Tumors
Craniopharyngioma (Grade I)
Having certain genetic syndromes may increase the risk of a CNS tumor.
The cause of most adult brain and spinal cord tumors is not known.
The signs and symptoms of adult brain and spinal cord tumors are not the same in every person.
Tests that examine the brain and spinal cord are used to diagnose adult brain and spinal cord tumors.
A biopsy is also used to diagnose a brain tumor.
Sometimes a biopsy or surgery cannot be done.
Certain factors affect prognosis (chance of recovery) and treatment options.
An adult central nervous system (CNS) tumor is a disease in which abnormal cells form in the tissues of the brain and/or spinal cord.
There are many types of brain and spinal cordtumors. The tumors are formed by the abnormal growth of cells and may begin in different parts of the brain or spinal cord. Together, the brain and spinal cord make up the central nervous system (CNS).
Benign brain and spinal cord tumors grow and press on nearby areas of the brain. They rarely spread into other tissues and may recur (come back).
Malignant brain and spinal cord tumors are likely to grow quickly and spread into other brain tissue.
When a tumor grows into or presses on an area of the brain, it may stop that part of the brain from working the way it should. Both benign and malignant brain tumors cause signs and symptoms and need treatment.
Brain and spinal cord tumors can occur in both adults and children. However, treatment for children may be different than treatment for adults.
A tumor that starts in another part of the body and spreads to the brain is called a metastatic brain tumor.
Tumors that start in the brain are called primary brain tumors. Primary brain tumors may spread to other parts of the brain or to the spine. They rarely spread to other parts of the body.
Often, tumors found in the brain have started somewhere else in the body and spread to one or more parts of the brain. These are called metastatic brain tumors (or brain metastases). Metastatic brain tumors are more common than primary brain tumors. Up to half of metastatic brain tumors are from lung cancer.
The cerebrum is the largest part of the brain. It is at the top of the head. The cerebrum controls thinking, learning, problem solving, emotions, speech, reading, writing, and voluntary movement.
The cerebellum is in the lower back of the brain (near the middle of the back of the head). It controls movement, balance, and posture.
The brain stem connects the brain to the spinal cord. It is in the lowest part of the brain (just above the back of the neck). The brain stem controls breathing, heart rate, and the nerves and muscles used to see, hear, walk, talk, and eat.
EnlargeAnatomy of the brain showing the cerebrum, ventricles (with cerebrospinal fluid shown in blue), cerebellum, brain stem (pons and medulla), and other parts of the brain.
The spinal cord connects the brain to nerves in most parts of the body.
The spinal cord is a column of nerve tissue that runs from the brain stem down the center of the back. It is covered by three thin layers of tissue called membranes. These membranes are surrounded by the vertebrae (back bones). Spinal cord nerves carry messages between the brain and the rest of the body, such as a message from the brain to cause muscles to move or a message from the skin to the brain to feel touch.
There are different types of brain and spinal cord tumors.
Brain and spinal cord tumors are named based on the type of cell they formed in and where the tumor first formed in the CNS. The grade of a tumor may be used to tell the difference between slow-growing and fast-growing types of the tumor. The World Health Organization (WHO) tumor grades are based on how abnormal the cancer cells look under a microscope and how quickly the tumor is likely to grow and spread.
WHO Tumor Grading System
Grade I (low-grade) — The tumor cells look more like normal cells under a microscope and grow and spread more slowly than grade II, III, and IV tumor cells. They rarely spread into nearby tissues. Grade I brain tumors may be completely removed by surgery.
Grade II — The tumor cells grow and spread more slowly than grade III and IV tumor cells. They may spread into nearby tissue and may recur (come back). Some tumors may become a higher-grade tumor.
Grade III — The tumor cells look very different from normal cells under a microscope and grow more quickly than grade I and II tumor cells. They are likely to spread into nearby tissue.
Grade IV (high-grade) — The tumor cells do not look like normal cells under a microscope and grow and spread very quickly. There may be areas of dead cells in the tumor. Grade IV tumors usually cannot be completely removed by surgery.
The following types of primary tumors can form in the brain or spinal cord:
Astrocytic Tumors
An astrocytic tumor begins in star-shaped brain cells called astrocytes, which help keep nerve cells healthy. An astrocyte is a type of glial cell. Glial cells sometimes form tumors called gliomas. Astrocytic tumors include the following:
Brain stem glioma (usually high grade): A brain stem glioma forms in the brain stem, which is the part of the brain connected to the spinal cord. It is often a high-grade tumor, which spreads widely through the brain stem. Brain stem gliomas are rare in adults.
Pineal astrocytic tumor (any grade): A pineal astrocytic tumor forms in tissue around the pineal gland and may be any grade. The pineal gland is a tiny organ in the brain that makes melatonin, a hormone that helps control the sleeping and waking cycle.
Pilocytic astrocytoma (grade I): A pilocyticastrocytoma grows slowly in the brain or spinal cord. It may be in the form of a cyst and rarely spreads into nearby tissues.
Diffuse astrocytoma (grade II): A diffuse astrocytoma grows slowly, but often spreads into nearby tissues. The tumor cells look something like normal cells. It is also called a low-grade diffuse astrocytoma.
Anaplastic astrocytoma (grade III): An anaplastic astrocytoma grows quickly and spreads into nearby tissues. The tumor cells look different from normal cells. An anaplastic astrocytoma is also called a malignant astrocytoma or high-grade astrocytoma.
Glioblastoma (grade IV): A glioblastoma grows and spreads very quickly. The tumor cells look very different from normal cells. It is also called glioblastoma multiforme.
Oligodendroglial Tumors
An oligodendroglial tumor begins in brain cells called oligodendrocytes, which help keep nerve cells healthy. An oligodendrocyte is a type of glial cell. Oligodendrocytes sometimes form tumors called oligodendrogliomas. Grades of oligodendroglial tumors include the following:
Oligodendroglioma (grade II): An oligodendroglioma grows slowly, but often spreads into nearby tissues. The tumor cells look something like normal cells.
Anaplastic oligodendroglioma (grade III): An anaplastic oligodendroglioma grows quickly and spreads into nearby tissues. The tumor cells look different from normal cells.
Mixed Gliomas
A mixed glioma is a brain tumor that has two types of tumor cells in it — oligodendrocytes and astrocytes. This type of mixed tumor is called an oligoastrocytoma.
Oligoastrocytoma (grade II): An oligoastrocytoma is a slow-growing tumor. The tumor cells look something like normal cells.
Anaplastic oligoastrocytoma (grade III): An anaplastic oligoastrocytoma grows quickly and spreads into nearby tissues. The tumor cells look different from normal cells. This type of tumor has a worse prognosis than oligoastrocytoma (grade II).
Ependymal Tumors
An ependymal tumor usually begins in cells that line the fluid-filled spaces in the brain and around the spinal cord. An ependymal tumor may also be called an ependymoma. Grades of ependymomas include the following:
Ependymoma (grade I or II): A grade I or II ependymoma grows slowly and has cells that look something like normal cells. There are two types of grade I ependymoma — myxopapillary ependymoma and subependymoma. A grade II ependymoma grows in a ventricle (fluid-filled space in the brain) and its connecting paths or in the spinal cord.
Anaplastic ependymoma (grade III): An anaplastic ependymoma grows quickly and spreads into nearby tissues. The tumor cells look different from normal cells. This type of tumor usually has a worse prognosis than a grade I or II ependymoma.
A pineal parenchymal tumor forms in parenchymal cells or pineocytes, which are the cells that make up most of the pineal gland. These tumors are different from pineal astrocytic tumors. Grades of pineal parenchymal tumors include the following:
Pineocytoma (grade II): A pineocytoma is a slow-growing pineal tumor.
Pineoblastoma (grade IV): A pineoblastoma is a rare tumor that is very likely to spread.
A meningeal tumor, also called a meningioma, forms in the meninges (thin layers of tissue that cover the brain and spinal cord). It can form from different types of brain or spinal cord cells. Meningiomas are most common in adults. Types of meningeal tumors include the following:
Meningioma (grade I): A grade I meningioma is the most common type of meningeal tumor. A grade I meningioma is a slow-growing tumor. It forms most often in the dura mater. A grade I meningioma may be completely removed by surgery.
Meningioma (grade II and III): This is a rare meningeal tumor. It grows quickly and is likely to spread within the brain and spinal cord. The prognosis is worse than a grade I meningioma because the tumor usually cannot be completely removed by surgery.
A hemangiopericytoma is not a meningeal tumor but is treated like a grade II or III meningioma. A hemangiopericytoma usually forms in the dura mater. The prognosis is worse than a grade I meningioma because the tumor usually cannot be completely removed by surgery.
Germ Cell Tumors
A germ cell tumor forms in germ cells, which are the cells that develop into sperm in men or ova (eggs) in women. There are different types of germ cell tumors. These include germinomas, teratomas, embryonal yolk sac carcinomas, and choriocarcinomas. Germ cell tumors can be either benign or malignant.
A craniopharyngioma is a rare tumor that usually forms in the center of the brain just above the pituitary gland (a pea-sized organ at the bottom of the brain that controls other glands). Craniopharyngiomas can form from different types of brain or spinal cord cells.
Having certain genetic syndromes may increase the risk of a CNS tumor.
Anything that increases a person’s chance of getting a disease is called a risk factor. Not every person with one or more of these risk factors will develop a brain or spinal cord tumor, and they can develop in people who don’t have any known risk factors. Talk with your doctor if you think you may be at risk. There are few known risk factors for brain tumors. The following conditions may increase the risk of certain types of brain tumors:
Being exposed to vinyl chloride may increase the risk of glioma.
The cause of most adult brain and spinal cord tumors is not known.
The signs and symptoms of adult brain and spinal cord tumors are not the same in every person.
Signs and symptoms depend on the following:
Where the tumor forms in the brain or spinal cord.
What the affected part of the brain controls.
The size of the tumor.
These and other signs and symptoms may be caused by CNS tumors or by other conditions, including cancer that has spread to the brain. Check with your doctor if you have any of the following:
Brain Tumor Symptoms
Morning headache or headache that goes away after vomiting.
Neurological exam: A series of questions and tests to check the brain, spinal cord, and nerve function. The exam checks a person’s mental status, coordination, and ability to walk normally, and how well the muscles, senses, and reflexes work. This may also be called a neuro exam or a neurologic exam.
Visual field exam: An exam to check a person’s field of vision (the total area in which objects can be seen). This test measures both central vision (how much a person can see when looking straight ahead) and peripheral vision (how much a person can see in all other directions while staring straight ahead). Any loss of vision may be a sign of a tumor that has damaged or pressed on the parts of the brain that affect eyesight.
Tumor marker test: A procedure in which a sample of blood, urine, or tissue is checked to measure the amounts of certain substances made by organs, tissues, or tumor cells in the body. Certain substances are linked to specific types of cancer when found in increased levels in the body. These are called tumor markers. This test may be done to diagnose a germ cell tumor.
Gene testing: A laboratory test in which cells or tissue are analyzed to look for changes in genes or chromosomes. These changes may be a sign that a person has or is at risk of having a specific disease or condition.
CT scan (CAT scan): A procedure that makes a series of detailed pictures of areas inside the body, taken from different angles. The pictures are made by a computer linked to an x-ray machine. A dye may be injected into a vein or swallowed to help the organs or tissues show up more clearly. This procedure is also called computed tomography, computerized tomography, or computerized axial tomography. EnlargeComputed tomography (CT) scan of the brain. The patient lies on a table that slides through the CT scanner, which takes x-ray pictures of the brain.
MRI (magnetic resonance imaging) with gadolinium: A procedure that uses a magnet, radio waves, and a computer to make a series of detailed pictures of the brain and spinal cord. A substance called gadolinium is injected into a vein. The gadolinium collects around the cancer cells so they show up brighter in the picture. This procedure is also called nuclear magnetic resonance imaging (NMRI). MRI is often used to diagnose tumors in the spinal cord. Sometimes a procedure called magnetic resonance spectroscopy (MRS) is done during the MRI scan. An MRS is used to diagnose tumors, based on their chemical make-up.
SPECT scan (single photon emission computed tomography scan): A procedure to find malignant tumor cells in the brain. A small amount of a radioactive substance is injected into a vein or inhaled through the nose. As the substance travels through the blood, a camera rotates around the head and takes pictures of the brain. A computer uses the pictures to make a 3-dimensional (3-D) image of the brain. There will be increased blood flow and more activity in areas where cancer cells are growing. These areas will show up brighter in the picture.
PET scan (positron emission tomography scan): A procedure to find malignant tumor cells in the body. A small amount of radioactive glucose (sugar) is injected into a vein. The PET scanner rotates around the body and makes a picture of where glucose is being used in the brain. Malignant tumor cells show up brighter in the picture because they are more active and take up more glucose than normal cells do. PET is used to tell the difference between a primary tumor and a tumor that has spread to the brain from somewhere else in the body. EnlargePET (positron emission tomography) scan. The patient lies on a table that slides through the PET machine. The head rest and white strap help the patient lie still. A small amount of radioactive glucose (sugar) is injected into the patient’s vein, and a scanner makes a picture of where the glucose is being used in the body. Cancer cells show up brighter in the picture because they take up more glucose than normal cells do.
A biopsy is also used to diagnose a brain tumor.
If imaging tests show there may be a brain tumor, a biopsy is usually done. One of the following types of biopsies may be used:
Stereotactic biopsy: When imaging tests show there may be a tumor deep in the brain in a hard to reach place, a stereotactic brain biopsy may be done. This kind of biopsy uses a computer and a 3-dimensional (3-D) scanning device to find the tumor and guide the needle used to remove the tissue. A small incision is made in the scalp, and a small hole is drilled through the skull. A biopsy needle is inserted through the hole to remove cells or tissues so they can be viewed under a microscope by a pathologist to check for signs of cancer.
Open biopsy: When imaging tests show that there may be a tumor that can be removed by surgery, an open biopsy may be done. A part of the skull is removed in an operation called a craniotomy. A sample of brain tissue is removed and viewed under a microscope by a pathologist. If cancer cells are found, some or all of the tumor may be removed during the same surgery. Tests are done before surgery to find the areas around the tumor that are important for normal brain function. There are also ways to test brain function during surgery. The doctor will use the results of these tests to remove as much of the tumor as possible with the least damage to normal tissue in the brain. EnlargeCraniotomy: An opening is made in the skull and a piece of the skull is removed to show part of the brain.
The pathologist checks the biopsy sample to find out the type and grade of the brain tumor. The grade of the tumor is based on how the tumor cells look under a microscope, and how quickly the tumor is likely to grow and spread.
The following tests may be done on the tumor tissue that is removed:
Immunohistochemistry: A laboratory test that uses antibodies to check for certain antigens (markers) in a sample of a patient’s tissue. The antibodies are usually linked to an enzyme or a fluorescent dye. After the antibodies bind to a specific antigen in the tissue sample, the enzyme or dye is activated, and the antigen can then be seen under a microscope. This type of test is used to help diagnose cancer and to help tell one type of cancer from another type of cancer.
Light and electron microscopy: A laboratory test in which cells in a sample of tissue are viewed under regular and high-powered microscopes to look for certain changes in the cells.
Cytogeneticanalysis: A laboratory test in which the chromosomes of cells in a sample of brain tissue are counted and checked for any changes, such as broken, missing, rearranged, or extra chromosomes. Changes in certain chromosomes may be a sign of cancer. Cytogenetic analysis is used to help diagnose cancer, plan treatment, or find out how well treatment is working.
Sometimes a biopsy or surgery cannot be done.
For some tumors, a biopsy or surgery cannot be done safely because of where the tumor formed in the brain or spinal cord. These tumors are diagnosed and treated based on the results of imaging tests and other procedures.
Sometimes the results of imaging tests and other procedures show that the tumor is very likely to be benign, and a biopsy is not done.
Certain factors affect prognosis (chance of recovery) and treatment options.
The prognosis and treatment options for primary brain and spinal cord tumors depend on the following:
The type and grade of the tumor.
Where the tumor is in the brain or spinal cord.
Whether the tumor can be removed by surgery.
Whether cancer cells remain after surgery.
Whether there are certain changes in the chromosomes.
Whether the cancer has just been diagnosed or has recurred (come back).
The patient’s general health.
The prognosis and treatment options for metastatic brain and spinal cord tumors depend on the following:
Whether there are more than two tumors in the brain or spinal cord.
Where the tumor is in the brain or spinal cord.
How well the tumor responds to treatment.
Whether the primary tumor continues to grow or spread.
Stages of Adult Central Nervous System Tumors
Key Points
There is no standard staging system for adult brain and spinal cord tumors.
Imaging tests may be repeated after surgery to help plan more treatment.
Central nervous system (CNS) tumors often recur, sometimes many years after treatment.
There is no standard staging system for adult brain and spinal cord tumors.
The process used to find out if cancer has spread to other areas of the brain or to other parts of the body is called staging. Brain tumors that begin in the brain rarely spread to other parts of the body. There is no standard staging system for brain and spinal cord tumors.
Treatment of primary brain and spinal cord tumors is based on the following:
The type of cell in which the tumor began.
Where the tumor formed in the brain or spinal cord.
Treatment of tumors that have spread to the brain from other parts of the body is based on the number of tumors in the brain.
Imaging tests may be repeated after surgery to help plan more treatment.
Some of the tests and procedures used to diagnose a brain or spinal cord tumor may be repeated after treatment to find out how much tumor is left.
Central nervous system (CNS) tumors often recur, sometimes many years after treatment.
A recurrent CNS tumor is a tumor that has recurred (come back) after it has been treated. The tumor may recur at the same place as the first tumor or in other parts of the CNS.
Treatment Option Overview
Key Points
There are different types of treatment for patients with adult brain and spinal cord tumors.
The following types of treatment are used:
Active surveillance
Surgery
Radiation therapy
Chemotherapy
Targeted therapy
Supportive care is given to lessen the problems caused by the disease or its treatment.
New types of treatment are being tested in clinical trials.
Proton beam radiation therapy
Immunotherapy
Treatment for adult central nervous system tumors may cause side effects.
Patients may want to think about taking part in a clinical trial.
Patients can enter clinical trials before, during, or after starting their cancer treatment.
Follow-up tests may be needed.
There are different types of treatment for patients with adult brain and spinal cord tumors.
Different types of treatment are available for patients with adult brain and spinal cord tumors. Some treatments are standard (the currently used treatment), and some are being tested in clinical trials. A treatment clinical trial is a research study meant to help improve current treatments or obtain information on new treatments for patients with cancer. When clinical trials show that a new treatment is better than the standard treatment, the new treatment may become the standard treatment. Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
The following types of treatment are used:
Active surveillance
Active surveillance is closely watching a patient’s condition but not giving any treatment unless there are changes in test results that show the condition is getting worse. Active surveillance may be used to avoid or delay the need for treatments such as radiation therapy or surgery, which can cause side effects or other problems. During active surveillance, certain exams and tests are done on a regular schedule. Active surveillance may be used for very slow-growing tumors that do not cause symptoms.
Surgery
Surgery may be used to diagnose and treat adult brain and spinal cord tumors. Removing tumor tissue helps decrease pressure of the tumor on nearby parts of the brain. See the General Information section of this summary.
After the doctor removes all the cancer that can be seen at the time of the surgery, some patients may be given chemotherapy or radiation therapy after surgery to kill any cancer cells that are left. Treatment given after the surgery, to lower the risk that the cancer will come back, is called adjuvant therapy.
Radiation therapy
Radiation therapy is a cancer treatment that uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. External radiation therapy uses a machine outside the body to send radiation toward the area of the body with cancer.
EnlargeExternal-beam radiation therapy of the brain. A machine is used to aim high-energy radiation. The machine can rotate around the patient, delivering radiation from many different angles. A mesh mask helps keep the patient’s head from moving during treatment. Small ink marks are put on the mask. The ink marks are used to line up the radiation machine in the same position before each treatment.
Certain ways of giving external radiation therapy can help keep radiation from damaging nearby healthy tissue. These types of radiation therapy include the following:
Conformal radiation therapy: Conformal radiation therapy uses a computer to make a 3-dimensional (3-D) picture of the tumor and shapes the radiation beams to fit the tumor.
Intensity-modulated radiation therapy (IMRT): IMRT is a type of 3-dimensional (3-D) radiation therapy that uses a computer to make pictures of the size and shape of the tumor. Thin beams of radiation of different intensities (strengths) are aimed at the tumor from many angles.
Stereotactic radiosurgery: Stereotactic radiosurgery uses a rigid head frame that is attached to the skull to keep the head still during the radiation treatment. A machine aims a single large dose of radiation directly at the tumor. This procedure does not involve surgery. It is also called stereotaxic radiosurgery, radiosurgery, and radiation surgery.
Chemotherapy
Chemotherapy is a cancer treatment that uses drugs to stop the growth of cancer cells, either by killing the cells or by stopping them from dividing. When chemotherapy is taken by mouth or injected into a vein or muscle, the drugs enter the bloodstream and can reach cancer cells throughout the body (systemic chemotherapy). Although most cannot, some chemotherapy drugs can cross the blood-brain barrier and reach tumor cells in the brain. Chemotherapy that is placed directly into the cerebrospinal fluid is called intrathecal chemotherapy. When chemotherapy is inserted in an organ, such as the brain, or a body cavity, the drugs mainly affect cancer cells in those areas (regional chemotherapy).
To treat brain tumors, a wafer that dissolves may be used to deliver a chemotherapy drug directly to the brain tumor site after the tumor has been removed by surgery. The way the chemotherapy is given depends on the type and grade of tumor and where it is in the brain.
Targeted therapy is a type of treatment that uses drugs or other substances to identify and attack specific cancer cells.
Monoclonal antibodytherapy: Monoclonal antibodies are immune systemproteins made in the laboratory to treat many diseases, including cancer. As a cancer treatment, these antibodies can attach to a specific target on cancer cells or other cells that may help cancer cells grow. The antibodies are able to then kill the cancer cells, block their growth, or keep them from spreading. Monoclonal antibodies are given by infusion. They may be used alone or to carry drugs, toxins, or radioactive material directly to cancer cells.
How do monoclonal antibodies work to treat cancer? This video shows how monoclonal antibodies, such as trastuzumab, pembrolizumab, and rituximab, block molecules cancer cells need to grow, flag cancer cells for destruction by the body’s immune system, or deliver harmful substances to cancer cells.
Other types of targeted therapies are being studied for adult brain tumors, including tyrosine kinase inhibitors and new VEGF inhibitors.
Supportive care is given to lessen the problems caused by the disease or its treatment.
This therapy controls problems or side effects caused by the disease or its treatment and improves quality of life. For brain tumors, supportive care includes drugs to control seizures and fluid buildup or swelling in the brain.
New types of treatment are being tested in clinical trials.
This summary section refers to new treatments being studied in clinical trials, but it may not mention every new treatment being studied. Information about clinical trials is available from the NCI website.
Proton beam radiation therapy
Proton beam radiation therapy is a type of high-energy, external radiation therapy that uses streams of protons (tiny particles with a positive charge) to kill tumor cells. This type of treatment can lower the amount of radiation damage to healthy tissue near a tumor. It is used to treat cancers of the head, neck, and spine and organs such as the brain, eye, lung, and prostate. Proton beam radiation is different from x-ray radiation.
Immunotherapy
Immunotherapy is a treatment that uses the patient’s immune system to fight cancer. Substances made by the body or made in a laboratory are used to boost, direct, or restore the body’s natural defenses against cancer.
Immunotherapy is being studied for the treatment of some types of brain tumors. Treatments may include the following:
Patients may want to think about taking part in a clinical trial.
For some patients, taking part in a clinical trial may be the best treatment choice. Clinical trials are part of the cancer research process. Clinical trials are done to find out if new cancer treatments are safe and effective or better than the standard treatment.
Many of today’s standard treatments for cancer are based on earlier clinical trials. Patients who take part in a clinical trial may receive the standard treatment or be among the first to receive a new treatment.
Patients who take part in clinical trials also help improve the way cancer will be treated in the future. Even when clinical trials do not lead to effective new treatments, they often answer important questions and help move research forward.
Patients can enter clinical trials before, during, or after starting their cancer treatment.
Some clinical trials only include patients who have not yet received treatment. Other trials test treatments for patients whose cancer has not gotten better. There are also clinical trials that test new ways to stop cancer from recurring (coming back) or reduce the side effects of cancer treatment.
Clinical trials are taking place in many parts of the country. Information about clinical trials supported by NCI can be found on NCI’s clinical trials search webpage. Clinical trials supported by other organizations can be found on the ClinicalTrials.gov website.
Follow-up tests may be needed.
As you go through treatment, you will have follow-up tests or check-ups. Some tests that were done to diagnose or stage the cancer may be repeated to see how well the treatment is working. Decisions about whether to continue, change, or stop treatment may be based on the results of these tests.
Some of the tests will continue to be done from time to time after treatment has ended. The results of these tests can show if your condition has changed or if the cancer has recurred (come back).
The following tests and procedures may be used to check whether a brain tumor has come back after treatment:
SPECT scan (single photon emission computed tomography scan): A procedure to find malignant tumor cells in the brain. A small amount of a radioactive substance is injected into a vein or inhaled through the nose. As the substance travels through the blood, a camera rotates around the head and takes pictures of the brain. A computer uses the pictures to make a 3-dimensional (3-D) image of the brain. There will be increased blood flow and more activity in areas where cancer cells are growing. These areas will show up brighter in the picture.
PET scan (positron emission tomography scan): A procedure to find malignant tumor cells in the body. A small amount of radioactive glucose (sugar) is injected into a vein. The PET scanner rotates around the body and makes a picture of where glucose is being used in the brain. Malignant tumor cells show up brighter in the picture because they are more active and take up more glucose than normal cells do. EnlargePET (positron emission tomography) scan. The patient lies on a table that slides through the PET machine. The head rest and white strap help the patient lie still. A small amount of radioactive glucose (sugar) is injected into the patient’s vein, and a scanner makes a picture of where the glucose is being used in the body. Cancer cells show up brighter in the picture because they take up more glucose than normal cells do.
Treatment of Primary Adult Brain Tumor by Type of Tumor
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Pilocytic Astrocytomas
Treatment of pilocyticastrocytomas may include surgery to remove the tumor. Radiation therapy may also be given if some of the tumor remains after surgery.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Surgery followed by radiation therapy and chemotherapy.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Glioblastomas
Treatment of glioblastomas may include the following:
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Ependymal Tumors
Treatment of grade I and grade II ependymomas may include surgery to remove the tumor. Radiation therapy may also be given if some of the tumor remains after surgery.
Treatment of grade III anaplastic ependymoma may include surgery and radiation therapy.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
For pineoblastomas, surgery, radiation therapy, and chemotherapy.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Radiation therapy for tumors that cannot be removed by surgery.
Treatment of grade II and III meningiomas and hemangiopericytomas may include the following:
Surgery and radiation therapy.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Surgery to remove as much of the tumor as possible, followed by radiation therapy.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
To Learn More About Adult Central Nervous System Tumors
Physician Data Query (PDQ) is the National Cancer Institute’s (NCI’s) comprehensive cancer information database. The PDQ database contains summaries of the latest published information on cancer prevention, detection, genetics, treatment, supportive care, and complementary and alternative medicine. Most summaries come in two versions. The health professional versions have detailed information written in technical language. The patient versions are written in easy-to-understand, nontechnical language. Both versions have cancer information that is accurate and up to date and most versions are also available in Spanish.
PDQ is a service of the NCI. The NCI is part of the National Institutes of Health (NIH). NIH is the federal government’s center of biomedical research. The PDQ summaries are based on an independent review of the medical literature. They are not policy statements of the NCI or the NIH.
Purpose of This Summary
This PDQ cancer information summary has current information about the treatment of adult central nervous system tumors. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.
Reviewers and Updates
Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.
The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Adult Treatment Editorial Board.
Clinical Trial Information
A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).
Permission to Use This Summary
PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”
The best way to cite this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Adult Central Nervous System Tumors Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/brain/patient/adult-brain-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389458]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use in the PDQ summaries only. If you want to use an image from a PDQ summary and you are not using the whole summary, you must get permission from the owner. It cannot be given by the National Cancer Institute. Information about using the images in this summary, along with many other images related to cancer can be found in Visuals Online. Visuals Online is a collection of more than 3,000 scientific images.
Disclaimer
The information in these summaries should not be used to make decisions about insurance reimbursement. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s E-mail Us.
Brain tumors account for 85% to 90% of all primary central nervous system (CNS) tumors.[1] Estimated new cases and deaths from brain tumors and other nervous system tumors in the United States in 2025:[2]
New cases: 24,820.
Deaths: 18,330.
Data from the Surveillance, Epidemiology, and End Results (SEER) Program database for 2017 to 2021 indicated that the combined incidence of brain and other CNS tumors in the United States was 6.2 per 100,000 people per year. The mortality rate was 4.4 deaths per 100,000 people per year based on age-adjusted deaths from 2018 to 2022.[3] Worldwide, approximately 321,476 new cases of brain and other CNS tumors were diagnosed in the year 2022, with an estimated 248,305 deaths.[4]
In general, the incidence of primary CNS tumors is higher in White individuals than in Black individuals, and mortality is higher in men than in women.[3]
Primary brain tumors include the following in decreasing order of frequency:[1]
Anaplastic astrocytomas and glioblastomas (38% of primary brain tumors).
Meningiomas and other mesenchymal tumors (27% of primary brain tumors).
Pituitary tumors.
Schwannomas.
CNS lymphomas.
Oligodendrogliomas.
Ependymomas.
Low-grade astrocytomas.
Medulloblastomas.
Primary spinal tumors include the following in decreasing order of frequency:
Schwannomas, meningiomas, and ependymomas (79% of primary spinal tumors).
Sarcomas.
Astrocytomas.
Vascular tumors.
Chordomas.
Primary brain tumors rarely spread to other areas of the body, but they can spread to other parts of the brain and to the spinal axis.
Anatomy
EnlargeAnatomy of the inside of the brain. The supratentorium contains the cerebrum, ventricles (with cerebrospinal fluid shown in blue), choroid plexus, hypothalamus, pineal gland, pituitary gland, and optic nerve. The infratentorium contains the cerebellum and brain stem.
Risk Factors
Few definitive observations have been made about environmental or occupational causes of primary CNS tumors.[1]
The following potential risk factors have been considered:
Exposure to vinyl chloride may be a risk factor for glioma.
Epstein-Barr virus infection has been implicated in the etiology of primary CNS lymphoma.
Transplant recipients and patients with AIDS have a substantially increased risk of primary CNS lymphoma.[1,5] For more information, see Primary CNS Lymphoma Treatment.
The familial tumor syndromes and related chromosomal abnormalities that are associated with CNS neoplasms include the following:[6,7]
Neurofibromatosis type 1 (17q11).
Neurofibromatosis type 2 (22q12).
von Hippel-Lindau disease (3p25-26).
Tuberous sclerosis (9q34, 16p13).
Li-Fraumeni syndrome (17p13).
Turcot syndrome type 1 (3p21, 7p22).
Turcot syndrome type 2 (5q21).
Nevoid basal cell carcinoma syndrome (9q22.3).
Clinical Features
The clinical presentation of various brain tumors is best appreciated by considering the relationship of signs and symptoms to anatomy.[1]
General signs and symptoms include the following:
Headaches.
Seizures.
Visual changes.
Gastrointestinal symptoms such as loss of appetite, nausea, and vomiting.
Changes in personality, mood, mental capacity, and concentration.
Seizures are a presenting symptom in approximately 20% of patients with supratentorial brain tumors and may antedate the clinical diagnosis by months to years in patients with slow-growing tumors. Among all patients with brain tumors, 70% with primary parenchymal tumors and 40% with metastatic brain tumors develop seizures at some time during the clinical course.[8]
Diagnostic Evaluation
All brain tumors, whether primary, metastatic, malignant, or benign, must be differentiated from other space-occupying lesions that can have similar clinical presentations, such as abscesses, arteriovenous malformations, and infarctions.[9]
Imaging tests
Contrast-enhanced computed tomography (CT) and magnetic resonance imaging (MRI) have complementary roles in the diagnosis of CNS neoplasms.[1,9,10]
The speed of CT is desirable for evaluating clinically unstable patients. CT is superior for detecting calcifications, skull lesions, and hyperacute hemorrhages (bleeding less than 24 hours old) and helps direct differential diagnosis and immediate management.
MRI has superior soft-tissue resolution. MRI can better detect isodense lesions, tumor enhancements, and associated findings such as edema, all phases of hemorrhagic states (except hyperacute), and infarctions. High-quality MRI is the diagnostic study of choice in the evaluation of intramedullary and extramedullary spinal cord lesions.[1]
In posttherapy imaging, single-photon emission computed tomography (SPECT) and positron emission tomography (PET) may be useful in differentiating tumor recurrence from radiation necrosis.[9]
Biopsy
Biopsy confirmation to corroborate the suspected diagnosis of a primary brain tumor is critical, whether before surgery by needle biopsy or at the time of surgical resection. The exception is cases in which the clinical and radiological evidence clearly points to a benign tumor, which could potentially be managed with active surveillance without biopsy or treatment. For other cases, radiological patterns may be misleading, and a definitive biopsy is needed to rule out other causes of space-occupying lesions, such as metastatic cancer or infection.
CT- or MRI-guided stereotactic techniques can be used to place a needle safely and accurately into almost all locations in the brain.
Prognostic Factors
Several genetic alterations have emerged as powerful prognostic factors in diffuse glioma (astrocytoma, oligodendroglioma, mixed glioma, and glioblastoma), and these alterations may guide patient management. Specific alterations include the following:
DNA methylation of the MGMT gene promoter.
IDH1 or IDH2 variants.
Codeletion of chromosomes 1p and 19q.
Other prognostic factors that confer poor prognosis include the following:[11,12]
Age older than 40 years.
Progressive disease.
Tumor size larger than 5 cm.
Tumor crossing the midline.
Contrast enhancement on MRI.
World Health Organization performance status (≥1).
Neurological symptoms.
Less than a gross total resection.
In an exploratory analysis of 318 patients with low-grade glioma treated with either radiation therapy alone or temozolomide chemotherapy alone, a combination of these prognostic factors demonstrated the following:[11]
Longer progression-free survival (PFS) in patients with IDH variants without codeletion of 1p/19q when treated with radiation therapy (hazard ratio, 1.86; 95% confidence interval, 1.21–2.87; log-rank P = .0043).
No significant treatment-dependent differences in PFS for patients with IDH variants with codeletion of 1p/19q and IDH wild-type tumors.
Patients with wild-type IDH tumors had the worst prognosis independent of treatment type.
Patients with IDH variants with codeletion of 1p/19q had the best prognosis.
The O6-methylguanine-DNA methyltransferase (MGMT) promoter status in low-grade tumors was methylated in:
All IDH variants with codeletion of 1p/19q (45/45).
Most, but not all (86%, 62/72), of the IDH variants without codeletion of 1p/19q.
Fifty-six percent (5/9) of the IDH wild-type cases.
Mehta M, Vogelbaum MA, Chang S, et al.: Neoplasms of the central nervous system. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 1700-49.
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
National Cancer Institute: SEER Cancer Stat Facts: Brain and Other Nervous System Cancer. Bethesda, Md: National Cancer Institute. Available online. Last accessed January 24, 2025.
Bray F, Laversanne M, Sung H, et al.: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 74 (3): 229-263, 2024. [PUBMED Abstract]
Hutter A, Schwetye KE, Bierhals AJ, et al.: Brain neoplasms: epidemiology, diagnosis, and prospects for cost-effective imaging. Neuroimaging Clin N Am 13 (2): 237-50, x-xi, 2003. [PUBMED Abstract]
Ricci PE: Imaging of adult brain tumors. Neuroimaging Clin N Am 9 (4): 651-69, 1999. [PUBMED Abstract]
Baumert BG, Hegi ME, van den Bent MJ, et al.: Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 17 (11): 1521-1532, 2016. [PUBMED Abstract]
Reijneveld JC, Taphoorn MJ, Coens C, et al.: Health-related quality of life in patients with high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 17 (11): 1533-1542, 2016. [PUBMED Abstract]
World Health Organization (WHO) Classification of Adult Primary CNS Tumors
This classification is based on the World Health Organization (WHO) classification of central nervous system (CNS) tumors.[1] The WHO approach incorporates and interrelates morphology, cytogenetics, molecular genetics, and immunological markers in an attempt to construct a cellular classification that is universally applicable and prognostically valid. Earlier attempts to develop a TNM (tumor, node, metastasis)-based classification were dropped for the following reasons:[2]
Tumor size (T) is less relevant than are tumor histology and location.
Nodal status (N) does not apply because the brain and spinal cord have no lymphatics.
Metastatic spread (M) rarely applies because most patients with CNS neoplasms do not live long enough to develop metastatic disease.
The WHO grading of CNS tumors establishes a malignancy scale based on histological features of the tumor.[3] The histological grades are as follows:
WHO grade I includes lesions with low proliferative potential, a frequently discrete nature, and the possibility of cure following surgical resection alone.
WHO grade II includes lesions that are generally infiltrating and low in mitotic activity but recur more frequently than do grade I malignant tumors after local therapy. Some tumor types tend to progress to higher grades of malignancy.
WHO grade III includes lesions with histological evidence of malignancy, including nuclear atypia and increased mitotic activity. These lesions have anaplastic histology and infiltrative capacity. They are usually treated with aggressive adjuvant therapy.
WHO grade IV includes lesions that are mitotically active, necrosis prone, and generally associated with a rapid preoperative and postoperative progression and fatal outcomes. The lesions are usually treated with aggressive adjuvant therapy.
Table 1 lists the tumor types and grades.[4] Tumors limited to the peripheral nervous system are not included. Histopathology, grading methods, incidence, and what is known about etiology specific to each tumor type have been described in detail elsewhere.[4,5]
Table 1. WHO Grades of CNS Tumorsa
I
II
III
IV
aReprinted with permission from Louis, DN, Ohgaki H, Wiestler, OD, Cavenee, WK. World Health Organization Classification of Tumours of the Central Nervous System. IARC, Lyon, 2007.
Astrocytic tumors
Subependymal giant cell astrocytoma
X
Pilocytic astrocytoma
X
Pilomyxoid astrocytoma
X
Diffuse astrocytoma
X
Pleomorphic xanthoastrocytoma
X
Anaplastic astrocytoma
X
Glioblastoma
X
Giant cell glioblastoma
X
Gliosarcoma
X
Oligodendroglial tumors
Oligodendroglioma
X
Anaplastic oligodendroglioma
X
Oligoastrocytic tumors
Oligoastrocytoma
X
Anaplastic oligoastrocytoma
X
Ependymal tumors
Subependymoma
X
Myxopapillary ependymoma
X
Ependymoma
X
Anaplastic ependymoma
X
Choroid plexus tumors
Choroid plexus papilloma
X
Atypical choroid plexus papilloma
X
Choroid plexus carcinoma
X
Other neuroepithelial tumors
Angiocentric glioma
X
Chordoid glioma of the third ventricle
X
Neuronal and mixed neuronal-glial tumors
Gangliocytoma
X
Ganglioglioma
X
Anaplastic ganglioma
X
Desmoplastic infantile astrocytoma and ganglioglioma
X
Dysembryoplastic neuroepithelial tumor
X
Central neurocytoma
X
Extraventricular neurocytoma
X
Cerebellar liponeurocytoma
X
Paraganglioma of the spinal cord
X
Papillary glioneuronal tumor
X
Rosette-forming glioneural tumor of the fourth ventricle
X
Pineal tumors
Pineocytoma
X
Pineal parenchymal tumor of intermediate differentiation
X
X
Pineoblastoma
X
Papillary tumor of the pineal region
X
X
Embryonal tumors
Medulloblastoma
X
CNS primitive neuroectodermal tumor
X
Atypical teratoid/rhabdoid tumor
X
Tumors of the cranial and paraspinal nerves
Schwannoma
X
Neurofibroma
X
Perineurioma
X
X
X
Malignant peripheral nerve sheath tumor
X
X
X
Meningeal tumors
Meningioma
X
Atypical meningioma
X
Anaplastic/malignant meningioma
X
Hemangiopericytoma
X
Anaplastic hemangiopericytoma
X
Hemangioblastoma
X
Tumors of the sellar region
Craniopharyngioma
X
Granular cell tumor of the neurohypophysis
X
Pituicytoma
X
Spindle cell oncocytoma of the adenohypophysis
X
Genomic Alterations
Alterations in the BRAF, IDH1, and IDH2 genes, and genomic 1p/19q codeletion, appear to be hallmark aberrations in particular glioma subtypes. Assessment for the presence of these variants aids diagnosis and prognosis and, with regard to 1p/19q codeletion, predicts for response to chemotherapy.
In pilocytic astrocytomas (WHO grade I), tandem duplication at 7q34 leading to a KIAA1549::BRAF gene fusion is found in approximately 70% of pilocytic astrocytomas.[6–8] Activating single nucleotide variants in BRAF (V600E) are found in an additional 5% to 9% of these tumors. Overall, RAF alterations occur in approximately 80% of pilocytic astrocytomas.
BRAF V600E variants are observed (in about 60%) of other benign gliomas, including pleomorphic xanthoastrocytoma and ganglioglioma, while BRAF tandem duplications are not found in these variant glioma tumors.[9–11]
Most WHO grade II and III diffuse gliomas (astrocytomas, oligodendrogliomas, and oligoastrocytomas) and 5% to 10% of glioblastomas (WHO grade IV) harbor single nucleotide variants in the R132 position of IDH1 or, rarely, the analogous codon in IDH2 (R172).[12–16] The presence of an IDH1 or IDH2 variant is a strong prognostic factor. Patients with these tumor variants have significantly longer survival independent of WHO grade or histological subtype.
Deletion of chromosomes 1p and 19q occurs through a translocation event [17] and is common in oligodendrogliomas. 1p/19q codeletion is a powerful prognostic factor and may predict for response to chemotherapy. For more information, see the Anaplastic oligodendrogliomas treatment section.
These genetic alterations have potential diagnostic utility. Presence of the IDH1 and IDH2 variants may distinguish diffuse gliomas from other gliomas, which often have BRAF genetic alterations, and nonneoplastic reactive astrocytosis.[18] Most (90%) IDH variants in gliomas result in an R132H substitution, which can be detected with a highly sensitive and specific monoclonal antibody. A rapid immunohistochemical analysis using the variant-specific IDH1 antibody can aid diagnostic analysis.[19]
Other CNS tumors are associated with characteristic patterns of altered oncogenes, altered tumor suppressor genes, and chromosomal abnormalities. Familial tumor syndromes with defined chromosomal abnormalities are associated with gliomas.
References
Kleihues P, Cavenee WK, eds.: Pathology and Genetics of Tumours of the Nervous System. International Agency for Research on Cancer, 2000.
Brain and Spinal Cord. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 857–69.
Kleihues P, Burger PC, Scheithauer BW: The new WHO classification of brain tumours. Brain Pathol 3 (3): 255-68, 1993. [PUBMED Abstract]
Louis DN, Ohgaki H, Wiestler OD, et al.: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114 (2): 97-109, 2007. [PUBMED Abstract]
Louis DN, Ohgaki H, Wiestler OD, et al., eds.: WHO Classification of Tumours of the Central Nervous System. 4th ed. IARC Press, 2007.
Sievert AJ, Jackson EM, Gai X, et al.: Duplication of 7q34 in pediatric low-grade astrocytomas detected by high-density single-nucleotide polymorphism-based genotype arrays results in a novel BRAF fusion gene. Brain Pathol 19 (3): 449-58, 2009. [PUBMED Abstract]
Pfister S, Janzarik WG, Remke M, et al.: BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas. J Clin Invest 118 (5): 1739-49, 2008. [PUBMED Abstract]
Jones DT, Kocialkowski S, Liu L, et al.: Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res 68 (21): 8673-7, 2008. [PUBMED Abstract]
Dias-Santagata D, Lam Q, Vernovsky K, et al.: BRAF V600E mutations are common in pleomorphic xanthoastrocytoma: diagnostic and therapeutic implications. PLoS One 6 (3): e17948, 2011. [PUBMED Abstract]
MacConaill LE, Campbell CD, Kehoe SM, et al.: Profiling critical cancer gene mutations in clinical tumor samples. PLoS One 4 (11): e7887, 2009. [PUBMED Abstract]
Parsons DW, Jones S, Zhang X, et al.: An integrated genomic analysis of human glioblastoma multiforme. Science 321 (5897): 1807-12, 2008. [PUBMED Abstract]
Yan H, Parsons DW, Jin G, et al.: IDH1 and IDH2 mutations in gliomas. N Engl J Med 360 (8): 765-73, 2009. [PUBMED Abstract]
Dubbink HJ, Taal W, van Marion R, et al.: IDH1 mutations in low-grade astrocytomas predict survival but not response to temozolomide. Neurology 73 (21): 1792-5, 2009. [PUBMED Abstract]
Sanson M, Marie Y, Paris S, et al.: Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol 27 (25): 4150-4, 2009. [PUBMED Abstract]
Hartmann C, Hentschel B, Wick W, et al.: Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol 120 (6): 707-18, 2010. [PUBMED Abstract]
Hartmann C, Meyer J, Balss J, et al.: Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol 118 (4): 469-74, 2009. [PUBMED Abstract]
Jenkins RB, Blair H, Ballman KV, et al.: A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res 66 (20): 9852-61, 2006. [PUBMED Abstract]
Camelo-Piragua S, Jansen M, Ganguly A, et al.: A sensitive and specific diagnostic panel to distinguish diffuse astrocytoma from astrocytosis: chromosome 7 gain with mutant isocitrate dehydrogenase 1 and p53. J Neuropathol Exp Neurol 70 (2): 110-5, 2011. [PUBMED Abstract]
Capper D, Weissert S, Balss J, et al.: Characterization of R132H mutation-specific IDH1 antibody binding in brain tumors. Brain Pathol 20 (1): 245-54, 2010. [PUBMED Abstract]
Treatment Option Overview for Adult Primary CNS Tumors
Radiation therapy and chemotherapy options vary according to histology and anatomical site of the CNS tumor. For glioblastoma, combined modality therapy with resection, radiation, and chemotherapy is standard. Anaplastic astrocytomas, anaplastic oligodendrogliomas, and anaplastic oligoastrocytomas represent only a small proportion of CNS gliomas; therefore, phase III randomized trials restricted to these tumor types are not generally practical. The natural histories of these tumors are variable, depending on histological and molecular factors; therefore, treatment guidelines are evolving. Therapy involving surgically implanted carmustine-impregnated polymer wafers combined with postoperative external-beam radiation therapy (EBRT) may play a role in the treatment of high-grade (grades III and IV) gliomas in some patients.[1]
Treatment options for primary CNS tumors include the following:
For most types of CNS tumors in most locations, complete or near-complete surgical removal is generally attempted, within the constraints of preserving neurological function and the patient’s underlying health. This practice is based on observational evidence that survival is better in patients who undergo tumor resection than in those who have closed biopsy alone.[2,3] The benefit of resection has not been tested in randomized trials. Selection bias can enter into observational studies despite attempts to adjust for patient differences that guide the decision to resect the tumor; therefore, the actual difference in outcome between radical surgery and biopsy alone may not be as large as noted in the retrospective studies.[3]
An exception to the use of resection is the case of deep-seated tumors such as pontine gliomas, which are diagnosed on clinical evidence and treated without initial surgery approximately 50% of the time. In most cases, however, diagnosis by biopsy is preferred. Stereotactic biopsy can be used for lesions that are difficult to reach and resect.
The primary goals of surgical resection include the following:[4]
To establish a histological diagnosis.
To reduce intracranial pressure by removing as much tumor as is safely possible to preserve neurological function.
Total elimination of primary malignant intraparenchymal tumors by surgery alone is rarely achievable. Therefore, intraoperative techniques have been developed to reach a balance between removing as much tumor as is practical and preserving functional status. For example, craniotomies with stereotactic resections of primary gliomas can be performed in cooperative patients while they are awake, with real-time assessment of neurological function.[5] Examples of intraoperative neurological assessment include the following:
Resection proceeds until either the magnetic resonance imaging (MRI) signal abnormality being used to monitor the extent of surgery is completely removed or subtle neurological dysfunction appears (e.g., a slight decrease in rapid alternating motor movement or anomia).
When the tumor is located in or near language centers in the cortex, intraoperative language mapping can be performed by electrode discharge-induced speech arrest while the patient is asked to count or read.[6]
As is the case with several other specialized operations [7,8] in which postoperative mortality has been associated with the number of procedures performed, postoperative mortality after surgery for primary brain tumors may be associated with hospital and/or surgeon volume.[9] Using the Nationwide Inpatient Sample hospital discharge database for the years 1988 to 2000, which represented 20% of inpatient admissions to nonfederal U.S. hospitals, investigators observed the following:[9]
Large-volume hospitals had lower in-hospital mortality rates after craniotomies for primary brain tumors (odds ratio [OR], 0.75 for a tenfold higher caseload; 95% confidence interval [CI], 0.62–0.90) and after needle biopsies (OR, 0.54; 95% CI, 0.35–0.83).
Although there was no specific sharp threshold in all-cause mortality outcomes between low-volume hospitals and high-volume hospitals, craniotomy-associated in-hospital mortality was 4.5% for hospitals with 5 or fewer procedures per year and 1.5% for hospitals with at least 42 procedures per year.
In-hospital mortality rates decreased over the study years (perhaps because the proportion of elective nonemergent operations increased from 45% to 57%), but the decrease was more rapid in high-volume hospitals than in low-volume hospitals.
High-volume surgeons had lower in-hospital patient mortality rates after craniotomy (OR, 0.60; 95% CI, 0.45–0.79).
As with any study of volume-outcome associations, these results may not be causal because of residual confounding factors such as referral patterns, private insurance, and patient selection, despite multivariable adjustment.
Radiation therapy
High-grade tumors
Radiation therapy has a major role in the treatment of patients with high-grade gliomas.
A systematic review and meta-analysis of five randomized trials (plus one trial with allocation by birth date) comparing PORT with no radiation therapy showed a statistically significant survival advantage with radiation (risk ratio, 0.81; 95% CI, 0.74–0.88).[10][Level of evidence A1]
A randomized trial comparing 60 Gy (in 30 fractions over 6 weeks) with 45 Gy (in 25 fractions over 4 weeks) showed superior survival in the first group (12 months vs. 9 months median survival; hazard ratio [HR], 0.81; 95% CI, 0.66–0.99). The accepted standard dose of EBRT for malignant gliomas is 60 Gy.[11][Level of evidence A1]
EBRT using either 3-dimensional conformal radiation therapy (3D-CRT) or intensity-modulated radiation therapy (IMRT) is considered an acceptable technique in radiation therapy delivery. Typically used are 2- to 3-cm margins on the MRI-based volumes (T1-weighted and fluid-attenuated inversion recovery [FLAIR]) to create the planning target volume.
Dose escalation using radiosurgery has not improved outcomes. A randomized trial tested radiosurgery as a boost added to standard EBRT, but the trial found no improvement in survival, quality of life, or patterns of relapse compared with EBRT without the boost.[12,13]
Brachytherapy has been used to deliver high doses of radiation locally to the tumor while sparing normal brain tissue. However, this approach is technically demanding and is less common since the advent of 3D-CRT and IMRT.
Low-grade tumors
Treatment options for patients with low-grade gliomas (i.e., low-grade astrocytomas, oligodendrogliomas, and mixed oligoastrocytomas) are not as clear as in the case of high-grade tumors and include observation, PORT, and chemotherapy with temozolomide.
Evidence (PORT vs. observation):
The European Organisation for Research and Treatment of Cancer (EORTC) randomly assigned 311 patients with low-grade gliomas to undergo either radiation or observation in the EORTC-22845 trial.[14,15] On review of central pathology, about 25% of patients in the trial were reported to have high-grade tumors. Most of the control patients received radiation therapy at the time of progression.
After a median follow-up of 93 months, the median progression-free survival (PFS) was 5.3 years in the radiation arm versus 3.4 years in the control arm (HR, 0.59; 95% CI, 0.45–0.77).[14,15][Level of evidence B1]
There was no difference in the overall survival (OS). The median survival was 7.4 years in the radiation arm and 7.2 years in the control arm (HR, 0.97; 95% CI, 0.71–1.34; P = .87).[14,15][Level of evidence A1] This was caused by a longer survival after progression in the control arm (3.4 years) than in the radiation arm (1.0 year) (P < .0001).
The investigators did not collect reliable quality-of-life measurements, so it is not clear whether the delay in initial relapse in the radiation therapy arm translated into improved function or quality of life.
Evidence (PORT versus temozolomide for patients with low-grade World Health Organization [WHO] grade II tumors with at least one high-risk feature):
The EORTC 22033-26033 trial (NCT00182819) included 707 patients with low-grade glioma (WHO grade II astrocytoma, oligoastrocytoma, or oligodendroglioma) and at least one high-risk feature (age >40 years, progressive disease, tumor size >5 cm, tumor crossing the midline, or neurological symptoms). Patients were randomly assigned to receive either radiation therapy (n = 240) or temozolomide chemotherapy (n = 237). Radiation therapy consisted of conformal treatment (up to 50.4 Gy; 28 doses of 1.8 Gy daily, 5 days a week, for up to 6.5 weeks). Chemotherapy was dose-dense oral temozolomide (75 mg/m2 daily for 21 days, repeated every 28 days [one cycle], for a maximum of 12 cycles).[16,17]
There was no significant difference in PFS (primary end point) or health-related quality of life (secondary end point).
At a median follow-up of 48 months (interquartile range, 31–56), median PFS was 39 months (95% CI, 35–44) in the temozolomide group and 46 months (95% CI, 40–56) in the radiation therapy group (unadjusted HR, 1.16; 95% CI, 0.9–1.5; P = .22).[16][Level of evidence B1]
An exploratory analysis of 318 molecularly defined patients found that patients with IDH gene variants without codeletion of 1p/19q displayed a significantly longer PFS when treated with radiation therapy (HR, 1.86; 95% CI, 1.21–2.87; log-rank P = .0043).
There were no significant treatment-dependent differences in PFS for patients with IDH variants with codeletion of 1p/19q and IDH wild-type tumors.
Patients with wild-type IDH tumors had the worst prognosis independent of treatment type.
Patients with IDH variants with codeletion of 1p/19q had the best prognosis.
The O6-methylguanine-DNA methyltransferase (MGMT) promoter status was methylated in the following:
All IDH variants with codeletion of 1p/19q (45/45).
Sixty-two of 72 (86%) of the IDH variants without codeletion of 1p/19q.
Five of nine (56%) of the IDH wild-type cases.
Disease progression, subsequent neoplasms, or recurrences
There are no randomized trials to delineate the role of repeat radiation after disease progression or the development of radiation-induced cancers. The literature is limited to small retrospective case series, which makes interpretation difficult.[18] The decision to repeat radiation must be made carefully because of the risk of neurocognitive deficits and radiation-induced necrosis. One advantage of radiosurgery is the ability to deliver therapeutic doses to recurrent tumors that may require the re-irradiation of previously irradiated brain tissue beyond tolerable dose limits.
Chemotherapy
Systemic chemotherapy
For many years, the nitrosourea carmustine ([bis-chloroethylnitrosourea] BCNU) was the standard chemotherapy agent added to surgery and radiation therapy for malignant gliomas, based on the Radiation Therapy Oncology Group’s (RTOG’s) randomized trial (RTOG-8302).[19][Level of evidence A1] A modest impact on survival with the use of nitrosourea-containing chemotherapy regimens for malignant gliomas was confirmed in a patient-level meta-analysis of 12 randomized trials (combined HRdeath, 0.85; 95% CI, 0.78–0.91).[20]
A large multicenter trial (NCT00006353) of patients with glioblastoma, conducted by the EORTC-National Cancer Institute of Canada, reported a survival advantage with the use of temozolomide in addition to radiation therapy.[21,22][Level of evidence A1] On the basis of these results, the oral agent temozolomide has replaced BCNU as the standard systemic chemotherapy for malignant gliomas. For more information, see the Glioblastomas treatment section.
Long-term results of randomized trials in high-risk, low-grade (WHO grade II) gliomas [23][Level of evidence A1] and anaplastic (WHO grade III) oligodendroglial tumors [24,25][Level of evidence A1] have demonstrated that the addition of procarbazine, lomustine, and vincristine (PCV) chemotherapy to radiation therapy after surgery extends survival. Radiation and PCV chemotherapy should be considered for patients deemed appropriate for therapy. For more information, see the Treatment of Primary Central Nervous System Tumors by Tumor Type section.
Localized chemotherapy (carmustine wafer)
The ability to give high doses of chemotherapy while avoiding systemic toxicity is desirable because malignant glioma–related deaths are usually due to uncontrolled intracranial disease rather than distant metastases. A biodegradable carmustine wafer has been developed for that purpose. The wafers contain 3.85% carmustine, and up to eight wafers are implanted into the tumor bed lining at the time of open resection, with an intended total dose of about 7.7 mg per wafer (61.6 mg maximum per patient) over a period of 2 to 3 weeks.
Two randomized placebo-controlled trials of this focal drug-delivery method have shown an OS advantage associated with the carmustine wafers versus radiation therapy alone. In both trials, the upper age limit for patients was 65 years.
Evidence (carmustine wafer):
A small trial was closed because of a lack of continued availability of the carmustine wafers after 32 patients with high-grade gliomas had been entered.[26]
Although OS was better in the carmustine-wafer group (median 58.1 vs. 39.9 weeks; P = .012), there was an imbalance in the study arms (only 11 of 16 patients in the carmustine-wafer group vs. 16 of the 16 patients in the placebo-wafer group had grade IV glioblastoma tumors).
A multicenter study of 240 patients with primary malignant gliomas, 207 of whom had glioblastoma, was more informative.[27,28] At initial surgery, patients received either carmustine wafers or placebo wafers, followed by radiation therapy (55–60 Gy). Systemic therapy was not allowed until recurrence, except in the case of anaplastic oligodendrogliomas (n = 9). Unlike the initial trial, patient characteristics were well balanced between the study arms.
Median survival in the two groups was 13.8 months in patients treated with carmustine wafers versus 11.6 months in placebo-treated patients (HR, 0.73; 95% CI, 0.56–0.96; P = .017).
A systematic review combining both studies [26–28] estimated an HR for overall mortality of 0.65; 95% CI, 0.48–0.86; P = .003.[29][Level of evidence A1]
Active surveillance
Active surveillance is appropriate in some circumstances. With the increasing use of sensitive neuroimaging tools, detection of asymptomatic low-grade meningiomas has increased; most appear to show minimal growth and can often be safely observed, with therapy deferred until the detection of tumor growth or the development of symptoms.[30,31]
Supportive therapy
Dexamethasone, mannitol, and furosemide are used to treat the peritumoral edema associated with brain tumors. The use of anticonvulsants is mandatory for patients with seizures.[4]
References
Lallana EC, Abrey LE: Update on the therapeutic approaches to brain tumors. Expert Rev Anticancer Ther 3 (5): 655-70, 2003. [PUBMED Abstract]
Laws ER, Parney IF, Huang W, et al.: Survival following surgery and prognostic factors for recently diagnosed malignant glioma: data from the Glioma Outcomes Project. J Neurosurg 99 (3): 467-73, 2003. [PUBMED Abstract]
Chang SM, Parney IF, Huang W, et al.: Patterns of care for adults with newly diagnosed malignant glioma. JAMA 293 (5): 557-64, 2005. [PUBMED Abstract]
Meyer FB, Bates LM, Goerss SJ, et al.: Awake craniotomy for aggressive resection of primary gliomas located in eloquent brain. Mayo Clin Proc 76 (7): 677-87, 2001. [PUBMED Abstract]
Sanai N, Mirzadeh Z, Berger MS: Functional outcome after language mapping for glioma resection. N Engl J Med 358 (1): 18-27, 2008. [PUBMED Abstract]
Begg CB, Cramer LD, Hoskins WJ, et al.: Impact of hospital volume on operative mortality for major cancer surgery. JAMA 280 (20): 1747-51, 1998. [PUBMED Abstract]
Birkmeyer JD, Finlayson EV, Birkmeyer CM: Volume standards for high-risk surgical procedures: potential benefits of the Leapfrog initiative. Surgery 130 (3): 415-22, 2001. [PUBMED Abstract]
Barker FG, Curry WT, Carter BS: Surgery for primary supratentorial brain tumors in the United States, 1988 to 2000: the effect of provider caseload and centralization of care. Neuro Oncol 7 (1): 49-63, 2005. [PUBMED Abstract]
Laperriere N, Zuraw L, Cairncross G, et al.: Radiotherapy for newly diagnosed malignant glioma in adults: a systematic review. Radiother Oncol 64 (3): 259-73, 2002. [PUBMED Abstract]
Bleehen NM, Stenning SP: A Medical Research Council trial of two radiotherapy doses in the treatment of grades 3 and 4 astrocytoma. The Medical Research Council Brain Tumour Working Party. Br J Cancer 64 (4): 769-74, 1991. [PUBMED Abstract]
Tsao MN, Mehta MP, Whelan TJ, et al.: The American Society for Therapeutic Radiology and Oncology (ASTRO) evidence-based review of the role of radiosurgery for malignant glioma. Int J Radiat Oncol Biol Phys 63 (1): 47-55, 2005. [PUBMED Abstract]
Souhami L, Seiferheld W, Brachman D, et al.: Randomized comparison of stereotactic radiosurgery followed by conventional radiotherapy with carmustine to conventional radiotherapy with carmustine for patients with glioblastoma multiforme: report of Radiation Therapy Oncology Group 93-05 protocol. Int J Radiat Oncol Biol Phys 60 (3): 853-60, 2004. [PUBMED Abstract]
Karim AB, Afra D, Cornu P, et al.: Randomized trial on the efficacy of radiotherapy for cerebral low-grade glioma in the adult: European Organization for Research and Treatment of Cancer Study 22845 with the Medical Research Council study BRO4: an interim analysis. Int J Radiat Oncol Biol Phys 52 (2): 316-24, 2002. [PUBMED Abstract]
van den Bent MJ, Afra D, de Witte O, et al.: Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 366 (9490): 985-90, 2005. [PUBMED Abstract]
Baumert BG, Hegi ME, van den Bent MJ, et al.: Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 17 (11): 1521-1532, 2016. [PUBMED Abstract]
Reijneveld JC, Taphoorn MJ, Coens C, et al.: Health-related quality of life in patients with high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study. Lancet Oncol 17 (11): 1533-1542, 2016. [PUBMED Abstract]
Paulino AC, Mai WY, Chintagumpala M, et al.: Radiation-induced malignant gliomas: is there a role for reirradiation? Int J Radiat Oncol Biol Phys 71 (5): 1381-7, 2008. [PUBMED Abstract]
Walker MD, Green SB, Byar DP, et al.: Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med 303 (23): 1323-9, 1980. [PUBMED Abstract]
Stewart LA: Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 359 (9311): 1011-8, 2002. [PUBMED Abstract]
Stupp R, Mason WP, van den Bent MJ, et al.: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352 (10): 987-96, 2005. [PUBMED Abstract]
Stupp R, Hegi ME, Mason WP, et al.: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10 (5): 459-66, 2009. [PUBMED Abstract]
Buckner JC, Pugh SL, Shaw EG, et al.: Phase III study of radiation therapy with or without procarbazine, CCNU, and vincristine (PCV) in low-grade glioma: RTOG 9802 with Alliance, ECOG, and SWOG. [Abstract] J Clin Oncol 32 (Suppl 5): A-2000, 2014.
van den Bent MJ, Brandes AA, Taphoorn MJ, et al.: Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 31 (3): 344-50, 2013. [PUBMED Abstract]
Cairncross G, Wang M, Shaw E, et al.: Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol 31 (3): 337-43, 2013. [PUBMED Abstract]
Valtonen S, Timonen U, Toivanen P, et al.: Interstitial chemotherapy with carmustine-loaded polymers for high-grade gliomas: a randomized double-blind study. Neurosurgery 41 (1): 44-8; discussion 48-9, 1997. [PUBMED Abstract]
Westphal M, Hilt DC, Bortey E, et al.: A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro-oncol 5 (2): 79-88, 2003. [PUBMED Abstract]
Westphal M, Ram Z, Riddle V, et al.: Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir (Wien) 148 (3): 269-75; discussion 275, 2006. [PUBMED Abstract]
Hart MG, Grant R, Garside R, et al.: Chemotherapeutic wafers for high grade glioma. Cochrane Database Syst Rev (3): CD007294, 2008. [PUBMED Abstract]
Nakamura M, Roser F, Michel J, et al.: The natural history of incidental meningiomas. Neurosurgery 53 (1): 62-70; discussion 70-1, 2003. [PUBMED Abstract]
Yano S, Kuratsu J; Kumamoto Brain Tumor Research Group: Indications for surgery in patients with asymptomatic meningiomas based on an extensive experience. J Neurosurg 105 (4): 538-43, 2006. [PUBMED Abstract]
Treatment of Primary CNS Tumors by Tumor Type
Table 2. Treatment of Primary Central Nervous System Tumors by Tumor Type
Patients with brain stem gliomas have relatively poor prognoses that correlate with histology (when biopsies are performed), location, and extent of tumor. The overall median survival time of patients in studies has been 44 to 74 weeks.
Treatment options for brain stem gliomas include the following:
Radiation therapy.
Pineal astrocytic tumors treatment
Depending on the degree of anaplasia, patients with pineal astrocytomas have variable prognoses. Patients with higher-grade tumors have worse prognoses.
Treatment options for pineal astrocytic tumors include the following:
Surgery plus radiation therapy for pineal astrocytoma.
Surgery plus radiation therapy and chemotherapy for higher-grade tumors.
Pilocytic astrocytomas treatment
This astrocytic tumor is classified as a World Health Organization (WHO) grade I tumor and is often curable.
Treatment options for pilocytic astrocytomas include the following:
Surgery alone if the tumor is totally resectable.
Surgery followed by radiation therapy to known or suspected residual tumor.
Diffuse astrocytomas treatment
This WHO grade II astrocytic tumor is less often curable than is a pilocytic astrocytoma.
Treatment options for diffuse astrocytomas (WHO grade II) include the following:
Surgery with or without radiation therapy.
Surgery followed by radiation therapy and chemotherapy.
Controversy exists about the timing of radiation therapy after surgery. For more information, see the Low-grade tumors section.
Radiation therapy improved progression-free survival (PFS) in patients who received early radiation therapy in the European Organisation for Research and Treatment of Cancer (EORTC) EORTC-22845 trial. For more information, see the Oligodendrogliomas treatment section.[1][Level of evidence A1]
In the same trial, there was no difference in overall survival (OS) between patients who had radiation therapy after surgery and those who were treated with radiation therapy at the time of progression.[1][Level of evidence A1]
Some physicians use surgery alone if a patient has clinical factors that are considered low risk, such as age younger than 40 years and the lack of contrast enhancement on a computed tomography scan.[2]
Evidence (surgery followed by radiation therapy and chemotherapy):
For patients with low-grade (WHO grade II) tumors, which are considered high risk, radiation therapy followed by six cycles of vincristine (PCV) chemotherapy is a recommended option. This recommendation is based on the long-term follow-up results of the Radiation Therapy Oncology Group’s (RTOG’s) 1986-initiated randomized trial (RTOG 9802 [NCT00003375]).[3][Level of evidence A1] In this trial, patients with high-risk, low-grade glioma, defined as patients aged 18 to 39 years with biopsy or subtotal resection, or patients aged 40 years or older, were randomly assigned to either 54 Gy of radiation therapy or radiation therapy followed by six cycles of PCV chemotherapy.
The addition of PCV to radiation therapy increased median PFS from 4.0 years to 10.4 years (hazard ratio [HR], 0.50; P = .002) and median OS from 7.8 years to 13.3 years (HR, 0.59; P = .03).
Notably, the RTOG 9802 study enrolled patients with a variety of tumors, including astrocytomas, oligodendrogliomas, and mixed oligoastrocytomas.
In a risk-adjusted multivariate analysis, patients treated with PCV and patients with an oligodendroglial histology had better survival outcomes. A subset analysis of histological type suggested that the addition of PCV mainly benefited patients with oligodendroglial tumors, although this data is yet to be validated.[4]
Median OS for PCV versus the control arm was not reached versus 10.8 years for oligodendrogliomas (P = .008), 11.4 years versus 5.9 years for oligoastrocytomas (P = .05), and 7.7 years versus 4.4 years for astrocytomas (P = .31).
The discovery of the IDH1 and IDH2 variants in diffuse gliomas has greatly helped to identify patients with high-risk disease. Large retrospective studies have demonstrated that IDH1 and IDH2 variants are powerful independent prognostic factors for improved survival.[5–9] Most WHO grade II and III gliomas harbor IDH1 and IDH2 variants,[6,10,11] and, therefore, those variants should be included in the assessment of high risk. Molecular correlative data from the RTOG 98-02 trial, which would be informative about which patients benefited the most from the addition of PCV, have not been reported.
Anaplastic astrocytomas treatment
Patients with anaplastic astrocytomas (WHO grade III) have a low cure rate with standard local treatment.
Treatment options for anaplastic astrocytomas include the following:
Surgery plus radiation therapy with or without chemotherapy.
Surgery plus chemotherapy.
A subset of anaplastic astrocytomas is aggressive; these tumors are frequently managed in the same way as glioblastomas, with surgery and radiation, and often with chemotherapy. However, the optimal treatment for these tumors is not established. Two phase III randomized trials restricted to patients with anaplastic gliomas (NCT00626990 and NCT00887146) are active, but efficacy data are not available. It is not known whether the improved survival of patients with chemotherapy-treated glioblastoma can be extrapolated to patients with anaplastic astrocytomas.
IDH1 and IDH2 variants are present in 50% to 70% of anaplastic astrocytomas and are independently associated with significantly improved survival.[6,9] Assessment of IDH1 and IDH2 variant status may guide decisions about treatment options.
Evidence (surgery plus radiation therapy or chemotherapy):
Postoperative radiation alone has been compared with postoperative chemotherapy alone in patients with anaplastic gliomas (i.e., 144 astrocytomas, 91 oligoastrocytomas, and 39 oligodendrogliomas), with crossover to the other modality at the time of tumor progression. Of the 139 patients randomly assigned to undergo radiation therapy, 135 were randomly assigned to receive chemotherapy, with a 32-week course of either PCV or single-agent temozolomide (2:1:1 randomization).[12][Levels of evidence A1 and B1]
The order of the modalities did not affect time-to-treatment failure (TTF) or OS.
Neither TTF nor OS differed across the treatment arms.
Patients with anaplastic astrocytomas are appropriate candidates for clinical trials designed to improve local control by adding newer forms of treatment to standard treatment. Information about ongoing clinical trials is available from the NCI website.
Glioblastomas treatment
For patients with glioblastoma (WHO grade IV), the cure rate is very low with standard local treatment.
Methylation of the promoter of the MGMT DNA repair enzyme gene is an independent prognostic factor for improved survival in newly diagnosed glioblastoma.[13,14] MGMT promoter methylation and concomitant inactivation of the DNA repair enzyme activities may also predict for response to temozolomide chemotherapy.[13] However, the clinical data that MGMT promoter methylation is a predictive marker is less certain.
Treatment options for patients with newly diagnosed glioblastoma include the following:
Surgery plus radiation therapy and chemotherapy.
Surgery plus radiation therapy.
Carmustine-impregnated polymer implanted during initial surgery.
Radiation therapy and concurrent chemotherapy.
The standard treatment for patients with newly diagnosed glioblastoma is surgery followed by concurrent radiation therapy and daily temozolomide, and then followed by six cycles of temozolomide. The addition of bevacizumab to radiation therapy and temozolomide did not improve OS.
Evidence (surgery plus radiation therapy and chemotherapy):
Standard therapy is based on a large, multicenter, randomized trial (NCT00006353) conducted by the EORTC and National Cancer Institute of Canada (NCIC). This trial reported a survival benefit with concurrent radiation therapy and temozolomide, compared with radiation therapy alone.[15,16][Level of evidence A1] In this study, 573 patients with glioblastoma were randomly assigned to receive standard radiation to the tumor volume with a 2- to 3-cm margin (60 Gy, 2 Gy per fraction, over 6 weeks) alone or with temozolomide (75 mg/m2 orally per day during radiation therapy for up to 49 days, followed by a 4-week break and then up to six cycles of five daily doses every 28 days at a dose of 150 mg/m2, increasing to 200 mg/m2 after the first cycle).
OS was statistically significantly better in the combined radiation therapy–temozolomide group (HRdeath, 0.6; 95% confidence interval [CI], 0.5–0.7; OS rate at 3 years was 16.0% for the radiation therapy–temozolomide group vs. 4.4% in the radiation therapy–alone group).
A companion molecular correlation subset study to the EORTC-NCIC trial provided strong evidence that epigenetic silencing of the MGMT DNA-repair gene by promoter DNA methylation was associated with increased OS in patients with newly diagnosed glioblastoma.[13]
MGMT promoter methylation was an independent favorable prognostic factor (HR, 0.45; 95% CI, 0.32–0.61; log-rank P < .001).
The median OS for patients with MGMT methylation was 18.2 months (95% CI, 15.5–22.0), compared with 12.2 months (95% CI, 11.4–13.5) for patients without MGMT methylation.
To test whether protracted (dose-dense) temozolomide enhances treatment response in patients with newly diagnosed glioblastoma, a multicenter, randomized, phase III trial conducted by the RTOG, EORTC, and the North Central Cancer Therapy Group, RTOG 0525 (NCT00304031), compared standard adjuvant temozolomide treatment (days 1–5 of a 28-day cycle) with a dose-dense schedule (days 1–21 of a 28-day cycle). All patients were treated with surgery followed by radiation therapy and concurrent daily temozolomide. Patients were then randomly assigned to receive either standard adjuvant temozolomide or dose-dense temozolomide.[14][Level of evidence A1]
Among 833 randomly assigned patients, no statistically significant difference between standard and dose-dense temozolomide was observed for median OS (16.6 months for standard temozolomide vs. 14.9 months for dose-dense temozolomide; HR, 1.03; P = .63) or for median PFS (5.5 vs. 6.7 months; HR, 0.87; P = .06).
Protracted temozolomide, which depletes intracellular MGMT, was predicted to have greater efficacy in tumors with MGMT-promoter methylation. To test this retrospectively, MGMT status was determined in 86% of randomly assigned patients. No difference in efficacy was observed in either the MGMT-methylated or MGMT-unmethylated subsets. There was no survival advantage for the use of dose-dense temozolomide versus standard-dose temozolomide in newly diagnosed glioblastoma patients, regardless of MGMT status. However, this study confirmed the strong prognostic effect of MGMT methylation because the median OS was 21.2 months (95% CI, 17.9–24.8) for patients with methylation versus 14 months (HR, 1.74; 95% CI, 12.9–14.7; P < .001) for patients without methylation.
The efficacy of dose-dense temozolomide for patients who have recurrent glioblastoma, however, is yet to be determined.
Evidence (surgery and chemoradiation therapy with or without bevacizumab):
In 2013, final data from two multicenter, phase III, randomized, double-blind, placebo-controlled trials of bevacizumab in patients who had newly diagnosed glioblastoma were reported: RTOG 0825 (NCT00884741) and the Roche-sponsored AVAglio (NCT00943826).[17,18][Level of evidence A1] Bevacizumab did not improve OS in either trial.
There was significant crossover in both trials. Approximately 40% of RTOG 0825 patients and approximately 30% of AVAglio patients received bevacizumab at the first sign of disease progression.
RTOG 0825 (NCT00884741): Patients were randomly assigned to receive standard therapy (chemoradiation therapy with temozolomide) or standard therapy plus bevacizumab. OS and PFS were coprimary end points.[17][Level of evidence A1]
Bevacizumab did not improve OS (median OS was 16–17 months for each arm). However, it increased median PFS (10.7 months in the bevacizumab arm vs. 7.3 months in the placebo arm; HR, 0.79; P = .007).
The PFS result in the RTOG 0825 trial did not meet the prespecified significance level (P = .004).
AVAglio (NCT00943826): Patients were randomly assigned to receive standard therapy (chemoradiation therapy with temozolomide) or standard therapy plus bevacizumab. OS and PFS were coprimary end points.[18][Level of evidence A1]
Bevacizumab did not improve OS (median OS was 16–17 months for each arm). However, it increased median PFS (10.6 months in the bevacizumab arm vs. 6.2 months in the placebo arm; HR, 0.64; P < .0001).
The PFS result was statistically significant and associated with clinical benefit because patients who received bevacizumab remained functionally independent longer (9.0 months in the bevacizumab arm vs. 6.0 months in the standard therapy arm) and had a longer time until their Karnofsky Performance status deteriorated (HR, 0.65; P < .0001).
Patients who received bevacizumab also had delayed initiation of corticosteroids (12.3 months vs. 3.7 months; HR, 0.71; P = .002), and more patients were able to discontinue corticosteroids if they were already taking them (66% in the bevacizumab arm vs. 47% in the standard therapy arm).
The two trials had contradictory results in health-related quality of life (HRQOL) and neurocognitive outcomes studies. In the mandatory HRQOL studies in the AVAglio trial, bevacizumab-treated patients experienced improved HRQOL, but bevacizumab-treated patients in the elective RTOG 0825 studies showed more decline in patient-reported HRQOL and neurocognitive function. The reasons for these discrepancies are unclear.
Based on these results, there is no definite evidence that the addition of bevacizumab to standard therapy is beneficial for all newly diagnosed glioblastoma patients. Certain subgroups may benefit from the addition of bevacizumab, but this is not yet known.
Patients with glioblastoma are appropriate candidates for clinical trials designed to improve local control by adding newer forms of treatment to standard treatment. Information about ongoing clinical trials is available from the NCI website.
Oligodendroglial Tumors Treatment
Oligodendrogliomas treatment
Patients who have oligodendrogliomas (WHO grade II) generally have better prognoses than do patients who have diffuse astrocytomas. In particular, patients who have oligodendrogliomas with 1p/19q codeletion have a much longer survival.[3] Most of the oligodendrogliomas eventually progress.
Treatment options for oligodendrogliomas include the following:
Surgery with or without radiation therapy.
Surgery with radiation therapy and chemotherapy.
Controversy exists concerning the timing of radiation therapy after surgery. A study (EORTC-22845) of 300 patients with low-grade gliomas who had surgery and were randomly assigned to either radiation therapy or watchful waiting, did not show a difference in OS between the two groups.[1][Level of evidence A1] For more information, see the Low-grade tumors section.
For low-grade (WHO grade II) tumors that are considered high risk, radiation therapy followed by six cycles of PCV chemotherapy is a recommended option based on the long-term follow-up results of RTOG-9802, a randomized trial for high-risk, low-grade gliomas.[3][Level of evidence A1] Notably, RTOG-9802 enrolled patients with a variety of tumors, including astrocytomas, oligodendrogliomas, and mixed oligoastrocytomas. In a retrospective subset analysis, only the oligodendroglial tumors appeared to benefit from the addition of PCV.[4]. For more information, see the Diffuse astrocytomas treatment section.
The discovery of the IDH1 and IDH2 variants, which are independent prognostic factors for significantly improved survival in diffuse gliomas, has greatly helped to identify patients with high-risk disease. For more information, see the Diffuse astrocytomas treatment section. In addition, a high proportion of WHO grade II oligodendrogliomas have 1p/19q codeletion, which is a powerful prognostic factor for improved survival.[19–21] Therefore, the presence of IDH1 and IDH2 variants and 1p/19q codeletion should be included in the assessment of high risk. Molecular correlative data from the RTOG-9802 trial, which would be informative about which patients benefited most from the addition of PCV, have not been reported.
Anaplastic oligodendrogliomas treatment
Patients with anaplastic oligodendrogliomas (WHO grade III) have a low cure rate with standard local treatment, but their prognoses are generally better than are the prognoses of patients with anaplastic astrocytomas. Prognoses are particularly better for patients with 1p/19q codeletion, which occurs in most of these tumors. Two phase III randomized trials restricted to patients with anaplastic gliomas (NCT00626990 and NCT00887146) are active; however, efficacy data are not yet available. For more information, see the Anaplastic astrocytomas treatment section. These patients are appropriate candidates for clinical trials designed to improve local control by adding newer forms of treatment.
Information about ongoing clinical trials is available from the NCI website.
Treatment options for anaplastic oligodendrogliomas include the following:
Surgery plus radiation therapy with or without chemotherapy.[22]
Evidence (surgery followed by radiation therapy with or without chemotherapy):
Mature results from the EORTC Brain Tumor Group Study 26951 (NCT00002840), a phase III randomized study with 11.7 years of follow-up, demonstrated increased OS and PFS in patients with anaplastic oligodendroglial tumors with six cycles of adjuvant PCV chemotherapy after radiation therapy, compared with radiation therapy alone.[23][Level of evidence A1]
OS was significantly longer in the radiation therapy and PCV arm (42.3 months vs. 30.6 months; HR, 0.75; 95% CI, 0.60–0.95).
Patients with 1p/19q-codeleted tumors derived more benefit from adjuvant PCV chemotherapy than did those with non–1p/19q-deleted tumors.[23]
In contrast, the RTOG trial (RTOG-9402 [NCT00002569]) demonstrated no differences in median survival by treatment arm between an 8-week, intensive PCV chemotherapy regimen followed by immediate involved-field-plus-radiation therapy and radiation therapy alone.[24]
In an unplanned subgroup analysis, patients with 1p/19q-codeleted anaplastic oligodendrogliomas and mixed anaplastic astrocytomas demonstrated a median survival of 14.7 years versus 7.3 years (HR, 0.59; 95% CI, 0.37–0.95; P = .03).
For patients with non-codeleted tumors, there was no difference in median survival by treatment arm (2.6 vs. 2.7 years; HR, 0.85; 95% CI, 0.58–1.23; P = .39).[24][Level of evidence A1]
Postoperative radiation therapy alone has been compared with postoperative chemotherapy alone in patients with anaplastic gliomas (including 144 astrocytomas, 91 oligoastrocytomas, and 39 oligodendrogliomas) with crossover to the other modality at the time of tumor progression. Of the 139 patients randomly assigned to undergo radiation therapy, 135 were randomly assigned to receive chemotherapy, with a 32-week course of either PCV or single-agent temozolomide (2:1:1 randomization).[12][Levels of evidence A1 and B1]
TTF or OS did not differ across the treatment arms and were not affected by the order of the modalities.
On the basis of these data, CODEL (NCT00887146), a study that randomly assigned patients to receive radiation therapy alone (control arm), radiation therapy with temozolomide, and temozolomide alone (exploratory arm), was halted because radiation therapy alone was no longer considered adequate treatment in patients with anaplastic oligodendroglioma with 1p/19q-codeletions.[25] Temozolomide and PCV chemotherapy in anaplastic oligodendroglioma have not been compared, although in the setting of grade III anaplastic gliomas, no survival difference was seen between PCV chemotherapy and temozolomide.[12,26]
The combination of radiation and chemotherapy is not known to be superior in outcome to sequential modality therapy.
A high proportion of anaplastic oligodendrogliomas have IDH1 andIDH2 variants and 1p/19q codeletion, both powerful prognostic factors for improved survival. For more information, see the Diffuse astrocytomas treatment section.[23,24] In addition, PCV chemotherapy has been shown to be predictive in a retrospective analysis of the phase III trials described earlier. Therefore, assessment of these molecular markers may aid management decisions for anaplastic oligodendrogliomas.
Mixed Gliomas Treatment
Patients with mixed glial tumors, which include oligoastrocytoma (WHO grade II) and anaplastic oligoastrocytoma (WHO grade III), have highly variable prognoses based on their status of the IDH1 and IDH2 genes and 1p/19q chromosomes.[27–29] Therefore, the optimal treatment for these tumors as a group is uncertain. Often, they are treated similarly to astrocytic tumors because a subset of tumors may have outcomes similar to WHO grade III astrocytic or WHO grade IV glioblastoma tumors. Testing for these known, strong prognostic molecular markers should be performed, which may help to guide the assessment of risk and subsequent management.
Treatment options for mixed gliomas include the following:
Surgery plus radiation therapy with or without chemotherapy.
Ependymal tumors (WHO grade I) and ependymomas (WHO grade II)—i.e., subependymomas and myxopapillary ependymomas—are often curable.
Treatment options for grades I and II ependymal tumors include the following:
Surgery alone if the tumor is totally resectable.
Surgery followed by radiation therapy to known or suspected residual tumor.
Patients with anaplastic ependymomas (WHO grade III) have variable prognoses that depend on the location and extent of disease. Frequently, but not invariably, patients with anaplastic ependymomas have worse prognoses than do those patients with lower-grade ependymal tumors.
Treatment options for anaplastic ependymomas include the following:
Treatment options for medulloblastomas include the following:
Surgery plus craniospinal radiation therapy for patients with good-risk disease.[32]
Surgery plus craniospinal radiation therapy and various chemotherapy regimens for patients with poor-risk disease (under clinical evaluation).[32]
Pineal Parenchymal Tumors Treatment
Pineocytomas (WHO grade II), pineoblastomas (WHO grade IV), and pineal parenchymal tumors of intermediate differentiation are diverse tumors that require special consideration. Pineocytomas are slow-growing tumors and prognosis varies.
Pineoblastomas grow more rapidly and patients with these tumors have worse prognoses. Pineal parenchymal tumors of intermediate differentiation have unpredictable growth and clinical behavior.
Treatment options for pineal parenchymal tumors include the following:
Surgery plus radiation therapy for pineocytoma.
Surgery plus radiation therapy and chemotherapy for pineoblastoma.
Meningeal Tumors Treatment
WHO grade I meningiomas are usually curable when they are resectable. With the increasing use of sensitive neuroimaging tools, there has been more detection of asymptomatic low-grade meningiomas. Most appear to show minimal growth and can often be safely observed while therapy is deferred until growth or the development of symptoms.[33,34]
Treatment options for meningeal tumors include the following:
Active surveillance with deferred treatment, especially for incidentally discovered asymptomatic tumors.[33,34]
Surgery.
Stereotactic radiosurgery for tumors smaller than 3 cm.
Surgery plus radiation therapy in selected cases, such as for patients with known or suspected residual disease or with recurrence after previous surgery.
Fractionated radiation therapy for patients with unresectable tumors.[35]
The prognoses for patients with WHO grade II meningiomas (atypical, clear cell, and chordoid), WHO grade III meningiomas (anaplastic/malignant, rhabdoid, and papillary), and hemangiopericytomas are worse than the prognoses for patients with low-grade meningiomas because complete resections are less commonly feasible, and the proliferative capacity is greater.
Treatment options for grades II and III meningiomas and hemangiopericytomas include the following:
Surgery plus radiation therapy.
Germ Cell Tumors Treatment
The prognoses and treatment of patients with germ cell tumors—which include germinomas, embryonal carcinomas, choriocarcinomas, and teratomas—depend on tumor histology, tumor location, presence and levels of biological markers, and surgical resectability.
Treatment of Tumors of the Sellar Region
Craniopharyngiomas (WHO grade I) are often curable.
Treatment options for craniopharyngiomas include the following:
Surgery alone if the tumor is totally resectable.
Debulking surgery plus radiation therapy if the tumor is unresectable.
Treatment Options Under Clinical Evaluation for Primary CNS Tumors
Patients who have central nervous system (CNS) tumors that are either infrequently curable or unresectable should consider enrollment in clinical trials. Information about ongoing clinical trials is available from the NCI website.
Heavy-particle radiation, such as proton-beam therapy, carries the theoretical advantage of delivering high doses of ionizing radiation to the tumor bed while sparing surrounding brain tissue. The data are preliminary for this investigational technique and are not widely available.
Novel biological therapies under clinical evaluation for patients with CNS tumors include the following:[36]
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
van den Bent MJ, Afra D, de Witte O, et al.: Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 366 (9490): 985-90, 2005. [PUBMED Abstract]
Buckner JC, Pugh SL, Shaw EG, et al.: Phase III study of radiation therapy with or without procarbazine, CCNU, and vincristine (PCV) in low-grade glioma: RTOG 9802 with Alliance, ECOG, and SWOG. [Abstract] J Clin Oncol 32 (Suppl 5): A-2000, 2014.
Buckner JC, Shaw E, Pugh S, et al.: R9802: Phase III study of radiation therapy with or without procarbazine, CCNU, and vincristine (PCV) in low-grade glioma: Results by histologic type. [Abstract] Neuro Oncol 16 (Suppl 5): A-AT-13, v11, 2014.
Parsons DW, Jones S, Zhang X, et al.: An integrated genomic analysis of human glioblastoma multiforme. Science 321 (5897): 1807-12, 2008. [PUBMED Abstract]
Yan H, Parsons DW, Jin G, et al.: IDH1 and IDH2 mutations in gliomas. N Engl J Med 360 (8): 765-73, 2009. [PUBMED Abstract]
Dubbink HJ, Taal W, van Marion R, et al.: IDH1 mutations in low-grade astrocytomas predict survival but not response to temozolomide. Neurology 73 (21): 1792-5, 2009. [PUBMED Abstract]
Sanson M, Marie Y, Paris S, et al.: Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol 27 (25): 4150-4, 2009. [PUBMED Abstract]
Hartmann C, Hentschel B, Wick W, et al.: Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol 120 (6): 707-18, 2010. [PUBMED Abstract]
Hartmann C, Meyer J, Balss J, et al.: Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol 118 (4): 469-74, 2009. [PUBMED Abstract]
Watanabe T, Nobusawa S, Kleihues P, et al.: IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am J Pathol 174 (4): 1149-53, 2009. [PUBMED Abstract]
Wick W, Hartmann C, Engel C, et al.: NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol 27 (35): 5874-80, 2009. [PUBMED Abstract]
Hegi ME, Diserens AC, Gorlia T, et al.: MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med 352 (10): 997-1003, 2005. [PUBMED Abstract]
Gilbert MR, Wang M, Aldape KD, et al.: Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J Clin Oncol 31 (32): 4085-91, 2013. [PUBMED Abstract]
Stupp R, Mason WP, van den Bent MJ, et al.: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352 (10): 987-96, 2005. [PUBMED Abstract]
Stupp R, Hegi ME, Mason WP, et al.: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 10 (5): 459-66, 2009. [PUBMED Abstract]
Gilbert MR, Dignam JJ, Armstrong TS, et al.: A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370 (8): 699-708, 2014. [PUBMED Abstract]
Chinot OL, Wick W, Mason W, et al.: Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370 (8): 709-22, 2014. [PUBMED Abstract]
Fallon KB, Palmer CA, Roth KA, et al.: Prognostic value of 1p, 19q, 9p, 10q, and EGFR-FISH analyses in recurrent oligodendrogliomas. J Neuropathol Exp Neurol 63 (4): 314-22, 2004. [PUBMED Abstract]
Smith JS, Perry A, Borell TJ, et al.: Alterations of chromosome arms 1p and 19q as predictors of survival in oligodendrogliomas, astrocytomas, and mixed oligoastrocytomas. J Clin Oncol 18 (3): 636-45, 2000. [PUBMED Abstract]
Okamoto Y, Di Patre PL, Burkhard C, et al.: Population-based study on incidence, survival rates, and genetic alterations of low-grade diffuse astrocytomas and oligodendrogliomas. Acta Neuropathol 108 (1): 49-56, 2004. [PUBMED Abstract]
van den Bent MJ, Chinot O, Boogerd W, et al.: Second-line chemotherapy with temozolomide in recurrent oligodendroglioma after PCV (procarbazine, lomustine and vincristine) chemotherapy: EORTC Brain Tumor Group phase II study 26972. Ann Oncol 14 (4): 599-602, 2003. [PUBMED Abstract]
van den Bent MJ, Brandes AA, Taphoorn MJ, et al.: Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 31 (3): 344-50, 2013. [PUBMED Abstract]
Cairncross G, Wang M, Shaw E, et al.: Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol 31 (3): 337-43, 2013. [PUBMED Abstract]
Gilbert MR: Minding the Ps and Qs: perseverance and quality studies lead to major advances in patients with anaplastic oligodendroglioma. J Clin Oncol 31 (3): 299-300, 2013. [PUBMED Abstract]
Brada M, Stenning S, Gabe R, et al.: Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma. J Clin Oncol 28 (30): 4601-8, 2010. [PUBMED Abstract]
Jiao Y, Killela PJ, Reitman ZJ, et al.: Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas. Oncotarget 3 (7): 709-22, 2012. [PUBMED Abstract]
Killela PJ, Reitman ZJ, Jiao Y, et al.: TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc Natl Acad Sci U S A 110 (15): 6021-6, 2013. [PUBMED Abstract]
Killela PJ, Pirozzi CJ, Healy P, et al.: Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas. Oncotarget 5 (6): 1515-25, 2014. [PUBMED Abstract]
Oya N, Shibamoto Y, Nagata Y, et al.: Postoperative radiotherapy for intracranial ependymoma: analysis of prognostic factors and patterns of failure. J Neurooncol 56 (1): 87-94, 2002. [PUBMED Abstract]
Brandes AA, Ermani M, Amista P, et al.: The treatment of adults with medulloblastoma: a prospective study. Int J Radiat Oncol Biol Phys 57 (3): 755-61, 2003. [PUBMED Abstract]
Brandes AA, Franceschi E, Tosoni A, et al.: Long-term results of a prospective study on the treatment of medulloblastoma in adults. Cancer 110 (9): 2035-41, 2007. [PUBMED Abstract]
Nakamura M, Roser F, Michel J, et al.: The natural history of incidental meningiomas. Neurosurgery 53 (1): 62-70; discussion 70-1, 2003. [PUBMED Abstract]
Yano S, Kuratsu J; Kumamoto Brain Tumor Research Group: Indications for surgery in patients with asymptomatic meningiomas based on an extensive experience. J Neurosurg 105 (4): 538-43, 2006. [PUBMED Abstract]
Debus J, Wuendrich M, Pirzkall A, et al.: High efficacy of fractionated stereotactic radiotherapy of large base-of-skull meningiomas: long-term results. J Clin Oncol 19 (15): 3547-53, 2001. [PUBMED Abstract]
Fine HA: Promising new therapies for malignant gliomas. Cancer J 13 (6): 349-54, 2007 Nov-Dec. [PUBMED Abstract]
Fecci PE, Mitchell DA, Archer GE, et al.: The history, evolution, and clinical use of dendritic cell-based immunization strategies in the therapy of brain tumors. J Neurooncol 64 (1-2): 161-76, 2003 Aug-Sep. [PUBMED Abstract]
Newton HB: Molecular neuro-oncology and development of targeted therapeutic strategies for brain tumors. Part 1: Growth factor and Ras signaling pathways. Expert Rev Anticancer Ther 3 (5): 595-614, 2003. [PUBMED Abstract]
Kew Y, Levin VA: Advances in gene therapy and immunotherapy for brain tumors. Curr Opin Neurol 16 (6): 665-70, 2003. [PUBMED Abstract]
Chiocca EA, Aghi M, Fulci G: Viral therapy for glioblastoma. Cancer J 9 (3): 167-79, 2003 May-Jun. [PUBMED Abstract]
Treatment of Primary Tumors of the Spinal Axis
Surgery and radiation therapy are the primary modalities used to treat tumors of the spinal axis. Therapeutic options vary according to the histology of the tumor.[1] The experience with chemotherapy for primary spinal cord tumors is limited; no reports of controlled clinical trials are available for these types of tumors.[1,2] Chemotherapy is indicated for most patients with leptomeningeal involvement from a primary or metastatic tumor and positive cerebrospinal fluid cytology.[1] Most patients require treatment with corticosteroids, particularly if they are receiving radiation therapy.
Patients who have spinal axis tumors that are either infrequently curable or unresectable should consider enrollment in clinical trials. Information about ongoing clinical trials is available from the NCI website.
Mehta M, Vogelbaum MA, Chang S, et al.: Neoplasms of the central nervous system. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 1700-49.
Metastatic Brain Tumors
General Information About Metastatic Brain Tumors
Brain metastases outnumber primary neoplasms by at least 10 to 1, and they occur in 20% to 40% of cancer patients, with subsequent median survival generally less than 6 months.[1] The exact incidence is unknown because no national cancer registry documents brain metastases, but it has been estimated that 98,000 to 170,000 new cases are diagnosed in the United States each year.[2,3] This number may be increasing because of the capacity of magnetic resonance imaging (MRI) to detect small metastases and because of prolonged survival resulting from improved systemic therapy.[1,2]
The most common primary tumors with brain metastases and the percentage of patients affected are as follows:[1,2]
Lung (18%–64%).
Breast (2%–21%).
Cancer of unknown primary (1%–18%).
Melanoma (4%–16%).
Colorectal (2%–12%).
Kidney (1%–8%).
Eighty percent of brain metastases occur in the cerebral hemispheres, 15% occur in the cerebellum, and 5% occur in the brain stem.[2] Metastases to the brain are multiple in more than 70% of cases, but solitary metastases also occur.[1]
Brain involvement can occur with cancers of the nasopharyngeal region by direct extension along the cranial nerves or through the foramina at the base of the skull. Dural metastases may constitute as much as 9% of total brain metastases.
Clinical Features
The diagnosis of brain metastases in cancer patients is based on the following:
Patient history.
Neurological examination.
Diagnostic procedures, including a contrast MRI of the brain.
Patients may describe any of the following:
Headaches.
Weakness.
Seizures.
Sensory defects.
Gait problems.
Often, family members or friends may notice the following:
Lethargy.
Emotional lability.
Personality change.
Diagnostic Evaluation
A physical examination may show objective neurological findings or only minor cognitive changes. The presence of multiple lesions and a high predilection of primary tumor metastasis may be sufficient to make the diagnosis of brain metastasis.
A lesion in the brain should not be assumed to be a metastasis just because a patient has had a previous cancer; such an assumption could result in overlooking appropriate treatment of a curable tumor.
Imaging tests
Computed tomography scans with contrast or MRIs with gadolinium are quite sensitive in diagnosing the presence of metastases. Positron emission tomography scanning and spectroscopic evaluation are new strategies to diagnose cerebral metastases and to differentiate the metastases from other intracranial lesions.[4]
Biopsy
In the case of a solitary lesion or a questionable relationship to the primary tumor, a brain biopsy (via resection or stereotactic biopsy) may be necessary.
Treatment of Metastatic Brain Tumors
The optimal therapy for patients with brain metastases continues to evolve.[1,2,5] The following treatments have been used in the management of metastatic brain tumors:
Radiation therapy.
Radiosurgery.
Surgical resection.
Corticosteroids.
Anticonvulsants.
Because most cases of brain metastases involve multiple metastases, a mainstay of therapy has historically been whole-brain radiation therapy (WBRT). However, stereotactic radiosurgery has become increasingly common. The role of radiosurgery continues to be defined. Stereotactic radiosurgery in combination with WBRT has been assessed.
Surgery is indicated to obtain tissue from a metastasis with an unknown primary tumor or to decompress a symptomatic dominant lesion that is causing significant mass effect.
Chemotherapy is usually not the primary therapy for most patients; however, it may have a role in the treatment of patients with brain metastases from chemosensitive tumors and can even be curative when combined with radiation for metastatic testicular germ cell tumors.[1,6] Intrathecal chemotherapy is also used for meningeal spread of metastatic tumors.
Treatment for patients with one to four metastases
Treatment options for patients with one to four metastases
About 10% to 15% of patients with cancer will have a single brain metastasis. Radiation therapy is the mainstay of palliation for these patients. The extent of extracranial disease can influence treatment of the brain lesions. In the presence of extensive active systemic disease, surgery provides little benefit for overall survival (OS). In patients with stable minimal extracranial disease, combined modality treatment may be considered, using surgical resection followed by radiation therapy. However, the published literature does not provide clear guidance.
Treatment options for patients with one to four metastases include the following:
WBRT with or without surgical resection.
WBRT with or without stereotactic radiosurgery.
Focal therapy alone (surgical resection or stereotactic radiosurgery).
Evidence (treatment for one to four metastases):
Three randomized trials examined resection of solitary brain metastases followed by WBRT versus WBRT alone, totaling 195 randomly assigned patients.[7–9] The process that necessarily goes into selecting appropriate patients for surgical resection may account for the small numbers in each trial. In the first trial,[7][Level of evidence B1] performed at a single center, all patients were selected and operated upon by one surgeon.
The first two trials showed an improvement in survival in the surgery group,[7,8] but the third trial showed a trend in favor of the WBRT-only group.[9]
The three trials were combined in a trial-level meta-analysis.[10] The combined analysis showed the following:
The combined analysis did not show a statistically significant difference in OS (hazard ratio [HR], 0.72; 95% confidence interval [CI], 0.34–1.53; P = .4); or in death from neurological causes (relative riskdeath, 0.68; 95% CI, 0.43–1.09; P = .11).[10]
One of the trials reported that combined therapy increased the duration of functionally independent survival.[7][Level of evidence B1]
None of the trials assessed or reported quality of life.
The need for WBRT after resection of solitary brain metastases has been studied.[11] Patients were randomly assigned to either undergo postoperative WBRT or receive no further treatment after resection.
Patients in the WBRT group were less likely to have tumor progression in the brain and were significantly less likely to die of neurological causes.
OS was the same in each group, and there was no difference in duration of functional independence.
One additional randomized study of observation versus WBRT after either surgery or stereotactic radiosurgery for solitary brain metastases was closed after 19 patients had been entered because of slow accrual; therefore, little can be deduced from the trial.[12]
A Radiation Therapy Oncology Group (RTOG) study (RTOG-9508) randomly assigned 333 patients with one to three metastases with a maximum diameter of 4 cm to WBRT (37.5 Gy over 3 weeks) with or without a stereotactic boost.[13] Patients with active systemic disease requiring therapy were excluded. The primary end point was OS with predefined hypotheses in both the full study population and the 186 patients with a solitary metastasis (and no statistical adjustment of P values for the two separate hypotheses).[13][Levels of evidence B1 for the full study population and A1 for patients with solitary metastases]
Mean OS in the combined-therapy group was 5.7 months, and mean OS in the WBRT–alone group was 6.5 months (P = .14).
In the subgroup with solitary metastases, OS was better in the combined-therapy group (6.5 months vs. 4.9 months; P = .039 in univariate analysis; P = .053 in a multivariable analysis adjusting for baseline prognostic factors).
In patients with multiple metastases, survival was 5.8 months in the combined-therapy group versus 6.7 months in the WBRT–only group (P = .98).
The combined-treatment group had a survival advantage of 2.5 months in patients with a single metastasis but not in patients with multiple lesions.
Local control was better in the full population with combined therapy.
At the 6-month follow-up, Karnofsky Performance status (considered a soft end point because of its imprecision and subjectivity) was better in the combined-therapy group, but there was no difference in mental status between the treatment groups. Acute and late toxicities were similar in both treatment arms. Quality of life was not assessed.
A phase III randomized trial compared adjuvant WBRT with observation after surgery or radiosurgery for a limited number of brain metastases in patients with stable solid tumors.[14][Level of evidence A3]
Health-related quality of life was improved in the observation-only arm, compared with WBRT.
Patients in the observation arm had better mean scores in physical, role, and cognitive functioning at 9 months.
In an exploratory analysis, statistically significant worse scores for bladder control, communication deficit, drowsiness, hair loss, motor dysfunction, leg weakness, appetite loss, constipation, nausea/vomiting, pain, and social functioning were observed in patients who underwent WBRT, compared with those who underwent observation only.
A meta-analysis of two trials with a total of 358 participants found no statistically significant difference in OS between the WBRT plus stereotactic radiosurgery group and the WBRT-alone group (HR, 0.82; 95% CI, 0.65–1.02).[15][Level of evidence B1]
Patients in the WBRT plus stereotactic radiosurgery group had decreased local failure, compared with patients who received WBRT alone (HR, 0.27; 95% CI, 0.14–0.52).
Unchanged or improved Karnofsky Performance status at 6 months was seen in 43% of patients in the combined-therapy group versus 28% in the WBRT-alone group (P = .03).
A study that had a primary end point of learning and neurocognition, using a standardized test for total recall, was stopped by the Data and Safety Monitoring Board because of worse outcomes in the WBRT group.[16][Level of evidence B1]
Given this body of information, focal therapy plus WBRT or focal therapy alone, with close follow-up with serial MRIs and initiation of salvage therapy when clinically indicated, appear to be reasonable treatment options. The pros and cons of each approach should be discussed with the patient.
Several randomized trials have been performed that were designed with varying primary end points to address whether WBRT is necessary after focal treatment. The results can be summarized as follows:[16–18]
Studies consistently show that the addition of WBRT to focal therapy decreases the risk of progression and new metastases in the brain.
The addition of WBRT does not improve OS.
The decrease in risk of intracranial disease progression does not translate into improved functional or neurological status, nor does it appear to decrease the risk of death from neurological deterioration.
About one-half or more of the patients who receive focal therapy alone ultimately require salvage therapy, such as WBRT or radiosurgery, compared with about one-quarter of the patients who are given up-front WBRT.
The impact of better local control associated with WBRT on quality of life has not been reported and remains an open question.
Leptomeningeal Carcinomatosis (LC)
LC occurs in about 5% of all cancer patients. The most common types of cancer to spread to the leptomeninges are:
Breast tumors (35%).
Lung tumors (24%).
Hematologic malignancies (16%).
Diagnosis includes a combination of neurospinal axis imaging and cerebrospinal fluid (CSF) cytology. Median OS is in the range of 10 to 12 weeks.
The management of LC includes the following:
Intrathecal chemotherapy.
Intrathecal chemotherapy and systemic chemotherapy.
Intrathecal chemotherapy and radiation therapy.
Supportive care.
In a series of 149 patients with metastatic non-small cell lung carcinoma, cytologically proven LC, poor performance status, high protein level in the CSF, and a high initial CSF white blood cell count were significant poor prognostic factors for survival.[19] Patients received active treatment, including intrathecal chemotherapy, WBRT, or epidermal growth factor receptor-tyrosine kinase inhibitors, or underwent a ventriculoperitoneal shunt procedure.
In a retrospective series of 38 patients with metastatic breast cancer and LC, the proportion of LC cases varied by breast cancer subtype:[20]
Luminal A (18.4%).
Luminal B (31.6%).
Human epidermal growth factor receptor 2 (HER2) positive (26.3%).
Triple-negative breast cancer subtype (23.7%).
Patients with triple-negative breast cancer had a shorter interval between metastatic breast cancer diagnosis and the development of LC. Median survival did not differ across breast cancer subtypes. Consideration of intrathecal administration of trastuzumab in patients with HER2-positive LC has also been described in case reports.[21]
References
Patchell RA: The management of brain metastases. Cancer Treat Rev 29 (6): 533-40, 2003. [PUBMED Abstract]
Mehta M, Vogelbaum MA, Chang S, et al.: Neoplasms of the central nervous system. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 1700-49.
Hutter A, Schwetye KE, Bierhals AJ, et al.: Brain neoplasms: epidemiology, diagnosis, and prospects for cost-effective imaging. Neuroimaging Clin N Am 13 (2): 237-50, x-xi, 2003. [PUBMED Abstract]
Schaefer PW, Budzik RF, Gonzalez RG: Imaging of cerebral metastases. Neurosurg Clin N Am 7 (3): 393-423, 1996. [PUBMED Abstract]
Soffietti R, Cornu P, Delattre JY, et al.: EFNS Guidelines on diagnosis and treatment of brain metastases: report of an EFNS Task Force. Eur J Neurol 13 (7): 674-81, 2006. [PUBMED Abstract]
Ogawa K, Yoshii Y, Nishimaki T, et al.: Treatment and prognosis of brain metastases from breast cancer. J Neurooncol 86 (2): 231-8, 2008. [PUBMED Abstract]
Patchell RA, Tibbs PA, Walsh JW, et al.: A randomized trial of surgery in the treatment of single metastases to the brain. N Engl J Med 322 (8): 494-500, 1990. [PUBMED Abstract]
Vecht CJ, Haaxma-Reiche H, Noordijk EM, et al.: Treatment of single brain metastasis: radiotherapy alone or combined with neurosurgery? Ann Neurol 33 (6): 583-90, 1993. [PUBMED Abstract]
Mintz AH, Kestle J, Rathbone MP, et al.: A randomized trial to assess the efficacy of surgery in addition to radiotherapy in patients with a single cerebral metastasis. Cancer 78 (7): 1470-6, 1996. [PUBMED Abstract]
Hart MG, Grant R, Garside R, et al.: Chemotherapeutic wafers for high grade glioma. Cochrane Database Syst Rev (3): CD007294, 2008. [PUBMED Abstract]
Patchell RA, Tibbs PA, Regine WF, et al.: Postoperative radiotherapy in the treatment of single metastases to the brain: a randomized trial. JAMA 280 (17): 1485-9, 1998. [PUBMED Abstract]
Roos DE, Wirth A, Burmeister BH, et al.: Whole brain irradiation following surgery or radiosurgery for solitary brain metastases: mature results of a prematurely closed randomized Trans-Tasman Radiation Oncology Group trial (TROG 98.05). Radiother Oncol 80 (3): 318-22, 2006. [PUBMED Abstract]
Andrews DW, Scott CB, Sperduto PW, et al.: Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet 363 (9422): 1665-72, 2004. [PUBMED Abstract]
Soffietti R, Kocher M, Abacioglu UM, et al.: A European Organisation for Research and Treatment of Cancer phase III trial of adjuvant whole-brain radiotherapy versus observation in patients with one to three brain metastases from solid tumors after surgical resection or radiosurgery: quality-of-life results. J Clin Oncol 31 (1): 65-72, 2013. [PUBMED Abstract]
Patil CG, Pricola K, Sarmiento JM, et al.: Whole brain radiation therapy (WBRT) alone versus WBRT and radiosurgery for the treatment of brain metastases. Cochrane Database Syst Rev 9: CD006121, 2012. [PUBMED Abstract]
Chang EL, Wefel JS, Hess KR, et al.: Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled trial. Lancet Oncol 10 (11): 1037-44, 2009. [PUBMED Abstract]
Aoyama H, Shirato H, Tago M, et al.: Stereotactic radiosurgery plus whole-brain radiation therapy vs stereotactic radiosurgery alone for treatment of brain metastases: a randomized controlled trial. JAMA 295 (21): 2483-91, 2006. [PUBMED Abstract]
Kocher M, Soffietti R, Abacioglu U, et al.: Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 study. J Clin Oncol 29 (2): 134-41, 2011. [PUBMED Abstract]
Lee SJ, Lee JI, Nam DH, et al.: Leptomeningeal carcinomatosis in non-small-cell lung cancer patients: impact on survival and correlated prognostic factors. J Thorac Oncol 8 (2): 185-91, 2013. [PUBMED Abstract]
Torrejón D, Oliveira M, Cortes J, et al.: Implication of breast cancer phenotype for patients with leptomeningeal carcinomatosis. Breast 22 (1): 19-23, 2013. [PUBMED Abstract]
Bartsch R, Berghoff AS, Preusser M: Optimal management of brain metastases from breast cancer. Issues and considerations. CNS Drugs 27 (2): 121-34, 2013. [PUBMED Abstract]
Treatment of Recurrent Adult CNS Tumors
Patients who have recurrent CNS tumors are rarely curable and should consider enrollment in clinical trials. Information about ongoing clinical trials is available from the NCI website.
Treatment options for recurrent CNS tumors include the following:
Carmustine wafers have been investigated for the treatment of recurrent malignant gliomas, but the impact on survival is less clear than at the time of initial diagnosis and resection.
Evidence (localized chemotherapy):
In a multicenter, randomized, placebo-controlled trial, 222 patients with recurrent malignant primary brain tumors requiring reoperation were randomly assigned to receive implanted carmustine wafers or placebo biodegradable wafers.[1][Level of evidence A1] Approximately one-half of the patients had received previous systemic chemotherapy. The two treatment groups were well balanced at baseline.
Median survival was 31 weeks in the group receiving carmustine wafers versus 23 weeks in the group receiving placebo wafers. The statistical significance between the two overall survival curves depended on the method of analysis.
The hazard ratio (HR) for risk of dying in the direct intention-to-treat comparison between the two groups was 0.83 (95% confidence interval [CI], 0.63–1.10; P = .19). The baseline characteristics were similar in the two groups, but the investigators performed an additional analysis, adjusting for prognostic factors, because they felt that even small differences in baseline characteristics could have a powerful influence on outcomes. In the adjusted proportional hazards model, the HR for risk of death was 0.67 (95% CI, 0.51–0.90; P = .006). The investigators emphasized this latter analysis and reported this as a positive trial.[1][Level of evidence A1]
A Cochrane Collaboration systematic review of chemotherapeutic wafers for high-grade glioma focused on the unadjusted analysis and reported the same trial as negative.[2]
Systemic chemotherapy
Systemic therapy (e.g., temozolomide, lomustine, or the combination of procarbazine, a nitrosourea, and vincristine [PCV] in patients who have not previously received the drugs) has been used at the time of recurrence of primary malignant brain tumors. However, this approach has not been tested in controlled studies. Patient-selection factors likely play a strong role in determining outcomes, so the impact of therapy on survival is not clear.
Antiangiogenesis Therapy
In 2009, the U.S. Food and Drug Administration (FDA) granted accelerated approval of bevacizumab monotherapy for patients with progressive glioblastoma. The indication was granted under the FDA’s accelerated approval program that permits the use of certain surrogate end points or an effect on a clinical end point other than survival or irreversible morbidity as bases for approvals of products intended for serious or life-threatening illnesses or conditions.
The approval was based on the demonstration of improved objective response rates observed in two historically controlled, single-arm, or noncomparative phase II trials.[3,4][Level of evidence C3] Based on these data and the FDA approval, bevacizumab monotherapy has become standard therapy for recurrent glioblastoma.
Evidence (antiangiogenesis therapy):
The FDA independently reviewed an open-label, multicenter, noncomparative phase II study that randomly assigned 167 patients with recurrent glioblastoma multiforme (GBM) to receive bevacizumab alone or bevacizumab in combination with irinotecan.[3] However, only efficacy data from the bevacizumab monotherapy arm (n = 85) were used to support drug approval.
Tumor responses were observed in 26% of patients treated with bevacizumab alone, and the median duration of response in these patients was 4.2 months.
Based on this externally controlled trial, the incidence of adverse events associated with bevacizumab did not appear to be significantly increased in GBM patients.
The FDA independently assessed another single-arm, single-institution trial in which 56 patients with recurrent glioblastoma were treated with bevacizumab alone.[4]
Responses were observed in 20% of patients, and the median duration of response was 3.9 months.
No data are available from prospective randomized controlled trials demonstrating improvement in health outcomes, such as disease-related symptoms or increased survival with the use of bevacizumab to treat glioblastoma.
Radiation Therapy
Because there are no randomized trials, the role of repeat radiation after disease progression or the development of radiation-induced cancers is also ill defined. Interpretation is difficult because the literature is limited to small retrospective case series.[5] The decision must be made carefully because of the risk of neurocognitive deficits and radiation necrosis.
Surgery
Re-resection of recurrent CNS tumors is an option for some patients. However, most patients do not qualify because of a deteriorating condition or technically inoperable tumors. The evidence is limited to noncontrolled studies and case series of patients who are healthy enough and have tumors that are small enough to technically debulk. The impact on survival of reoperation versus patient selection is not known.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Brem H, Piantadosi S, Burger PC, et al.: Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group. Lancet 345 (8956): 1008-12, 1995. [PUBMED Abstract]
Hart MG, Grant R, Garside R, et al.: Chemotherapeutic wafers for high grade glioma. Cochrane Database Syst Rev (3): CD007294, 2008. [PUBMED Abstract]
Friedman HS, Prados MD, Wen PY, et al.: Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27 (28): 4733-40, 2009. [PUBMED Abstract]
Kreisl TN, Kim L, Moore K, et al.: Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 27 (5): 740-5, 2009. [PUBMED Abstract]
Paulino AC, Mai WY, Chintagumpala M, et al.: Radiation-induced malignant gliomas: is there a role for reirradiation? Int J Radiat Oncol Biol Phys 71 (5): 1381-7, 2008. [PUBMED Abstract]
Latest Updates to This Summary (02/12/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 2).
Updated statistics about incidence and mortality rates for the United States. Also updated statistics with worldwide cases and deaths (cited Bray et al. as reference 4).
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of adult central nervous system tumors. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Adult Central Nervous System Tumors Treatment are:
Solmaz Sahebjam, MD ()
Minh Tam Truong, MD (Boston University Medical Center)
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Adult Central Nervous System Tumors Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/brain/hp/adult-brain-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389419]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
We offer evidence-based supportive and palliative care information for health professionals on the assessment and management of cancer-related symptoms and conditions.
Brain and spinal cord (also known as central nervous system, or CNS) tumors can be benign or malignant. Explore the links on this page to learn more about the many different CNS tumor types and how they are treated. We also have information about brain cancer statistics, research, and clinical trials.
We offer evidence-based supportive and palliative care information for health professionals on the assessment and management of cancer-related symptoms and conditions.
Cancerprevention is action taken to lower the chance of getting cancer. By preventing cancer, the number of new cases of cancer in a group or population is lowered. Hopefully, this will lower the number of deaths caused by cancer.
To prevent new cancers from starting, scientists look at risk factors and protective factors. Anything that increases your chance of developing cancer is called a cancer risk factor; anything that decreases your chance of developing cancer is called a cancer protective factor.
Some risk factors for cancer can be avoided, but many cannot. For example, both smoking and inheriting certain genes are risk factors for some types of cancer, but only smoking can be avoided. Regular exercise and a healthy diet may be protective factors for some types of cancer. Avoiding risk factors and increasing protective factors may lower your risk, but it does not mean that you will not get cancer.
Different ways to prevent cancer are being studied, including:
Anal cancer is a disease in which malignant (cancer) cells form in the tissues of the anus.
Squamous cell carcinoma is the most common type of anal cancer.
In the United States, the number of new cases of anal cancer has increased in recent years.
Anal cancer is a disease in which malignant (cancer) cells form in the tissues of the anus.
The anus is the end of the large intestine, below the rectum. It is where stool (solid waste) leaves the body. The anus is formed partly from the outer skin layers of the body and partly from the intestine. The anus is connected to the rectum by the anal canal, which is about 1–1½ inches long. This area is controlled by two ring-like sphincter muscles, which open and close to let stool pass out of the body.
EnlargeAnatomy of the lower gastrointestinal (digestive) system showing the colon, rectum, and anus. Other organs that make up the digestive system are also shown.
The skin around the outside of the anus is called the perianal area. Tumors in this area are skin tumors, not anal cancer.
Another type of anal cancer, called anal adenocarcinoma, is very rare and is not discussed in this summary.
In the United States, the number of new cases of anal cancer has increased in recent years.
Between 2012 and 2021, the number of new cases of anal cancer increased each year. Between 2013 and 2022, the rate of deaths from this disease also increased each year.
Anal Cancer Prevention
Key Points
Avoiding risk factors and increasing protective factors may help prevent cancer.
The following are risk factors for anal cancer:
Anal HPV infection
Certain medical conditions
History of cervical, vaginal, or vulvar cancer
HIV infection/AIDS
Immunosuppression
Certain sexual practices
Cigarette smoking
The following protective factors decrease the risk of anal cancer:
HPV vaccination
Screening for anal cancer using anoscopy
Treatment of anal lesions
It is not clear if the following protective factor decreases the risk of anal cancer:
Condom use
Cancer prevention clinical trials are used to study ways to prevent cancer.
New ways to prevent anal cancer are being studied in clinical trials.
Avoiding risk factors and increasing protective factors may help prevent cancer.
Avoiding cancerrisk factors may help prevent certain cancers. Risk factors include smoking, having overweight, and not getting enough exercise. Increasing protective factors such as quitting smoking and exercising may also help prevent some cancers. Talk to your doctor or other health care professional about how you might lower your risk of cancer.
People with healthy immune systems are usually able to fight HPV infections. People with weakened immune systems who are infected with HPV have a higher risk of anal cancer.
Certain medical conditions
History of cervical, vaginal, or vulvar cancer
Cervical cancer, vaginal cancer, and vulvar cancer are related to HPV infection. Women who have had cervical, vaginal, or vulvar cancer have a higher risk of anal cancer.
HIV infection/AIDS
Being infected with HIV is a strong risk factor for anal cancer. HIV is the cause of AIDS. HIV weakens the body’s immune system and its ability to fight infection. HPV infection of the anus is common among people who are HIV positive.
The risk of anal cancer is higher in men who are HIV positive and have sex with men compared with men who are HIV negative and have sex with men. Women who are HIV positive also have an increased risk of anal cancer compared with women who are HIV negative.
Studies show that intravenousdrug use or cigarette smoking may further increase the risk of anal cancer in people who are HIV positive.
Immunosuppression
Immunosuppression is a condition that weakens the body’s immune system and its ability to fight infections and other diseases. Chronic (long-term) immunosuppression may increase the risk of anal cancer because it lowers the body’s ability to fight HPV infection.
Having an autoimmune disorder, such as Crohn disease or psoriasis, may increase the risk of anal cancer. It is not clear if the increased risk is due to the autoimmune condition, the treatment for the condition, or a combination of both.
Certain sexual practices
The following sexual practices increase the risk of anal cancer because they increase the chance of being infected with HPV:
Studies show that cigarette smoking increases the risk of anal cancer. Studies also show that current smokers have a higher risk of anal cancer than smokers who have quit or people who have never smoked.
The following protective factors decrease the risk of anal cancer:
HPV vaccination
HPV vaccines help protect the body against infection with certain types of HPV. They are used to prevent anal cancer, cervical cancer, vulvar cancer, vaginal cancer, and some other types of cancer caused by HPV. They are also used to prevent abnormallesions caused by HPV that may lead to some of these cancers.
Studies show that being vaccinated against HPV lowers the risk of anal cancer. The vaccine may work best when it is given before a person is exposed to HPV.
Screening for anal cancer using anoscopy
Cancer screening is looking for cancer before a person has any symptoms. This can help find cancer at an early stage. When abnormal tissue or cancer is found early, it may be easier to treat.
An anoscopy is a procedure that uses a short, lighted tube called an anoscope to check for abnormal areas in the anus and lower rectum.
Studies suggest that using anoscopy to screen for anal cancer could reduce the number of deaths from the disease in people who are HIV positive.
It is not clear if the following protective factor decreases the risk of anal cancer:
Condom use
It is not known if the use of condoms protects against anal HPV infection. This is because not enough studies have been done to prove this.
Cancer prevention clinical trials are used to study ways to prevent cancer.
Cancer prevention clinical trials are used to study ways to lower the risk of developing certain types of cancer. Some cancer prevention trials include healthy people who may or may not have an increased risk of cancer. Other prevention trials include people who have had cancer and are trying to prevent recurrence or a second cancer.
The purpose of some cancer prevention clinical trials is to find out whether actions people take can prevent cancer. These may include eating fruits and vegetables, exercising, quitting smoking, or taking certain medicines, vitamins, minerals, or food supplements.
New ways to prevent anal cancer are being studied in clinical trials.
Physician Data Query (PDQ) is the National Cancer Institute’s (NCI’s) comprehensive cancer information database. The PDQ database contains summaries of the latest published information on cancer prevention, detection, genetics, treatment, supportive care, and complementary and alternative medicine. Most summaries come in two versions. The health professional versions have detailed information written in technical language. The patient versions are written in easy-to-understand, nontechnical language. Both versions have cancer information that is accurate and up to date and most versions are also available in Spanish.
PDQ is a service of the NCI. The NCI is part of the National Institutes of Health (NIH). NIH is the federal government’s center of biomedical research. The PDQ summaries are based on an independent review of the medical literature. They are not policy statements of the NCI or the NIH.
Purpose of This Summary
This PDQ cancer information summary has current information about anal cancer prevention. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.
Reviewers and Updates
Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.
The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Screening and Prevention Editorial Board.
Clinical Trial Information
A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).
Permission to Use This Summary
PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”
The best way to cite this PDQ summary is:
PDQ® Screening and Prevention Editorial Board. PDQ Anal Cancer Prevention. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/anal/patient/anal-prevention-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389512]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use in the PDQ summaries only. If you want to use an image from a PDQ summary and you are not using the whole summary, you must get permission from the owner. It cannot be given by the National Cancer Institute. Information about using the images in this summary, along with many other images related to cancer can be found in Visuals Online. Visuals Online is a collection of more than 3,000 scientific images.
Disclaimer
The information in these summaries should not be used to make decisions about insurance reimbursement. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s E-mail Us.
Anal cancer is a type of cancer that forms in the tissues of the anus.
Most anal cancers are related to human papillomavirus (HPV) infection.
Signs of anal cancer include bleeding from the anus or rectum or a lump near the anus.
Tests that examine the rectum and anus are used to diagnose anal cancer.
After anal cancer has been diagnosed, tests are done to find out if cancer cells have spread within the anus or to other parts of the body.
Some people decide to get a second opinion.
Certain factors affect the prognosis (chance of recovery) and treatment options.
Anal cancer is a type of cancer that forms in the tissues of the anus.
The anus is the end of the large intestine. It is where stool (solid waste) leaves the body. The anus is formed partly from the outer skin layers of the body and partly from the intestine. The anus is connected to the rectum by the anal canal, which is about 1 to 1½ inches long. This area is controlled by two ring-like sphincter muscles, which contract to hold stool in and relax to allow its passage out of the body.
EnlargeAnatomy of the lower gastrointestinal (digestive) system showing the colon, rectum, and anus. Other organs that make up the digestive system are also shown.
Anal cancer can start in the lining of the anal canal, called the mucosa, or in the perianal skin, the squamous cells outside of the anus that contain hair follicles and sweat glands.
Tumors of the perianal skin that do not involve the anal sphincter are usually treated the same as anal cancers, although local therapy (treatment directed to a limited area of skin) may be used for some.
Most anal cancers are related to human papillomavirus (HPV) infection.
Anal cancer is caused by certain changes to the way anal cells function, especially how they grow and divide into new cells. There are many risk factors for anal cancer, but many do not directly cause cancer. Instead, they increase the chance of DNA damage in cells that may lead to anal cancer. Learn more about how cancer develops at What Is Cancer?
A risk factor is anything that increases the chance of getting a disease. Some risk factors for anal cancer can be changed. However, risk factors also include things people cannot change, like getting older and their health history. Learning about risk factors for anal cancer can help you make changes that might lower your risk of getting it.
Digital rectal examination (DRE) is an exam of the anus and rectum. The doctor or nurse inserts a lubricated, gloved finger into the lower part of the rectum to feel for lumps or anything else that seems unusual. EnlargeDigital rectal exam (DRE). The doctor inserts a gloved, lubricated finger into the rectum and feels the rectum, anus, and prostate (in males) to check for anything abnormal.
Anoscopy is an exam of the anus and lower rectum using a short, lighted tube called an anoscope.
Proctoscopy is a procedure to look inside the rectum and anus to check for abnormal areas, using a proctoscope. A proctoscope is a thin, tube-like instrument with a light and a lens for viewing the inside of the rectum and anus. It may also have a tool to remove tissue samples, which are checked under a microscope for signs of cancer.
Endo-anal or endorectal ultrasound is a procedure in which an ultrasound transducer (probe) is inserted into the anus or rectum and used to bounce high-energy sound waves (ultrasound) off internal tissues or organs and make echoes. The echoes form a picture of body tissues called a sonogram.
Biopsy is the removal of cells or tissues so they can be viewed under a microscope by a pathologist to check for signs of cancer. If an abnormal area is seen during the anoscopy, a biopsy may be done at that time.
After anal cancer has been diagnosed, tests are done to find out if cancer cells have spread within the anus or to other parts of the body.
The process used to find out if cancer has spread within the anus or to other parts of the body is called staging. The information gathered from this staging process determines the stage of the disease. It is important to know the stage in order to plan treatment. The following tests may be used in the staging process:
CT scan (CAT scan) uses a computer linked to an x-ray machine to make a series of detailed pictures of areas inside the body, such as the abdomen, pelvis, or chest. The pictures are taken from different angles and are used to create 3-D views of tissues and organs. A dye may be injected into a vein or swallowed to help the organs or tissues show up more clearly. This procedure is also called computed tomography, computerized tomography, or computerized axial tomography.
Chest x-ray is a type of radiation that can go through the body and make pictures of the organs and bones inside the chest.
MRI (magnetic resonance imaging) uses a magnet, radio waves, and a computer to make a series of detailed pictures of areas inside the body. This procedure is also called nuclear magnetic resonance imaging (NMRI).
PET scan (positron emission tomography scan) uses a small amount of radioactive sugar (also called radioactive glucose) that is injected into a vein. The PET scanner rotates around the body and makes pictures of where glucose is being used in the body. Cancer cells show up brighter in the picture because they are more active and take up more glucose than normal cells do.
Pelvic exam is an exam of the vagina, cervix, uterus, fallopian tubes, ovaries, and rectum. A speculum is inserted into the vagina and the doctor or nurse looks at the vagina and cervix for signs of disease. A Pap test of the cervix is usually done. The doctor or nurse also inserts one or two lubricated, gloved fingers of one hand into the vagina and places the other hand over the lower abdomen to feel the size, shape, and position of the uterus and ovaries. The doctor or nurse also inserts a lubricated, gloved finger into the rectum to feel for lumps or abnormal areas. EnlargePelvic exam. A doctor or nurse inserts one or two lubricated, gloved fingers of one hand into the vagina and presses on the lower abdomen with the other hand. This is done to feel the size, shape, and position of the uterus and ovaries. The vagina, cervix, fallopian tubes, and rectum are also checked.
Some people decide to get a second opinion.
You may want to get a second opinion to confirm your anal cancer diagnosis and treatment plan. If you seek a second opinion, you will need to get medical test results and reports from the first doctor to share with the second doctor. The second doctor will review the pathology report, slides, and scans. They may agree with the first doctor, suggest changes or another treatment approach, or provide more information about your cancer.
whether cancer remains after initial treatment or has recurred (come back)
Stages of Anal Cancer
Key Points
The following stages are used for anal cancer:
Stage 0 (carcinoma in situ)
Stage I (also called stage 1) anal cancer
Stage II (also called stage 2) anal cancer
Stage III (also called stage 3) anal cancer
Stage IV (also called stage 4) anal cancer
Anal cancer can recur (come back) after it has been treated.
Cancer stage describes the extent of cancer in the body, such as the size of the tumor, whether it has spread, and how far it has spread from where it first formed. It is important to know the stage of the anal cancer to plan the best treatment.
There are several staging systems for cancer that describe the extent of the cancer. Anal cancer staging usually uses the TNM staging system. The cancer may be described by this staging system in your pathology report. Based on the TNM results, a stage (I, II, III, or IV, also written as 1, 2, 3, or 4) is assigned to your cancer. When talking to you about your diagnosis, your doctor may describe the cancer as one of these stages.
In stage 0, abnormal cells are found in the mucosa (innermost layer) of the anus. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called high-grade intraepithelial lesion (HSIL).
EnlargeTumor sizes are often measured in centimeters (cm) or inches. Common food items that can be used to show tumor size in cm include: a pea (1 cm), a peanut (2 cm), a grape (3 cm), a walnut (4 cm), a lime (5 cm or 2 inches), an egg (6 cm), a peach (7 cm), and a grapefruit (10 cm or 4 inches).
Stage I (also called stage 1) anal cancer
In stage I, cancer has formed and the tumor is 2 centimeters or smaller.
Stage II (also called stage 2) anal cancer
Stage II anal cancer is divided into stages IIA and IIB.
In stage IIA, the tumor is larger than 2 centimeters but not larger than 5 centimeters.
In stage IIB, the tumor is larger than 5 centimeters.
Stage III (also called stage 3) anal cancer
Stage III anal cancer is divided into stages IIIA, IIIB, and IIIC.
In stage IIIB, the tumor is any size and has spread to nearby organs, such as the vagina, urethra, or bladder. Cancer has not spread to lymph nodes.
In stage IIIC, the tumor is any size and may have spread to nearby organs. Cancer has spread to lymph nodes near the anus or groin.
Stage IV (also called stage 4) anal cancer
In stage IV, the tumor is any size. Cancer may have spread to lymph nodes or nearby organs and has spread to other parts of the body, such as the liver or lungs.
Stage IV anal cancer is also called metastatic anal cancer. Metastatic cancer happens when cancer cells travel through the lymphatic system or blood and form tumors in other parts of the body. The metastatic tumor is the same type of cancer as the primary tumor. For example, if anal cancer spreads to the liver, the cancer cells in the liver are actually anal cancer cells. The disease is called metastatic anal cancer, not liver cancer. Learn more in Metastatic Cancer: When Cancer Spreads.
Anal cancer can recur (come back) after it has been treated.
Recurrent anal cancer is cancer that has come back after it has been treated. If anal cancer comes back, it may come back in the anus or in other parts of the body, such as the liver or lungs. Tests will be done to help determine where the cancer has returned. The type of treatment for recurrent anal cancer will depend on where it has come back.
There are different types of treatment for people with anal cancer.
The following types of treatment are used:
Surgery
Radiation therapy
Chemotherapy
New types of treatment are being tested in clinical trials.
Treatment for anal cancer may cause side effects.
Follow-up care may be needed.
There are different types of treatment for people with anal cancer.
Different types of treatments are available for anal cancer. You and your cancer care team will work together to decide your treatment plan, which may include more than one type of treatment. Many factors will be considered, such as the stage of the cancer, your overall health, and your preferences. Your plan will include information about your cancer, the goals of treatment, your treatment options and the possible side effects, and the expected length of treatment.
Talking with your cancer care team before treatment begins about what to expect will be helpful. You’ll want to learn what you need to do before treatment begins, how you’ll feel while going through it, and what kind of help you will need. To learn more, visit Questions to Ask Your Doctor About Treatment.
The following types of treatment are used:
Surgery
Local resection is a surgical procedure in which the tumor is cut from the anus along with some of the healthy tissue around it. Local resection may be used if the cancer is small and has not spread. This procedure may save the sphincter muscles so the person can still control bowel movements. Tumors that form in the lower part of the anus can often be removed with local resection.
Abdominoperineal resection is a surgical procedure in which the anus, the rectum, and part of the sigmoid colon are removed through an incision made in the abdomen. The doctor sews the end of the intestine to an opening, called a stoma, made in the surface of the abdomen so body waste can be collected in a disposable bag outside of the body. This is called a colostomy. Lymph nodes that contain cancer may also be removed during this operation. This procedure is used only for cancer that remains or comes back after treatment with radiation therapy and chemotherapy. EnlargeResection of the colon with colostomy. Part of the colon containing the cancer and nearby healthy tissue are removed, a stoma is created, and a colostomy bag is attached to the stoma.
Radiation therapy
Radiation therapy uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. There are two types of radiation therapy used to treat anal cancer:
External radiation therapy uses a machine outside the body to send radiation toward the area of the body with cancer.
Chemotherapy (also called chemo) uses drugs to stop the growth of cancer cells, either by killing the cells or by stopping the cells from dividing. When chemotherapy is taken by mouth or injected into a vein or muscle, the drugs enter the bloodstream and can reach cancer cells throughout the body (systemic chemotherapy).
Chemotherapy drugs used to treat anal cancer include:
New types of treatment are being tested in clinical trials.
For some people, joining a clinical trial may be an option. There are different types of clinical trials for people with cancer. For example, a treatment trial tests new treatments or new ways of using current treatments. Supportive care and palliative care trials look at ways to improve quality of life, especially for those who have side effects from cancer and its treatment.
You can use the clinical trial search to find NCI-supported cancer clinical trials accepting participants. The search allows you to filter trials based on the type of cancer, your age, and where the trials are being done. Clinical trials supported by other organizations can be found on the ClinicalTrials.gov website.
As you go through treatment, you will have follow-up tests or check-ups. Some tests that were done to diagnose or stage the cancer may be repeated to see how well the treatment is working. Decisions about whether to continue, change, or stop treatment may be based on the results of these tests.
Some of the tests will continue to be done from time to time after treatment has ended. The results of these tests can show if your condition has changed or if the cancer has recurred (come back).
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
abdominoperineal resection, if cancer remains or comes back after treatment with radiation therapy and chemotherapy, or other options that may include treatment with additional chemoradiation therapy, chemotherapy alone, or immunotherapy
Those who have had treatment that saves the sphincter muscles may receive follow-up exams every 3 months for the first 2 years, including rectal exams with endoscopy and biopsy, as needed to check for recurrence.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Treatment of HIV and Anal Cancer
In general, treatment for people who have anal cancer and HIV is similar to treatment for other people, and these patients have similar outcomes. However, this treatment can further damage the weakened immune systems of people who have HIV. Treatment for people with a history of AIDS-related complications may require lower doses of anticancer drugs and radiation therapy than doses used for patients who do not have HIV.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Physician Data Query (PDQ) is the National Cancer Institute’s (NCI’s) comprehensive cancer information database. The PDQ database contains summaries of the latest published information on cancer prevention, detection, genetics, treatment, supportive care, and complementary and alternative medicine. Most summaries come in two versions. The health professional versions have detailed information written in technical language. The patient versions are written in easy-to-understand, nontechnical language. Both versions have cancer information that is accurate and up to date and most versions are also available in Spanish.
PDQ is a service of the NCI. The NCI is part of the National Institutes of Health (NIH). NIH is the federal government’s center of biomedical research. The PDQ summaries are based on an independent review of the medical literature. They are not policy statements of the NCI or the NIH.
Purpose of This Summary
This PDQ cancer information summary has current information about the treatment of anal cancer. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.
Reviewers and Updates
Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.
The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Adult Treatment Editorial Board.
Clinical Trial Information
A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).
Permission to Use This Summary
PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”
The best way to cite this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Anal Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/anal/patient/anal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389368]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use in the PDQ summaries only. If you want to use an image from a PDQ summary and you are not using the whole summary, you must get permission from the owner. It cannot be given by the National Cancer Institute. Information about using the images in this summary, along with many other images related to cancer can be found in Visuals Online. Visuals Online is a collection of more than 3,000 scientific images.
Disclaimer
The information in these summaries should not be used to make decisions about insurance reimbursement. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s E-mail Us.
Human papillomavirus (HPV) infection is the strongest risk factor for anal cancer and is accepted as a causal agent of squamous cell carcinoma of the anus and its precursor lesions.[1] Behaviors or medical conditions that either indicate HPV infection or facilitate HPV transmission or persistence are associated with increased risk. These behaviors and conditions include a history of HPV-related cancers, high-risk sexual practices such as sex between men, receptive anal intercourse and numerous sexual partners, human immunodeficiency virus (HIV) infection, and chronic immunosuppressive states.[2] Cigarette smoking is also a risk factor.[3]
Factors Associated With Increased Risk of Anal Cancer
Anal HPV infection
Based on solid evidence, HPV infection causes squamous cell carcinoma of the anus.
Magnitude of Effect: About 90% of anal squamous cell cancers occur in individuals with detectable HPV infection.[4] Of those, HPV strain 16 (HPV-16) and/or HPV-18 are detectable in more than 90% of cases.[4] Eighty-five percent of anal cancers have squamous cell histology.[2]
Study Design: Case series in men and women (HPV typing of tumor tissue).
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Behaviors or medical conditions associated with HPV infection
Based on solid evidence, behaviors or medical conditions that either indicate HPV infection or facilitate HPV transmission or persistence increase the risk or are associated with increased risk of anal cancer.
Magnitude of Effect: Risk varies by behavior and medical condition.
History of cervical, vaginal, and vulvar cancer increases risk at least threefold.[5–8]
High-risk sexual practices increase risk at least twofold, higher for individuals with many sexual partners and those who engage in receptive anal intercourse.[3,9–11]
Chronic immunosuppressive states increase risk about 30-fold for individuals who are HIV positive, and risk is much higher for men who both are HIV positive and have sex with men.[12]
Risk is at least threefold higher for organ transplant recipients.[11]
Study Design: Cohort, cancer registries, case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Cigarette smoking
Based on solid evidence, cigarette smoking increases the risk of anal cancer.
Magnitude of Effect: Risk is about twofold to threefold for ever-smokers; current smokers are at higher risk.[3,11,13]
Study Design: Cohort, case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Interventions Associated With a Decreased Risk of Anal Cancer
HPV vaccination
Based on solid evidence, HPV vaccination of men aged 16 to 26 years who have sex with men in the year before vaccination reduces anal intraepithelial neoplasia (AIN), a precursor lesion of anal cancer.
Magnitude of Effect: Vaccine efficacy against HPV-6, -11, -16, or -18–related AIN is between 50% and 75%.[14] Among those individuals who were naïve to vaccine types, incident quadrivalent HPV-type–associated anal low-grade squamous cell intraepithelial lesions (LSIL)/high-grade squamous cell intraepithelial lesions (HSIL) were not detected. In comparison, 11.1, 2.2, 4.5, and 2.8 cases per 100 person-years were reported for HPV-6, -11, -16, and -18–associated LSIL/HSIL, respectively, among those who were previously exposed to that respective HPV-type.[15]
Study Design: Randomized controlled trial, phase II open-label study.
Internal Validity: Good.
Consistency: Not applicable (N/A)—only one study.
External Validity: Good.
Based on national population-level observational data trends, HPV vaccination appears to decrease the risk of anal carcinoma in situ and invasive anal cancer among individuals aged 20 to 44 years after 2008 (when HPV vaccines were widely available).
Magnitude of Effect: Although direct efficacy of the HPV vaccine could not be measured in this population-level analysis, results showed a statistically significant decrease in the incidence of both anal carcinoma in situ (24% decrease) and invasive anal cancer (15% decrease). Older age groups (>45 years) who were not eligible for the HPV vaccine demonstrated increased HPV incidence after 2008.[16]
Study Design: National population-level cancer data.
Internal Validity: Good.
Consistency: Data from other HPV-related cancers, including population-level cervical cancer data, have shown similar decreases in overall cervical cancer incidence.
External Validity: Good.
Screening with high-resolution anoscopy (HRA) and treatment for high-grade squamous intraepithelial lesions (HSIL)
Utilizing data from a cohort of 28,175 individuals undergoing treatment for HIV in the Netherlands, anal cancer incidence significantly decreased over time. In a subcohort of 3,866 men who have sex with men (MSM) and had HRA-based anal cancer screening at least once, anal cancer mortality decreased by 31% when compared with those who did not participate in anal cancer screening (even after controlling for CD4 count less than 200).
Even though people living with HIV represent a high-risk population, anal cancer is still rare, and there were few anal cancers in this cohort, rendering conclusions difficult. In particular, there were only 37 anal cancers in men who do not have sex with men and only 10 among women. As a result, confidence intervals (CIs) were wide, and conclusions were suggestive at best.
Magnitude of Effect: Although direct efficacy of screening with HRA could not be measured in this cohort study, it found a statistically significant decrease in anal cancer mortality from 24% in the unscreened population to 3.7% in the screened population.
Study Design: Retrospective observational.
Internal Validity: Fair.
Consistency: Good.
External Validity: Fair.
Treatment of anal HSIL
Based on solid evidence, treating anal HSIL prevents anal cancer in HIV-positive individuals older than 35 years. When compared with the active monitoring arm, active HSIL treatment was associated with a decreased progression to anal cancer incidence by 57%.[17]
Study Design: Randomized phase III clinical trial.
Internal Validity: Good.
Consistency: N/A—only one study.
External Validity: Fair.
Interventions With Inadequate Evidence as to Whether They Reduce the Risk of Anal Cancer
Condom use
In a study of HPV transmission, MSM, recently had anal sex, and never use condoms were more likely to be infected with oncogenic HPV strains than were those who always used condoms. However, the association was not statistically significant.
Magnitude of Effect: About twofold but not statistically significant (odds ratio, 1.81; 95% CI, 0.58–5.68).[18]
Study Design: Case-control study.
Internal Validity: Fair.
Consistency: N/A—only one study.
External Validity: Fair.
References
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Human papillomaviruses. IARC Monogr Eval Carcinog Risks Hum 90: 1-636, 2007. [PUBMED Abstract]
Zandberg DP, Bhargava R, Badin S, et al.: The role of human papillomavirus in nongenital cancers. CA Cancer J Clin 63 (1): 57-81, 2013. [PUBMED Abstract]
Daling JR, Madeleine MM, Johnson LG, et al.: Human papillomavirus, smoking, and sexual practices in the etiology of anal cancer. Cancer 101 (2): 270-80, 2004. [PUBMED Abstract]
Parkin DM, Bray F: Chapter 2: The burden of HPV-related cancers. Vaccine 24 (Suppl 3): S3/11-25, 2006. [PUBMED Abstract]
Chaturvedi AK, Engels EA, Gilbert ES, et al.: Second cancers among 104,760 survivors of cervical cancer: evaluation of long-term risk. J Natl Cancer Inst 99 (21): 1634-43, 2007. [PUBMED Abstract]
Hemminki K, Dong C, Vaittinen P: Second primary cancer after in situ and invasive cervical cancer. Epidemiology 11 (4): 457-61, 2000. [PUBMED Abstract]
Ruth A, Kosary A, Hildesheim A: New malignancies following cancer of the cervix uteri, vagina, and vulva. In: Curtis RE, Freedman DM, Ron E, et al., eds.: New Malignancies Among Cancer Survivors: SEER Cancer Registries, 1973-2000. National Cancer Institute, 2006. NIH Pub. No. 05-5302, pp 207-30.
Saleem AM, Paulus JK, Shapter AP, et al.: Risk of anal cancer in a cohort with human papillomavirus-related gynecologic neoplasm. Obstet Gynecol 117 (3): 643-9, 2011. [PUBMED Abstract]
Daling JR, Weiss NS, Hislop TG, et al.: Sexual practices, sexually transmitted diseases, and the incidence of anal cancer. N Engl J Med 317 (16): 973-7, 1987. [PUBMED Abstract]
Frisch M, Glimelius B, van den Brule AJ, et al.: Sexually transmitted infection as a cause of anal cancer. N Engl J Med 337 (19): 1350-8, 1997. [PUBMED Abstract]
van der Zee RP, Richel O, de Vries HJ, et al.: The increasing incidence of anal cancer: can it be explained by trends in risk groups? Neth J Med 71 (8): 401-11, 2013. [PUBMED Abstract]
Silverberg MJ, Lau B, Justice AC, et al.: Risk of anal cancer in HIV-infected and HIV-uninfected individuals in North America. Clin Infect Dis 54 (7): 1026-34, 2012. [PUBMED Abstract]
Nordenvall C, Nilsson PJ, Ye W, et al.: Smoking, snus use and risk of right- and left-sided colon, rectal and anal cancer: a 37-year follow-up study. Int J Cancer 128 (1): 157-65, 2011. [PUBMED Abstract]
Palefsky JM, Giuliano AR, Goldstone S, et al.: HPV vaccine against anal HPV infection and anal intraepithelial neoplasia. N Engl J Med 365 (17): 1576-85, 2011. [PUBMED Abstract]
Palefsky JM, Lensing SY, Belzer M, et al.: High Prevalence of Anal High-Grade Squamous Intraepithelial Lesions, and Prevention Through Human Papillomavirus Vaccination, in Young Men Who Have Sex With Men Living With Human Immunodeficiency Virus. Clin Infect Dis 73 (8): 1388-1396, 2021. [PUBMED Abstract]
Berenson AB, Guo F, Chang M: Association of Human Papillomavirus Vaccination With the Incidence of Squamous Cell Carcinomas of the Anus in the US. JAMA Oncol 8 (4): 1-3, 2022. [PUBMED Abstract]
Palefsky JM, Lee JY, Jay N, et al.: Treatment of Anal High-Grade Squamous Intraepithelial Lesions to Prevent Anal Cancer. N Engl J Med 386 (24): 2273-2282, 2022. [PUBMED Abstract]
Nyitray AG, Carvalho da Silva RJ, Baggio ML, et al.: Age-specific prevalence of and risk factors for anal human papillomavirus (HPV) among men who have sex with women and men who have sex with men: the HPV in men (HIM) study. J Infect Dis 203 (1): 49-57, 2011. [PUBMED Abstract]
Incidence, Mortality, and Survival
United States
The Surveillance, Epidemiology, and End Results (SEER) Program age-adjusted annual incidence rate of anal cancer in the United States from 2017 to 2021 was 1.9 cases per 100,000 persons per year, and the mortality rate was 0.4 cases per 100,000 persons per year from 2018 to 2022. Incidence rates were slightly higher for women than for men (2.3 vs. 1.6 per 100,000 person-years, respectively), but mortality rates were about the same.[1] In 2025, it is estimated that 10,930 Americans will be diagnosed with anal cancer and 2,030 will die of this disease.[2] Incidence rates increased annually from 2012 to 2021 (average increase, 1.3%), and mortality rates increased annually from 2013 to 2022 (average increase, 5.1%). All incidence and mortality increases were statistically different from zero.[3] The 5-year survival rate has remained fairly constant since 1975, and based on data from 2014 to 2020, it was 70.6%.[1]
World
An estimated 27,000 new cases of anal cancer were diagnosed worldwide in 2008.[4] No global incidence rates, mortality rates, or survival statistics are available.
References
National Cancer Institute: SEER Stat Fact Sheets: Anal Cancer. Bethesda, Md: National Cancer Institute. Available online. Last accessed April 8, 2025.
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
Surveillance Research Program, National Cancer Institute: SEER*Explorer: An interactive website for SEER cancer statistics. Bethesda, MD: National Cancer Institute. Available online. Last accessed December 30, 2024.
Forman D, de Martel C, Lacey CJ, et al.: Global burden of human papillomavirus and related diseases. Vaccine 30 (Suppl 5): F12-23, 2012. [PUBMED Abstract]
Histology
About 85% of anal cancers in the United States have squamous cell histology or a histological variant.[1] Nearly all other anal cancers are adenocarcinomas.[2] Human papillomavirus (HPV) vaccination, HPV screening, and screening for the presence of anal cancer precursor lesions will probably change the histological distribution of anal cancer in years to come, as HPV is implicated only in squamous cell carcinomas,[3] and identification of precursor lesions is expected to reduce invasive squamous cell disease.
Precursor Lesions
Squamous cell cancer of the anus is preceded by grade 2 or 3 anal intraepithelial neoplasia (AIN), also referred to as high-grade AIN. Grade 1 AIN is not considered a precursor lesion of anal cancer but may precede high-grade AIN.[4] The cytological terms for low- and high-grade AIN are low-grade squamous cell intraepithelial lesions (LSIL) and high-grade squamous cell intraepithelial lesions (HSIL).[4]
One study reported that 11% of AIN cases progressed to invasive disease over an 8-year period.[5] However, results from another study suggested that progression is much less frequent. Using AIN prevalence and anal cancer incidence data, the investigators estimated hypothetical annual rates of progression from high-grade AIN to anal cancer. For men who have sex with men (MSM) and who are human immunodeficiency virus (HIV) positive, the rate was about 1 case in 600 patients. For HIV-negative MSM, the rate was 1 case in 4,000 patients.[6] Using meta-analysis techniques to combine data from numerous studies worldwide, the investigators estimated that the prevalence of LSIL is 27.5% (95% confidence interval [CI], 21.9%–33.2%) and the prevalence of HSIL is 6.7% (95% CI, 4.4%–9.0%) in HIV-positive MSM. Among HIV-negative MSM, the prevalence of LSIL was 6.6% (95% CI, 1.1%–12.1%), and the prevalence of HSIL was 2.7% (95% CI, 0.0%–5.1%).[6]
References
Zandberg DP, Bhargava R, Badin S, et al.: The role of human papillomavirus in nongenital cancers. CA Cancer J Clin 63 (1): 57-81, 2013. [PUBMED Abstract]
Ries LAG, Young JL, Keel GE, et al., eds.: SEER Survival Monograph: Cancer Survival Among Adults: U. S. SEER Program, 1988-2001, Patient and Tumor Characteristics. National Cancer Institute, 2007. NIH Pub. No. 07-6215.
Joseph DA, Miller JW, Wu X, et al.: Understanding the burden of human papillomavirus-associated anal cancers in the US. Cancer 113 (10 Suppl): 2892-900, 2008. [PUBMED Abstract]
Hoots BE, Palefsky JM, Pimenta JM, et al.: Human papillomavirus type distribution in anal cancer and anal intraepithelial lesions. Int J Cancer 124 (10): 2375-83, 2009. [PUBMED Abstract]
Watson AJ, Smith BB, Whitehead MR, et al.: Malignant progression of anal intra-epithelial neoplasia. ANZ J Surg 76 (8): 715-7, 2006. [PUBMED Abstract]
Machalek DA, Poynten M, Jin F, et al.: Anal human papillomavirus infection and associated neoplastic lesions in men who have sex with men: a systematic review and meta-analysis. Lancet Oncol 13 (5): 487-500, 2012. [PUBMED Abstract]
Risk Factors
Factors Associated With Increased Risk of Anal Cancer
Anal HPV infection
Human papillomavirus (HPV) infection is the strongest risk factor for anal cancer. About 90% of anal cancers occur in individuals with detectable HPV infection.[1] HPV infection with oncogenic HPV strains is accepted as a causal agent and necessary condition for development of squamous cell carcinoma of the anus and its precursor lesions.[2] In a 2009 meta-analysis of about 1,000 invasive squamous cell lesions, HPV-16 was present in about two-thirds of lesions, and HPV-18 was present in about 5% of lesions.[3] Because 85% of anal cancers have a squamous cell carcinoma histology or a histological variant,[4] it is probable that elimination of oncogenic HPV infection would nearly eradicate anal cancer.
HPVs are typically cleared rapidly in healthy individuals. Persistence of the oncogenic HPV strains is more likely in individuals with compromised immune systems; therefore, the risk of squamous cell anal cancer is much higher in these individuals. Behaviors that facilitate transmission of HPVs also increase risk.[4] While these conditions or behaviors will probably have, at most, little independent effect on squamous cell anal cancer risk (that is, in the absence of HPV), data that fully address this hypothesis are very limited.
Given the paucity of cases of anal adenocarcinoma and other nonsquamous histological subtypes, it is unknown what role, if any, HPV plays in the development of these lesions.
Behaviors or medical conditions associated with HPV infection
History of cervical, vaginal, and vulvar cancer
Cancers of the cervix, vagina, and vulva are HPV-related cancers.[5] Long-term registry-based monitoring of cervical, vaginal, and vulvar cancer survivors demonstrates an increase in anal cancer risk for these individuals, although the magnitude of the relationship varies.[6–9] For survivors of invasive cervical cancer, the standardized incidence ratio (SIR) for anal cancer was 3.1 (95% confidence interval [CI], 1.9–4.9) in a cohort of more than 100,000 cervical cancer survivors from Denmark, Finland, Norway, Sweden, and the United States.[6] The SIR for anal cancer was 6.2 (95% CI, 4.1–8.7) for survivors of invasive cervical cancer in the Surveillance, Epidemiology, and End Results (SEER) Program registry data from 1973 to 2007 (more than 1 million person-years).[8] In the latter cohort, the SIR for women with in situ cervical cancer was 16.4 (95% CI, 13.7–19.2). In an analysis of data from the Swedish Family-Cancer Database, which used data from 1958 to 1996, SIRs were 3.8 (95% CI, 2.9–4.7) among the women with in situ cervical cancer and 3.9 (95% CI, 2.3–6.0) among the women with invasive cervical cancer.[8] In the aforementioned multicountry cohort,[6] the anal cancer SIRs for in situ and invasive vaginal cancer were 7.6 (95% CI, 2.4–15.6) and 1.8 (95% CI, 0.2–5.3), respectively; the anal cancer SIRs for in situ and invasive vulvar cancer were 22.2 (95% CI, 16.7–28.4) and 17.4 (95% CI, 16.7–28.4), respectively.
Individuals with cancer of the oropharynx [10] and penis,[11] two other HPV-associated cancers, are hypothesized to be at increased risk of anal cancer. From 1973 to 2007 (more than 1 million person-years),[7] it was estimated that the observed-to-expected ratio for anal cancer among people with oropharyngeal cancer was twofold (significantly different from one). In that same data source, it was also estimated that no anal cancers occurred after penile cancer, although the expected number of cases was 0.36.
HIV infection/AIDS
The association between HIV infection and anal cancer is strong. One meta-analysis indicated a 30-fold increase in anal cancer in HIV-infected people, compared with the general population (SIR, 28.8; 95% CI, 21.6–38.3).[12] A nationwide Danish cohort study with data from 1995 to 2009 observed an even stronger association (incidence rate ratio, 77.9; 95% CI, 36.2–167.7).[13] This association between HIV infection and anal cancer is confounded or modified by other factors associated with anal cancer, such as HPV status, high-risk behaviors, and level of immunocompromise. For example, the magnitude of the association between HIV infection and anal cancer risk varies by sexual preference. In one study,[14] the highest SIR and the highest incidence rate were observed for HIV-positive men who have sex with men (MSM), compared with HIV-negative men (SIR, 80.3; 95% CI, 42.7–151.1). The incidence rate for HIV-positive MSM is 131 cases per 100,000 person-years. The SIR for HIV-positive men who did not have sex with men was lower but nonnegligible (SIR, 26.7; 95% CI, 11.5–61.7; incidence rate, 46 cases per 100,000 person-years). In the same study, 30 of 8,842 HIV-positive women had anal cancer diagnoses (incidence rate, 2 cases per 100,000 person-years) but none of the 11,653 HIV-negative women were diagnosed with anal cancer; thus, no SIR could be calculated, and the incidence rate was zero. Among men with anal cancer, the Danish study observed a mortality rate ratio of 3.2 (95% CI, 1.1–9.2) for HIV-positive men compared with men in the general population.[13]
Anal HPV infection is common in HIV-positive individuals. Studies suggest an HPV prevalence of 85% to 95% among HIV-positive MSM, 76% to 90% in HIV-positive women, and 60% in HIV-positive heterosexual men.[14]
In a cohort of almost 7,000 men with AIDS, 28 anal cancers occurred, and the odds ratios (OR) suggested relatively modest elevations (about twofold) in risk as the prevalence of high-risk behaviors increased. However, the only statistically significant OR relating to sexual practices was for seven or more unprotected anal receptive sexual partners during the time between study onset and the third study visit (OR, 4.0; 95% CI, 1.1–14.6).[15] In a cohort of nearly half a million AIDS patients, intravenous drug use was associated with anal cancer (SIR, 11.7; 95% CI, 4.2–25.5 for men and SIR, 38.0; 95% CI, 10.3–97.3 for women).[16] Current cigarette smoking, relative to never smoking, has also been observed to increase anal cancer risk in HIV-positive individuals (OR, 2.6; 95% CI, 1.3–5.3).[17]
Anal cancer risk is positively associated with severity of immunosuppression in HIV-positive and AIDS patients.[14] When combined antiretroviral therapy (cART) became available in 1996, the incidence of anal cancer among these patients was expected to decrease. While decreases have been observed for other HIV-associated cancers, such trends have not been observed for anal cancer. It has been proposed that timing of cART treatment influences the risk of anal cancer, and that to be effective against anal cancer, cART must be administered to those with HPV infection earlier in the course of infection than has been clinically practiced.[18] One study suggests that immunosuppression levels 6 to 7 years before anal cancer diagnosis may be more strongly associated with odds of developing the disease than immunosuppression levels in the 12 months before anal cancer diagnosis.
Investigators reported ORs for CD4+ counts 6 to 7 years before anal cancer diagnosis as follows:[17]
For counts of 350 to 499: OR, 2.8 (95% CI, 0.6–13.0).
For counts of 200 to 349: OR , 5.9 (95% CI, 1.5–23.0).
For counts lower than 200: OR, 14.0 (95% CI, 3.9–50.9).
ORs for CD4+ counts in the 12 months before diagnosis were as follows:
For counts of 350 to 499: OR, 2.0 (95% CI, 0.9–4.6).
For counts of 200 to 349: OR, 2.2 (95% CI, 1.1–4.6).
For counts lower than 200: OR, 4.6 (95% CI, 1.8–11.4).
Similar patterns were observed for CD8+ cell counts and for CD4+/CD8+ ratios.
Sexual practices associated with increased risk
Sexual practices that confer elevation in anal cancer risk include receptive anal intercourse, numerous sexual partners, and sex between men.[19] These are practices that are known or believed to increase anal exposure to oncogenic strains of HPV. Because HPV and HIV infection are highly correlated with high-risk sexual practices, few data exist that assess the independent effects of sexual behaviors. Before the HIV/AIDS era, the epidemiology of anal cancer received little attention; it was only as the concurrent emergence of AIDS and the increase in anal cancer occurred that sexual practices were investigated as possible risk factors.
Regardless of the underlying reason, MSM have the highest rates of anal cancer when compared with other men and women. As previously mentioned, HIV-positive MSM have the highest anal cancer rates (about 50 cases per 100,000 person-years),[20] but HIV-negative MSM have significantly higher rates than do men in the general population; their incidence is estimated to be 5 cases per 100,000 person-years.[20] Case-control studies have observed a modest (about twofold) increase in risk for women who practice receptive anal intercourse;[21,22] however, one study found the association to exist only among women who first had anal intercourse before age 30 years (OR, 3.4; 95% CI, 1.7–6.6).[21] In the same study, adjusted ORs for both men and women increased with increasing lifetime number of sexual partners. The OR associated with 10 or more partners was 4.5 (95% CI, 2.7–7.4) for women and 2.5 (95% CI, 1.1–5.5) for men. Increased risk for both men and women has been observed with a history of anal warts and certain other sexually transmitted diseases.[21]
Chronic immunosuppressive states other than HIV infection
Chronic immunosuppression in general is thought to increase risk of anal cancer because of its impact on the ability to clear HPV infection.[19] Organ transplant recipients are at elevated risk of anal cancer because immunosuppressant medications are used to prevent organ rejection. Three large transplant cohort studies have observed SIRs for anal cancer of 2.8 (95% CI, 1.5–4.6),[12] 5.8 (95% CI, 4.7–7.2),[23] and 10.3 (95% CI, 2.8–26.6).[12] Autoimmune disorders are hypothesized to increase risk of anal cancer because of the condition, the treatment, or both. However, the rarity of anal cancer and relative rarity of many of these disorders have led to conflicting findings or limited data. A cohort study of the Denmark National Patient Registry that included nearly 30 years’ experience observed statistically significant threefold increases in risk for Crohn disease (SIR, 3.1; 95% CI, 1.2–6.4) and psoriasis (SIR, 3.1; 95% CI, 1.8–5.1), as well as a ninefold increase for polyarteritis nodosa (SIR, 8.8; 95% CI, 1.5–29.0) and a 12-fold increase in Wegener granulomatosis (SIR, 12.4; 95% CI, 2.1–40.8).[24]
Cigarette smoking
Cigarette smoking was among the first risk factors for anal cancer to be identified. In 1987, a case-control study of 58 men and 90 women observed a ninefold increase in risk (relative risk [RR], 9.4; 95% CI, 2.3–38.5) for men and an eightfold increase in risk for women (RR, 7.7; 95% CI, 3.5–17.2) for current smokers after adjustment for number of sexual partners.[25] RRs for former smokers were not statistically significant and less than twofold. Another case-control study of 306 patients suggested that current cigarette smoking may be an independent risk factor for anal cancer because adjustment for HPV status and number of sexual partners dampened but did not eliminate the significant associations observed in the 1987 study. The OR was 3.9 (95% CI, 1.9–8.0) for men and 3.8 (95% CI, 2.3–6.2) for women.[22] Given the rarity of anal cancer, studies have not been able to rigorously explore whether risk of anal cancer varies by other aspects of smoking history, such as pack-years smoked and time since cessation. The latter would be of particular interest, given the observation of strong risk in current smokers but no risk in former smokers.
References
Parkin DM, Bray F: Chapter 2: The burden of HPV-related cancers. Vaccine 24 (Suppl 3): S3/11-25, 2006. [PUBMED Abstract]
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Human papillomaviruses. IARC Monogr Eval Carcinog Risks Hum 90: 1-636, 2007. [PUBMED Abstract]
Hoots BE, Palefsky JM, Pimenta JM, et al.: Human papillomavirus type distribution in anal cancer and anal intraepithelial lesions. Int J Cancer 124 (10): 2375-83, 2009. [PUBMED Abstract]
Zandberg DP, Bhargava R, Badin S, et al.: The role of human papillomavirus in nongenital cancers. CA Cancer J Clin 63 (1): 57-81, 2013. [PUBMED Abstract]
Forman D, de Martel C, Lacey CJ, et al.: Global burden of human papillomavirus and related diseases. Vaccine 30 (Suppl 5): F12-23, 2012. [PUBMED Abstract]
Chaturvedi AK, Engels EA, Gilbert ES, et al.: Second cancers among 104,760 survivors of cervical cancer: evaluation of long-term risk. J Natl Cancer Inst 99 (21): 1634-43, 2007. [PUBMED Abstract]
Saleem AM, Paulus JK, Shapter AP, et al.: Risk of anal cancer in a cohort with human papillomavirus-related gynecologic neoplasm. Obstet Gynecol 117 (3): 643-9, 2011. [PUBMED Abstract]
Hemminki K, Dong C, Vaittinen P: Second primary cancer after in situ and invasive cervical cancer. Epidemiology 11 (4): 457-61, 2000. [PUBMED Abstract]
Ruth A, Kosary A, Hildesheim A: New malignancies following cancer of the cervix uteri, vagina, and vulva. In: Curtis RE, Freedman DM, Ron E, et al., eds.: New Malignancies Among Cancer Survivors: SEER Cancer Registries, 1973-2000. National Cancer Institute, 2006. NIH Pub. No. 05-5302, pp 207-30.
Frisch M, Melbye M: Anal cancer. In: Schottenfeld D, Fraumeni JF Jr, eds.: Cancer Epidemiology and Prevention. 3rd ed. Oxford University Press, 2006, pp 830-40.
McMaster ML, Feuer EJ, Tucker MA: New malignancies following cancer of the male genital tract. In: Curtis RE, Freedman DM, Ron E, et al., eds.: New Malignancies Among Cancer Survivors: SEER Cancer Registries, 1973-2000. National Cancer Institute, 2006. NIH Pub. No. 05-5302, pp 257-84.
Grulich AE, van Leeuwen MT, Falster MO, et al.: Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet 370 (9581): 59-67, 2007. [PUBMED Abstract]
Legarth R, Helleberg M, Kronborg G, et al.: Anal carcinoma in HIV-infected patients in the period 1995-2009: a Danish nationwide cohort study. Scand J Infect Dis 45 (6): 453-9, 2013. [PUBMED Abstract]
Silverberg MJ, Lau B, Justice AC, et al.: Risk of anal cancer in HIV-infected and HIV-uninfected individuals in North America. Clin Infect Dis 54 (7): 1026-34, 2012. [PUBMED Abstract]
D’Souza G, Wiley DJ, Li X, et al.: Incidence and epidemiology of anal cancer in the multicenter AIDS cohort study. J Acquir Immune Defic Syndr 48 (4): 491-9, 2008. [PUBMED Abstract]
Chaturvedi AK, Madeleine MM, Biggar RJ, et al.: Risk of human papillomavirus-associated cancers among persons with AIDS. J Natl Cancer Inst 101 (16): 1120-30, 2009. [PUBMED Abstract]
Bertisch B, Franceschi S, Lise M, et al.: Risk factors for anal cancer in persons infected with HIV: a nested case-control study in the Swiss HIV Cohort Study. Am J Epidemiol 178 (6): 877-84, 2013. [PUBMED Abstract]
Engels EA, Madeleine MM: Invited commentary: Biological and clinical insights from epidemiologic research into HIV, HPV, and anal cancer. Am J Epidemiol 178 (6): 885-7, 2013. [PUBMED Abstract]
van der Zee RP, Richel O, de Vries HJ, et al.: The increasing incidence of anal cancer: can it be explained by trends in risk groups? Neth J Med 71 (8): 401-11, 2013. [PUBMED Abstract]
Machalek DA, Poynten M, Jin F, et al.: Anal human papillomavirus infection and associated neoplastic lesions in men who have sex with men: a systematic review and meta-analysis. Lancet Oncol 13 (5): 487-500, 2012. [PUBMED Abstract]
Frisch M, Glimelius B, van den Brule AJ, et al.: Sexually transmitted infection as a cause of anal cancer. N Engl J Med 337 (19): 1350-8, 1997. [PUBMED Abstract]
Daling JR, Madeleine MM, Johnson LG, et al.: Human papillomavirus, smoking, and sexual practices in the etiology of anal cancer. Cancer 101 (2): 270-80, 2004. [PUBMED Abstract]
Engels EA, Pfeiffer RM, Fraumeni JF, et al.: Spectrum of cancer risk among US solid organ transplant recipients. JAMA 306 (17): 1891-901, 2011. [PUBMED Abstract]
Sunesen KG, Nørgaard M, Thorlacius-Ussing O, et al.: Immunosuppressive disorders and risk of anal squamous cell carcinoma: a nationwide cohort study in Denmark, 1978-2005. Int J Cancer 127 (3): 675-84, 2010. [PUBMED Abstract]
Daling JR, Weiss NS, Hislop TG, et al.: Sexual practices, sexually transmitted diseases, and the incidence of anal cancer. N Engl J Med 317 (16): 973-7, 1987. [PUBMED Abstract]
Interventions Associated With a Decreased Risk of Anal Cancer
HPV Vaccination
Because human papillomavirus (HPV) is a causal condition for squamous cell anal cancer development, vaccination against the oncogenic strains of HPV before exposure may reduce the risk of anal cancer. Conducted from 2004 to 2008, a multicountry trial randomly assigned 4,065 boys and men to receive either the three-shot quadrivalent HPV vaccine regimen (for HPV-6, -11, -16, and -18) or a three-shot placebo injection regimen. Of the 4,065 patients, 602 reported having sex with male partners in the year before enrollment. Heterosexual participants were between the ages of 16 years and 23 years and had no more than five lifetime female partners. Patients who reported sex with male partners were between the ages of 16 years and 26 years and had no more than five lifetime male or female partners. Persistent infection was defined as detection of the same HPV type in anogenital swabs or biopsy specimens collected on two or more consecutive visits, with an interval of 6 months between visits. In the intent-to-treat analysis, which included participants regardless of their baseline HPV status, the efficacy against persistent HPV-6, -11, -16, and -18 infection was 48% (95% confidence interval [CI], 36.0%–57.6%). Among those who were negative for the four HPV strains of interest at baseline (per the protocol analysis, which included 1,397 intervention-arm and 1,408 control-arm participants), vaccine efficacy against persistent HPV-6, -11, -16, and -18 infection was 90% (95% CI, 69.2%–98.1%).[1]
A nonrandomized, phase II, open-label trial (AMC 072 [NCT01209325]) was conducted in 149 men who have sex with men living with HIV. This study did not show a statistically significant difference between incident-persistent infections in the naïve, per-protocol, and previously exposed per-protocol groups. However, there was a statistically significant reduction in incident HPV-16–associated histological high-grade squamous intraepithelial lesions (HSIL) in the naïve group when compared with the previously exposed group (P = .014). The authors conducted a secondary analysis that compared the per-protocol quadrivalent HPV (qHPV)-type naïve participants in the AMC 072 trial to the original, per-protocol placebo group in the Merck 020 trial. This analysis demonstrated that vaccinated qHPV-naïve AMC-072 participants had significantly reduced disease in a combined analysis of all four qHPV types (5.8 per 100 person-years vs. 0 per 100 person-years; P = .008).[2]
Among the 602 patients who had sex with men, the vaccine efficacy against persistent HPV-6, -11, -16, and -18 infection was 59% (95% CI, 43.0%–71.4%) in the intent-to-treat analysis and 95% (95% CI, 80.4%–99.4%) in the per-protocol analysis. Efficacy against HPV-6, -11, -16 or -18–associated anal intraepithelial neoplasia (AIN) was 50% (95% CI, 25.7%–67.2%) in the intention-to-treat analysis and 77.5% (95% CI, 39.6%–93.9%) in the per-protocol analysis (275 intervention-arm and 276 control-arm participants). Efficacy against HPV-6, -11, -16, or -18–associated high-grade AIN was 54.2% (95% CI, 18.0%–75.3%) in the intent-to-treat analysis and 74.9% (95% CI, 8.8%–95.4%) in the per-protocol analysis (194 intervention-arm and 208 control-arm participants).[3]
Efficacy of the bivalent (HPV-16 and HPV-18) vaccine against anal infection was evaluated in the context of a randomized controlled trial of cervical cancer prevention. Conducted in 6,300 Costa Rican women aged 18 to 25 years at enrollment, the trial compared the efficacy of the three-dose bivalent vaccine with that of a control vaccine. Four years after vaccination, most women were offered the option of providing an anal specimen. Among the 2,103 intervention-arm and 2,107 control-arm participants who provided specimens, vaccine efficacy (i.e., absence of HPV-16 or -18 in the specimen) was 62% (95% CI, 47.1%–73.1%). Among the 1,003 intervention-arm and 986 control-arm participants who provided anal specimens, received the three doses, had no evidence of cervical HPV-16 or -18 infection before vaccination, and were seronegative before vaccination, vaccine efficacy was 84% (95% CI, 66.7%–92.8%).[4]
These data strongly suggest that vaccination against oncogenic HPV strains will lead to reductions in anal cancer. They also suggest that vaccination before exposure will provide the most benefit.
Treatment of Anal HSIL
Until the Anal Cancer–HSIL Outcomes Research (ANCHOR) trial was published, there had not been confirmation that treating anal HSIL decreased an individual’s risk for invasive cancer.[5] However, the findings from the ANCHOR trial provided solid evidence that treatment of anal HSIL prevented the incidence of anal cancer. This trial, which was a phase III trial conducted at 25 U.S. sites, included individuals living with HIV who were aged 35 years or older and had biopsy-proven anal HSIL. Participants were randomly assigned in a 1:1 ratio to receive either HSIL treatment or active monitoring without treatment. Nine cases were diagnosed in the treatment group (173 per 100,000 person-years; 95% CI, 90.0–332.0), and 21 cases were diagnosed in the active-monitoring group (402 per 100,000 person-years; 95% CI, 262.0–616.0) after a median follow-up of 25.8 months. The rate of progression to anal cancer was 57% lower in the treatment group than it was in the active-monitoring group (95% CI, 6.0–80.0; P = .03 by log-rank test).
In a recent cohort study of 28,175 individuals being treated for HIV (59.7%, men who have sex with men [MSM]), 227 primary anal cancer cases were diagnosed.[6] Despite the increasing average age of the cohort, crude incidence rates of anal cancer in MSM declined slowly over time from 107.0 per 100,000 person-years (95% CI, 75.7–147.0) in 1996 to 2005 to 93.7 per 100,000 person-years (95% CI, 75.3–115.0) in 2013 to 2020 (P = .49). Crude incidence rates in men who do not have sex with men (non-MSM) and women were generally lower than those in MSM. However, crude incidence rates increased slightly over time from 51.08 per 100,000 person-years (95% CI, 20.54–105.25) to 67.82 per 100,000 person-years (95% CI, 40.83–105.91; P = .52) in non-MSM and from 8.09 per 100,000 person-years (95% CI, 0.20–45.06) to 24.95 per 100,000 person-years (95% CI, 10.03–51.40; P = .29) in women. In addition, the authors compared a subcohort of 3,866 MSM who received high-resolution anoscopy (HRA) screening at least once and treatment for HSIL. Furthermore, if screened individuals had low-grade squamous intraepithelial lesions (LSIL), they continued HRA screening once yearly, and those who had HSIL treatment received HRA screening every 6 months. TNM tumor staging was more favorable (Cochrane-Armitage test for trend, P = .033) in individuals diagnosed with anal cancer during screening. Crude anal cancer–associated 5-year mortality in people living with HIV decreased from 30.4% (1996–2005) to 18.3% (2013–2020; odds ratio, 0.48; P = .070). Anal cancer–related mortality was 3.7% (95% CI, 0.5–23.5) in all men who had been screened and 24.0% (95% CI, 18.1–31.3) in men who had not been screened (P = .023). In men, screening participation (hazard ratio [HR], 0.31; P = .051) and cumulative exposure to CD4 counts of less than 200 cells per µL (HR, 1.11 per year; P = .0022) were independently associated with anal cancer–related mortality.
Even though people living with HIV represent a high-risk population, anal cancer is still rare, and there were few anal cancers in this cohort, rendering conclusions difficult. In particular, there were only 37 anal cancers in non-MSM and only 10 among women. As a result, CIs were wide, and conclusions were suggestive at best. Of note, there are no known randomized controlled trials that provide evidence to support the conclusion of this study.
Data do not support the conclusion that men had improved survival when they were diagnosed with anal cancer after screening or that it is important to screen those who are at high risk of developing anal cancer. The study purported to show reduced mortality after an anal cancer diagnosis among screened men, but the curves in the study’s results did not make adjustments for lead-time bias, selection bias, nor the possibility of overdiagnosis bias. The study also suggested that there was a larger proportion of early-stage cases among screened individuals, but the proportional stage shift caused by anal cancer screening is known to be influenced by lead-time and overdiagnosis biases. There was no mention of adjustment for these biases in the study’s results. In addition, there was a high proportion of unknown stages, further suggesting that the conclusions in this study were not supported by the data. This is an additional rationale for why the data did not support the conclusion of the study.
References
Giuliano AR, Palefsky JM, Goldstone S, et al.: Efficacy of quadrivalent HPV vaccine against HPV Infection and disease in males. N Engl J Med 364 (5): 401-11, 2011. [PUBMED Abstract]
Palefsky JM, Lensing SY, Belzer M, et al.: High Prevalence of Anal High-Grade Squamous Intraepithelial Lesions, and Prevention Through Human Papillomavirus Vaccination, in Young Men Who Have Sex With Men Living With Human Immunodeficiency Virus. Clin Infect Dis 73 (8): 1388-1396, 2021. [PUBMED Abstract]
Palefsky JM, Giuliano AR, Goldstone S, et al.: HPV vaccine against anal HPV infection and anal intraepithelial neoplasia. N Engl J Med 365 (17): 1576-85, 2011. [PUBMED Abstract]
Kreimer AR, González P, Katki HA, et al.: Efficacy of a bivalent HPV 16/18 vaccine against anal HPV 16/18 infection among young women: a nested analysis within the Costa Rica Vaccine Trial. Lancet Oncol 12 (9): 862-70, 2011. [PUBMED Abstract]
Palefsky JM, Lee JY, Jay N, et al.: Treatment of Anal High-Grade Squamous Intraepithelial Lesions to Prevent Anal Cancer. N Engl J Med 386 (24): 2273-2282, 2022. [PUBMED Abstract]
van der Zee RP, Wit FWNM, Richel O, et al.: Effect of the introduction of screening for cancer precursor lesions on anal cancer incidence over time in people living with HIV: a nationwide cohort study. Lancet HIV 10 (2): e97-e106, 2023. [PUBMED Abstract]
Interventions With Inadequate Evidence as to Whether They Reduce the Risk of Anal Cancer
Condom Use
Because human papillomavirus (HPV) can be transmitted through microabrasions, as well as through more pronounced exposures such as exchange of certain bodily fluids,[1] restriction of condom use to penetrative activity will not protect against transmission that occurs as part of other sexual contact. Nevertheless, condom use would be expected to reduce some risk of transmission and thus anal cancer risk. Few data that explore these hypotheses exist, and those that do suggest a very modest effect, if any. Of note, the ability of condom use to reduce cervical cancer risk is still uncertain and the subject of debate.[2]
In an Italian cohort of 258 HIV-negative men who have sex with men (MSM), the odds ratio (OR) for infection with high-risk HPV strains was 1.7 (95% confidence interval [CI], 0.52–6.3) for inconsistent or no use of condoms in receptive anal sex, compared with consistent condom use.[3] In a Brazilian cohort that included 176 MSM, the OR for oncogenic HPV infection was 1.8 (95% CI, 0.77–4.35) for men who sometimes used condoms for anal sex and 1.8 (95% CI, 0.58–5.68) for men who never used condoms, compared with men who always used condoms.[4]
References
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Human papillomaviruses. IARC Monogr Eval Carcinog Risks Hum 90: 1-636, 2007. [PUBMED Abstract]
Chelimo C, Wouldes TA, Cameron LD, et al.: Risk factors for and prevention of human papillomaviruses (HPV), genital warts and cervical cancer. J Infect 66 (3): 207-17, 2013. [PUBMED Abstract]
Donà MG, Palamara G, Di Carlo A, et al.: Prevalence, genotype diversity and determinants of anal HPV infection in HIV-uninfected men having sex with men. J Clin Virol 54 (2): 185-9, 2012. [PUBMED Abstract]
Nyitray AG, Carvalho da Silva RJ, Baggio ML, et al.: Age-specific prevalence of and risk factors for anal human papillomavirus (HPV) among men who have sex with women and men who have sex with men: the HPV in men (HIM) study. J Infect Dis 203 (1): 49-57, 2011. [PUBMED Abstract]
Latest Updates to This Summary (04/08/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 2).
This summary is written and maintained by the PDQ Screening and Prevention Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about anal cancer prevention. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Screening and Prevention Editorial Board. PDQ Anal Cancer Prevention. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/anal/hp/anal-prevention-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389511]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
Estimated new cases and deaths from anal, anal canal, and anorectal cancer in the United States in 2025:[1]
New cases: 10,930.
Deaths: 2,030.
Prognosis and Survival
The two major prognostic factors for anal cancer are tumor size and nodal status. Primary tumors smaller than 2 cm have a better prognosis.[2] Nodal drainage of the anus follows the inguinal vein. The initial evaluation of a patient with anal cancer will include a careful clinical examination of the inguinal region and biopsy of any palpable lymph nodes. For more information, see the American Joint Committee on Cancer Stage Groupings and TNM Definitions section.
Anal cancer is usually curable. At presentation, most patients have T1 or T2 disease (≤5 cm), and fewer than 20% of patients have node-positive disease. The 5-year survival rate for these early-stage patients exceeds 85%.[3,4] Even in patients with node-positive disease, 5-year survival rates exceed 50% in the absence of invasion into adjacent organs or distant metastases.[5]
Risk Factors
Overall, the risk of anal cancer is rising due to increased incidence of human papillomavirus (HPV) infection.[6,7] Ninety-five percent of anal cancers are HPV related, with the highest risk for serotypes 16 and 18. Involvement of HPV can be pathologically correlated with P16+ staining.[8] Patients with HIV have a higher risk of HPV coinfection, and consequently have a higher risk of anal cancer.
Data suggest that certain sexual practices, such as receptive anal intercourse or a high lifetime number of sexual partners, portend an increased risk of anal cancer. These practices may have led to an increase in the number of individuals at risk of infection with HPV.[6]
References
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
Ajani JA, Winter KA, Gunderson LL, et al.: Prognostic factors derived from a prospective database dictate clinical biology of anal cancer: the intergroup trial (RTOG 98-11). Cancer 116 (17): 4007-13, 2010. [PUBMED Abstract]
Klas JV, Rothenberger DA, Wong WD, et al.: Malignant tumors of the anal canal: the spectrum of disease, treatment, and outcomes. Cancer 85 (8): 1686-93, 1999. [PUBMED Abstract]
Touboul E, Schlienger M, Buffat L, et al.: Epidermoid carcinoma of the anal canal. Results of curative-intent radiation therapy in a series of 270 patients. Cancer 73 (6): 1569-79, 1994. [PUBMED Abstract]
Anus. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 275–84.
Johnson LG, Madeleine MM, Newcomer LM, et al.: Anal cancer incidence and survival: the surveillance, epidemiology, and end results experience, 1973-2000. Cancer 101 (2): 281-8, 2004. [PUBMED Abstract]
Holly EA, Ralston ML, Darragh TM, et al.: Prevalence and risk factors for anal squamous intraepithelial lesions in women. J Natl Cancer Inst 93 (11): 843-9, 2001. [PUBMED Abstract]
Ryan DP, Compton CC, Mayer RJ: Carcinoma of the anal canal. N Engl J Med 342 (11): 792-800, 2000. [PUBMED Abstract]
Cellular Classification of Anal Cancer
Squamous cell (epidermoid) carcinomas make up most primary anal cancers. Historically, a subset of tumors arising from the epithelial transitional zone were categorized as cloacogenic or basaloid tumors. However, these tumors are now recognized as nonkeratinizing squamous cell cancers and are similarly associated with human papillomavirus.[1,2]
Lesions in the hair-bearing skin distal to the squamous mucocutaneous junction are defined as perianal cancers. These are typically treated the same as anal canal cancers, although local therapy alone can be considered for discrete skin lesions with significant separation from the anal verge.
Adenocarcinomas starting in anal glands or fistulae formation are rare and generally have clinical features that are similar to rectal adenocarcinoma. For more information, see the Clinical Features section in Rectal Cancer Treatment.
Treatment of anal melanoma is not included in this summary.
References
Palefsky JM, Holly EA, Gonzales J, et al.: Detection of human papillomavirus DNA in anal intraepithelial neoplasia and anal cancer. Cancer Res 51 (3): 1014-9, 1991. [PUBMED Abstract]
Pirog EC, Quint KD, Yantiss RK: P16/CDKN2A and Ki-67 enhance the detection of anal intraepithelial neoplasia and condyloma and correlate with human papillomavirus detection by polymerase chain reaction. Am J Surg Pathol 34 (10): 1449-55, 2010. [PUBMED Abstract]
Stage Information for Anal Cancer
The anal canal extends from the rectum to the perianal skin and is lined by a mucous membrane that covers the internal sphincter. Tumors of the anal margin (below the anal verge and involving the perianal hair-bearing skin) are classified with skin tumors.
American Joint Committee on Cancer (AJCC) Stage Groupings and TNM Definitions
The following is a staging system for anal canal cancer that has been described by the AJCC and the International Union Against Cancer.[1] The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define anal cancer.
Table 1. Definitions of TNM Stage 0a
Stage
TNM
Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Anus. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 275–84.
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Anus. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 275–84.
I
T1, N0, M0
T1 = Tumor ≤2 cm.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 3. Definitions of TNM Stages IIA and IIBa
Stage
TNM
Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Anus. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 275–84.
IIA
T2, N0, M0
T2 = Tumor >2 cm but ≤5 cm.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
IIB
T3, N0, M0
T3 = Tumor >5 cm.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 4. Definitions of TNM Stages IIIA, IIIB, and IIICa
Stage
TNM
Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Anus. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 275–84.
IIIA
T1, N1, M0
T1 = Tumor ≤2 cm.
N1 = Metastasis in inguinal, mesorectal, internal iliac, or external iliac nodes.
M0 = No distant metastasis.
T2, N1, M0
T2 = Tumor >2 cm but ≤5 cm.
N1 = Metastasis in inguinal, mesorectal, internal iliac, or external iliac nodes.
M0 = No distant metastasis.
IIIB
T4, N0, M0
T4 = Tumor of any size invading adjacent organ(s), such as the vagina, urethra, or bladder.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
IIIC
T3, N1, M0
T3 = Tumor >5 cm.
N1 = Metastasis in inguinal, mesorectal, internal iliac, or external iliac nodes.
M0 = No distant metastasis.
T4, N1, M0
T4 = Tumor of any size invading adjacent organ(s), such as the vagina, urethra, or bladder.
N1 = Metastasis in inguinal, mesorectal, internal iliac, or external iliac nodes.
M0 = No distant metastasis.
Table 5. Definitions of Stage IVa
Stage
TNM
Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Anus. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 275–84.
The optimal approach in patients with advanced disease is still under clinical evaluation. Information about ongoing clinical trials is available from the NCI website.
Capecitabine and Fluorouracil Dosing
The DPYD gene encodes an enzyme that catabolizes pyrimidines and fluoropyrimidines, like capecitabine and fluorouracil. An estimated 1% to 2% of the population has germline pathogenic variants in DPYD, which lead to reduced DPD protein function and an accumulation of pyrimidines and fluoropyrimidines in the body.[1,2] Patients with the DPYD*2A variant who receive fluoropyrimidines may experience severe, life-threatening toxicities that are sometimes fatal. Many other DPYD variants have been identified, with a range of clinical effects.[1–3] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[4–6] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[7] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[8]
References
Sharma BB, Rai K, Blunt H, et al.: Pathogenic DPYD Variants and Treatment-Related Mortality in Patients Receiving Fluoropyrimidine Chemotherapy: A Systematic Review and Meta-Analysis. Oncologist 26 (12): 1008-1016, 2021. [PUBMED Abstract]
Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
Shakeel F, Fang F, Kwon JW, et al.: Patients carrying DPYD variant alleles have increased risk of severe toxicity and related treatment modifications during fluoropyrimidine chemotherapy. Pharmacogenomics 22 (3): 145-155, 2021. [PUBMED Abstract]
Amstutz U, Henricks LM, Offer SM, et al.: Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther 103 (2): 210-216, 2018. [PUBMED Abstract]
Henricks LM, Lunenburg CATC, de Man FM, et al.: DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol 19 (11): 1459-1467, 2018. [PUBMED Abstract]
Lau-Min KS, Varughese LA, Nelson MN, et al.: Preemptive pharmacogenetic testing to guide chemotherapy dosing in patients with gastrointestinal malignancies: a qualitative study of barriers to implementation. BMC Cancer 22 (1): 47, 2022. [PUBMED Abstract]
Brooks GA, Tapp S, Daly AT, et al.: Cost-effectiveness of DPYD Genotyping Prior to Fluoropyrimidine-based Adjuvant Chemotherapy for Colon Cancer. Clin Colorectal Cancer 21 (3): e189-e195, 2022. [PUBMED Abstract]
Baker SD, Bates SE, Brooks GA, et al.: DPYD Testing: Time to Put Patient Safety First. J Clin Oncol 41 (15): 2701-2705, 2023. [PUBMED Abstract]
Treatment of Stage 0 Anal Cancer
Treatment Options for Stage 0 Anal Cancer
Stage 0 anal cancer is carcinoma in situ. Rarely diagnosed, it is a very early cancer that has not spread below the limiting membrane of the first layer of anal tissue.
Surgical resection is used to treat lesions of the perianal area not involving the anal sphincter. The surgical approach depends on the location of the lesion in the anal canal.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Treatment of Stages I, II, and III Anal Cancer
Treatment Options for Stages I, II, and III Anal Cancer
Current sphincter-sparing therapies include wide local excision for small tumors of the perianal skin or anal margin, or definitive chemoradiation therapy (fluorouracil [5-FU] and mitomycin) for cancers of the anal canal. Radical resection is reserved for patients with incomplete responses or recurrent disease.
Continued surveillance with rectal examination every 3 months for the first 2 years and endoscopy with biopsy when indicated after completion of sphincter-preserving therapy is important to monitor for recurrence.
Small tumors of the perianal skin or anal margin not involving the anal sphincter may be adequately treated with local resection.[1]
The standard of care for all other stage I, II, and III anal cancers in appropriate patients is chemoradiation therapy (external-beam radiation therapy [EBRT] with chemotherapy).
Alternative strategies such as radiation therapy alone or surgery alone may be considered, depending on the clinical context.
Radical resection is reserved for residual or recurrent cancer in the anal canal after nonoperative therapy.
Chemoradiation therapy
Because of historically high rates of recurrence with colostomy alone, chemoradiation therapy is the preferred approach for patients with anal cancer in the absence of distant metastases.
Evidence (chemoradiation therapy):
The Anal Cancer Trial (ACT I) from the United Kingdom Co-ordinating Committee on Cancer Research demonstrated the superiority of chemoradiation with 5-FU and mitomycin over radiation therapy alone with regard to local failure and deaths from anal cancer.[2,8][Level of evidence A1]
In this prospective trial, 585 patients were randomly assigned to receive 45 Gy of radiation in 20 or 25 fractions with or without 5-FU. The 5-FU was given by continuous infusion (750 mg/m2 for 5 days or 1,000 mg/m2 for 4 days) during the first and final weeks of radiation therapy, along with a single dose of mitomycin (12 mg/m2) on the first day.
After a median follow-up of 13.1 years, patients who received chemoradiation therapy had a reduction in local failure (36% vs. 59%; hazard ratio [HR], 0.46; 95% confidence interval [CI], 0.35−0.60; P < .001), risk of death from anal cancer (HR, 0.61; 95% CI, 0.49−0.76; P < .001), and relapse at 12 years (17.7% vs. 29.7%; HR, 0.70; 95% CI, 9.58−0.84; P < .001).[2][Level of evidence A1]
There was no significant difference in overall survival (OS) in this trial (HR, 0.86; 95% CI, 0.70−1.04; P = .12).
An initial 9.1% increase in non–anal cancer deaths was observed in the first 5 years after chemoradiation therapy but was not seen at 10 years.
A European Organisation for Research and Treatment of Cancer (EORTC) trial prospectively randomly assigned 100 patients with T3 to T4 or N1 to N3 disease to receive 45 Gy of radiation with a 15-Gy or 30-Gy boost with or without 5-FU infusion (750 mg/m2 for 5 days starting on days 1 and 29) plus mitomycin (15 mg/m2 on day 1).[3][Level of evidence B1]
Outcomes favored chemoradiation therapy with respect to 5-year colostomy-free survival rates (75% vs. 48%; P = .002) and 5-year progression-free survival (PFS) rates (60% vs. 48%; P = .05).
Subsequent trials have found capecitabine to be a reasonable replacement for 5-FU in combination with mitomycin and radiation therapy.[4,5]
While the ACT I and EORTC randomized trials established chemoradiation therapy as the preferred approach for nonmetastatic anal cancer, the substantial hematological, renal, and pulmonary toxicity of mitomycin has prompted studies of alternative regimens.
A Radiation Therapy Oncology Group (RTOG)/Eastern Cooperative Oncology Group trial of 310 patients studied chemoradiation therapy (5-FU infusion + 45 Gy of radiation) with or without mitomycin.
After 4-years of follow-up, patients who received mitomycin had an improved colostomy-free survival rate (71% vs. 59%; P = .014) and disease-free survival (DFS) rate (73% vs. 51%; P = .0003).[9][Level of evidence B1]
Two large intergroup trials studied the substitution of cisplatin for mitomycin, with differing conclusions.
In a phase III U.S. Intergroup trial (RTOG 9811 [NCT00003596]), patients in the cisplatin arm received two cycles of induction 5-FU and cisplatin before receiving concurrent chemoradiation therapy with 5-FU and cisplatin.[6]
Patients who received mitomycin had improved local control and an improved colostomy-free survival rate (90% vs. 81%; P = .02). Subsequent long-term follow-up demonstrated a borderline significant difference in the 5-year colostomy-free survival rate (71.9% vs. 65%; P = .05).[10]
Long-term follow-up also demonstrated a superior 5-year DFS rate (67.8% vs. 57.8%; P = .006) and OS rate (78.3% vs. 70.7%; P = .074) for patients who received mitomycin.[11][Level of evidence B1]
One potential explanation for the inferiority of cisplatin in this study was the delay in time to radiation therapy during induction chemotherapy.
In the prospective randomized ACT II trial, 940 patients were assigned in a 2 × 2 factorial design to receive the following: (1) either mitomycin or cisplatin during induction chemoradiation therapy and (2) either maintenance therapy with 5-FU and cisplatin in weeks 11 and 14 or no maintenance therapy.[7]
The complete remission rate was equivalent in patients who received mitomycin or cisplatin after a median follow-up of 5.1 years (90.5% vs. 89.6%; 95% CI, -4.9 to 3.1; P = .64). The 3-year PFS rate was also equivalent in both study groups (73% for mitomycin vs. 72% for cisplatin; HR, 0.95; 95% CI, 0.75−1.19; P = .063).[7][Level of evidence B1]
There was also no significant effect on 3-year PFS rates among patients who received maintenance therapy or no maintenance therapy (74% vs. 73%; HR, 0.95; 95% CI, 0.75−1.21; P = .70).
This study suggests that cisplatin might reasonably substitute for mitomycin in a chemoradiation strategy.
The best time to assess a complete clinical response after chemoradiation therapy is generally after 26 weeks because delayed responses are seen.[12] Residual disease or subsequent local recurrence require further treatment.
The standard salvage therapy for patients with either gross or microscopic residual disease after chemoradiation therapy has been abdominoperineal resection. Alternatively, patients may be treated with additional salvage chemoradiation therapy, chemotherapy alone, or immunotherapy.[12,13]
The optimal radiation dose in various situations has not been determined. There is insufficient evidence to determine whether the dose should be escalated for patients with T3 to T4 disease or nodal metastases, or potentially de-escalated for patients with early-stage tumors smaller than 1 cm. It is also unclear whether the chemotherapy backbone can be safely omitted for some patients with early-stage tumors, and whether such a strategy would affect the optimal dose of radiation. The roles for newer strategies such as intensity-modulated radiation therapy, proton beam therapy, and brachytherapy have yet to be conclusively determined.[14–16] Based on the National Cancer Database, higher volume radiation oncology centers report improved OS for patients with anal cancer.[17]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Enker WE, Heilwell M, Janov AJ, et al.: Improved survival in epidermoid carcinoma of the anus in association with preoperative multidisciplinary therapy. Arch Surg 121 (12): 1386-90, 1986. [PUBMED Abstract]
Northover J, Glynne-Jones R, Sebag-Montefiore D, et al.: Chemoradiation for the treatment of epidermoid anal cancer: 13-year follow-up of the first randomised UKCCCR Anal Cancer Trial (ACT I). Br J Cancer 102 (7): 1123-8, 2010. [PUBMED Abstract]
Bartelink H, Roelofsen F, Eschwege F, et al.: Concomitant radiotherapy and chemotherapy is superior to radiotherapy alone in the treatment of locally advanced anal cancer: results of a phase III randomized trial of the European Organization for Research and Treatment of Cancer Radiotherapy and Gastrointestinal Cooperative Groups. J Clin Oncol 15 (5): 2040-9, 1997. [PUBMED Abstract]
Goodman KA, Julie D, Cercek A, et al.: Capecitabine With Mitomycin Reduces Acute Hematologic Toxicity and Treatment Delays in Patients Undergoing Definitive Chemoradiation Using Intensity Modulated Radiation Therapy for Anal Cancer. Int J Radiat Oncol Biol Phys 98 (5): 1087-1095, 2017. [PUBMED Abstract]
Meulendijks D, Dewit L, Tomasoa NB, et al.: Chemoradiotherapy with capecitabine for locally advanced anal carcinoma: an alternative treatment option. Br J Cancer 111 (9): 1726-33, 2014. [PUBMED Abstract]
Ajani JA, Winter KA, Gunderson LL, et al.: Fluorouracil, mitomycin, and radiotherapy vs fluorouracil, cisplatin, and radiotherapy for carcinoma of the anal canal: a randomized controlled trial. JAMA 299 (16): 1914-21, 2008. [PUBMED Abstract]
James RD, Glynne-Jones R, Meadows HM, et al.: Mitomycin or cisplatin chemoradiation with or without maintenance chemotherapy for treatment of squamous-cell carcinoma of the anus (ACT II): a randomised, phase 3, open-label, 2 × 2 factorial trial. Lancet Oncol 14 (6): 516-24, 2013. [PUBMED Abstract]
Epidermoid anal cancer: results from the UKCCCR randomised trial of radiotherapy alone versus radiotherapy, 5-fluorouracil, and mitomycin. UKCCCR Anal Cancer Trial Working Party. UK Co-ordinating Committee on Cancer Research. Lancet 348 (9034): 1049-54, 1996. [PUBMED Abstract]
Flam M, John M, Pajak TF, et al.: Role of mitomycin in combination with fluorouracil and radiotherapy, and of salvage chemoradiation in the definitive nonsurgical treatment of epidermoid carcinoma of the anal canal: results of a phase III randomized intergroup study. J Clin Oncol 14 (9): 2527-39, 1996. [PUBMED Abstract]
Eng C, Ciombor KK, Cho M, et al.: Anal Cancer: Emerging Standards in a Rare Disease. J Clin Oncol 40 (24): 2774-2788, 2022. [PUBMED Abstract]
Gunderson LL, Winter KA, Ajani JA, et al.: Long-term update of US GI intergroup RTOG 98-11 phase III trial for anal carcinoma: survival, relapse, and colostomy failure with concurrent chemoradiation involving fluorouracil/mitomycin versus fluorouracil/cisplatin. J Clin Oncol 30 (35): 4344-51, 2012. [PUBMED Abstract]
Pedersen TB, Gocht-Jensen P, Klein MF: 30-day and long-term outcome following salvage surgery for squamous cell carcinoma of the anus. Eur J Surg Oncol 44 (10): 1518-1521, 2018. [PUBMED Abstract]
Guerra GR, Kong JC, Bernardi MP, et al.: Salvage Surgery for Locoregional Failure in Anal Squamous Cell Carcinoma. Dis Colon Rectum 61 (2): 179-186, 2018. [PUBMED Abstract]
Cordoba A, Escande A, Leroy T, et al.: Low-dose-rate interstitial brachytherapy boost for the treatment of anal canal cancers. Brachytherapy 16 (1): 230-235, 2017 Jan – Feb. [PUBMED Abstract]
Call JA, Prendergast BM, Jensen LG, et al.: Intensity-modulated Radiation Therapy for Anal Cancer: Results From a Multi-Institutional Retrospective Cohort Study. Am J Clin Oncol 39 (1): 8-12, 2016. [PUBMED Abstract]
Gryc T, Ott O, Putz F, et al.: Interstitial brachytherapy as a boost to patients with anal carcinoma and poor response to chemoradiation: Single-institution long-term results. Brachytherapy 15 (6): 865-872, 2016 Nov – Dec. [PUBMED Abstract]
Amini A, Jones BL, Ghosh D, et al.: Impact of facility volume on outcomes in patients with squamous cell carcinoma of the anal canal: Analysis of the National Cancer Data Base. Cancer 123 (2): 228-236, 2017. [PUBMED Abstract]
In the multicenter, randomized, phase II International Advanced Anal Cancer InterAACT trial (NCT02560298), carboplatin (area under the curve 5) and weekly paclitaxel was compared with standard infusional 5-FU and bolus cisplatin in patients with advanced-stage anal cancer.[2]
With a median follow-up of 25.3 months, the median overall survival (OS) with carboplatin and paclitaxel was improved compared with cisplatin and 5-FU (20 months vs. 12.3 months; hazard ratio [HR], 2.0; P = .014).[Level of evidence A1]
Serious adverse events were more common in patients treated with cisplatin plus 5-FU (62% vs. 36%; P = .016).
These promising findings have led international investigators to use carboplatin and paclitaxel as a new backbone in trials for patients with advanced-stage disease, as well as a potential partner for use with radiation therapy. Other chemotherapy regimens, such as modified docetaxel, cisplatin, and 5-FU, are under clinical evaluation.[3]
The checkpoint inhibitors have also shown activity for patients with metastatic disease. The phase II NCI96773 trial (NCT02314169) of single-agent nivolumab (3 mg/kg every 2 weeks) enrolled 37 patients.[4]
The overall response rate was 24%, including two complete responses.[4][Level of evidence C3]
The phase Ib KEYNOTE-028 trial (NCT02054806) for patients with advanced tumors with programmed death ligand-1 of at least 1% enrolled a cohort of 24 patients with anal squamous cell carcinoma.[5]
The overall response rate was 17%, and an additional stable disease rate was 42%.[5][Level of evidence C3]
Although there is no clear standard of care for patients with metastatic disease, recent studies are uncovering promising new avenues for systemic treatment. Palliation of symptoms from the primary lesion is important. Patients with stage IV disease should strongly consider enrolling in clinical trials.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
James RD, Glynne-Jones R, Meadows HM, et al.: Mitomycin or cisplatin chemoradiation with or without maintenance chemotherapy for treatment of squamous-cell carcinoma of the anus (ACT II): a randomised, phase 3, open-label, 2 × 2 factorial trial. Lancet Oncol 14 (6): 516-24, 2013. [PUBMED Abstract]
Rao S, Sclafani F, Eng C, et al.: International Rare Cancers Initiative Multicenter Randomized Phase II Trial of Cisplatin and Fluorouracil Versus Carboplatin and Paclitaxel in Advanced Anal Cancer: InterAAct. J Clin Oncol 38 (22): 2510-2518, 2020. [PUBMED Abstract]
Kim S, François E, André T, et al.: Docetaxel, cisplatin, and fluorouracil chemotherapy for metastatic or unresectable locally recurrent anal squamous cell carcinoma (Epitopes-HPV02): a multicentre, single-arm, phase 2 study. Lancet Oncol 19 (8): 1094-1106, 2018. [PUBMED Abstract]
Morris VK, Salem ME, Nimeiri H, et al.: Nivolumab for previously treated unresectable metastatic anal cancer (NCI9673): a multicentre, single-arm, phase 2 study. Lancet Oncol 18 (4): 446-453, 2017. [PUBMED Abstract]
Ott PA, Piha-Paul SA, Munster P, et al.: Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with recurrent carcinoma of the anal canal. Ann Oncol 28 (5): 1036-1041, 2017. [PUBMED Abstract]
Treatment of HIV and Anal Cancer
The tolerance of patients with HIV and anal carcinoma to standard fluorouracil and mitomycin chemoradiation therapy is not well defined.[1,2] In general, patients with HIV are treated similarly to other patients and have similar outcomes, particularly in the era of highly active antiretroviral therapy (HAART). Patients with pretreatment CD4 counts of fewer than 200 cells/μl may have increased acute and late toxic effects.[3,4] Therefore, patients with a history of AIDS-related complications may have difficulty tolerating a standard regimen, necessitating a dose adjustment or omission of mitomycin.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Holland JM, Swift PS: Tolerance of patients with human immunodeficiency virus and anal carcinoma to treatment with combined chemotherapy and radiation therapy. Radiology 193 (1): 251-4, 1994. [PUBMED Abstract]
Peddada AV, Smith DE, Rao AR, et al.: Chemotherapy and low-dose radiotherapy in the treatment of HIV-infected patients with carcinoma of the anal canal. Int J Radiat Oncol Biol Phys 37 (5): 1101-5, 1997. [PUBMED Abstract]
Hoffman R, Welton ML, Klencke B, et al.: The significance of pretreatment CD4 count on the outcome and treatment tolerance of HIV-positive patients with anal cancer. Int J Radiat Oncol Biol Phys 44 (1): 127-31, 1999. [PUBMED Abstract]
Place RJ, Gregorcyk SG, Huber PJ, et al.: Outcome analysis of HIV-positive patients with anal squamous cell carcinoma. Dis Colon Rectum 44 (4): 506-12, 2001. [PUBMED Abstract]
Treatment of Recurrent Anal Cancer
Local recurrences and persistent disease after treatment with radiation therapy and chemotherapy or surgery as the primary treatment may be controlled by using the alternate treatment (surgical resection after radiation and vice versa).[1] Salvage chemoradiation therapy with fluorouracil and cisplatin plus a radiation boost may avoid permanent colostomy in patients with residual tumor after initial nonoperative therapy.[2] Clinical trials are exploring the use of radiation therapy with chemotherapy and radiosensitizers to improve local control.
Preliminary studies in patients with stage IV disease suggest that alternative chemotherapy regimens (such as carboplatin and paclitaxel in the InterACCT trial [NCT02560298]) or immune checkpoint inhibitors (as in NCI9673 [NCT02314169] and KEYNOTE-028 [NCT02054806]) may be beneficial in this setting.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Longo WE, Vernava AM, Wade TP, et al.: Recurrent squamous cell carcinoma of the anal canal. Predictors of initial treatment failure and results of salvage therapy. Ann Surg 220 (1): 40-9, 1994. [PUBMED Abstract]
Flam M, John M, Pajak TF, et al.: Role of mitomycin in combination with fluorouracil and radiotherapy, and of salvage chemoradiation in the definitive nonsurgical treatment of epidermoid carcinoma of the anal canal: results of a phase III randomized intergroup study. J Clin Oncol 14 (9): 2527-39, 1996. [PUBMED Abstract]
Latest Updates to This Summary (02/12/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 1).
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of anal cancer. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Anal Cancer Treatment are:
Amit Chowdhry, MD, PhD (University of Rochester Medical Center)
Leon Pappas, MD, PhD (Massachusetts General Hospital)
Ari Seifter, MD (Advocate Health Care)
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Anal Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/anal/hp/anal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389221]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
Leaving ChemoPrescribe
You are now leaving ChemoPrescribe website.
To return to the website, click on the back arrow on your browser.