Childhood Nasopharyngeal Cancer (PDQ®)–Patient Version

Childhood Nasopharyngeal Cancer (PDQ®)–Patient Version

What is childhood nasopharyngeal cancer?

Childhood nasopharyngeal cancer is a rare type of cancer that forms in the tissue lining the nasopharynx. The nasopharynx is the upper part of the throat behind the nose. The nostrils allow air into the nasopharynx. Air and food pass through the pharynx on the way to the trachea or esophagus.

Nasopharyngeal cancer is more common in children aged 10 to 19 years than in children younger than 10 years.

EnlargeAnatomy of the nasopharynx; drawing shows the three parts of the pharynx (throat): the nasopharynx, oropharynx, and hypopharynx. Also shown are the nasal cavity, oral cavity, larynx, esophagus, and trachea.
Anatomy of the nasopharynx. The nasopharynx is in the upper part of the pharynx (throat) behind the nose. The nostrils lead into the nasopharynx. An opening on each side of the nasopharynx leads into the ear.

Causes and risk factors for childhood nasopharyngeal cancer

Childhood nasopharyngeal cancer is caused by certain changes in the way the cells in the nasal cavity and top part of the throat function, especially how they grow and divide into new cells. Often, the exact cause of these 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. A known risk factor for childhood nasopharyngeal cancer is infection from the Epstein-Barr virus (EBV). Not every child with this risk factor will develop nasopharyngeal cancer. And it will develop in some children who don’t have a known risk factor. Talk with your child’s doctor if you think your child may be at risk.

Symptoms of childhood nasopharyngeal cancer

Children may not have symptoms of nasopharyngeal cancer until the tumor has grown bigger. It’s important to check with your child’s doctor if your child has:

  • a headache
  • a blocked or stuffy nose
  • nosebleeds
  • an earache
  • an ear infection
  • hearing loss
  • problems moving the jaw
  • trouble speaking
  • trouble seeing or a droopy eyelid
  • lumps in the neck that may be painful

These symptoms may be caused by problems other than nasopharyngeal cancer. The only way to know is for your child to see a doctor.

Tests to diagnose childhood nasopharyngeal cancer

If your child has symptoms that suggest nasopharyngeal cancer, the doctor will need to find out if these are due to cancer or another problem. The doctor will ask when the symptoms started and how often your child has been having them. They will also ask about your child’s personal and family medical history and do a physical exam. Depending on these results, the doctor may recommend other tests. If your child is diagnosed with nasopharyngeal cancer, the results of these tests will help plan treatment.

Diagnostic tests

The tests used to diagnose nasopharyngeal cancer may include:

Magnetic resonance imaging (MRI)

MRI uses a magnet, radio waves, and a computer to make a series of detailed pictures of areas of the body, such as the head and neck. This procedure is also called nuclear magnetic resonance imaging (NMRI).

Nasal endoscopy

Nasal endoscopy is a procedure to look at organs and tissues inside the body to check for abnormal areas. A flexible or rigid endoscope is inserted through the nose. An endoscope is a thin, tube-like instrument with a light and a lens for viewing. It may also have a tool to remove tissue samples, which are checked under a microscope by a pathologist for signs of disease.

Epstein-Barr virus (EBV) tests

Epstein-Barr virus (EBV) tests use a sample of blood to check for antibodies to the Epstein-Barr virus and DNA markers of the Epstein-Barr virus. These are found in the blood of patients who have been infected with EBV.

Tests to stage nasopharyngeal cancer

If your child is diagnosed with nasopharyngeal cancer, they will be referred to a pediatric oncologist. This is a doctor who specializes in staging and treating nasopharyngeal cancer and other cancers. They will recommend tests to determine the extent (stage) of cancer. Sometimes the cancer is only in the nasopharynx. Or, it may have spread to other parts of the body. The process of learning the extent of cancer in the body is called staging. It is important to know the stage of the nasopharyngeal cancer in order to plan the best treatment.

Most children with nasopharyngeal cancer are at an advanced stage at the time of diagnosis. Nasopharyngeal cancer spreads most often to the bone, lung, and liver.

For information about a specific stage of nasopharyngeal cancer, see Childhood Nasopharyngeal Cancer Stages.

The following procedures may be used to determine the nasopharyngeal cancer stage:

Neurological exam

A neurologic exam is a series of questions and tests done 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.

Chest x-ray

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.

PET-CT scan

PET-CT scan combines the pictures from a positron emission tomography (PET) scan and a computed tomography (CT) scan. The PET and CT scans are done at the same time on the same machine. The combined scans make more detailed pictures than either test would make by itself.

  • For the PET scan, a small amount of radioactive sugar (also called radioactive glucose) is injected into a vein. The PET scanner rotates around the body and makes a picture of where sugar is being used in the body. Cancer cells show up brighter in the picture because they are more active and take up more sugar than normal cells do.
  • For the CT scan (CAT scan), a computer linked to an x-ray machine makes a series of detailed pictures of areas inside the body. 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. Often, a chest CT scan is done. This procedure is also called computed tomography, computerized tomography, or computerized axial tomography. Learn more about Computed Tomography (CT) Scans and Cancer.

Getting a second opinion

You may want to get a second opinion to confirm your child’s 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. This doctor may agree with the first doctor, suggest changes to the treatment plan, or provide more information about your child’s cancer.

To learn more about choosing a doctor and getting a second opinion, visit 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, visit Questions to Ask Your Doctor about Cancer.

Stages of childhood nasopharyngeal cancer

The 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 nasopharyngeal cancer to plan the best treatment.

There are several staging systems for cancer. Nasopharyngeal cancer staging usually uses the TNM staging system. You may see your child’s cancer described by this staging system in the 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 child’s cancer. When talking with you about your child’s cancer, your child’s doctor may describe it as one of these stages.

Learn more about Cancer Staging.

Learn about the tests and procedures used to determine the stage of nasopharyngeal cancer. 

Stage 0 nasopharyngeal cancer

In stage 0, abnormal cells are found in the lining of the nasopharynx. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called carcinoma in situ.

Stage I nasopharyngeal cancer

In stage I, nasopharyngeal cancer has formed and is found in the nasopharynx only or has spread from the nasopharynx to the oropharynx and/or nasal cavity.

EnlargeDrawing shows different sizes of a tumor in centimeters (cm) compared to the size of a pea (1 cm), a peanut (2 cm), a grape (3 cm), a walnut (4 cm), a lime (5 cm), an egg (6 cm), a peach (7 cm), and a grapefruit (10 cm). Also shown is a 10-cm ruler and a 4-inch ruler.
Tumor 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 II nasopharyngeal cancer

Stage II is based on the location of the cancer and whether it has spread. A child may have one of the following:

  • Cancer has spread to one or more lymph nodes on one side of the neck and/or to one or more lymph nodes on one or both sides of the back of the throat. The affected lymph nodes are 6 centimeters or smaller. Cancer is found:
    • in the nasopharynx only or has spread from the nasopharynx to the oropharynx and/or to the nasal cavity; or
    • only in the lymph nodes in the neck. The cancer cells in the lymph nodes are infected with Epstein-Barr virus (a virus linked to nasopharyngeal cancer). Cancer was not found in the nasopharynx.
  • Cancer has spread to the parapharyngeal space and/or nearby muscles. Cancer may have also spread to one or more lymph nodes on one side of the neck and/or to one or more lymph nodes on one or both sides of the back of the throat. The affected lymph nodes are 6 centimeters or smaller.

Stage III nasopharyngeal cancer

Stage III is based on the location of the cancer and whether it has spread. A child may have one of the following:

  • Cancer has spread to one or more lymph nodes on both sides of the neck. The affected lymph nodes are 6 centimeters or smaller. Cancer is found:
    • in the nasopharynx only or has spread from the nasopharynx to the oropharynx and/or to the nasal cavity; or
    • only in the lymph nodes in the neck. The cancer cells in the lymph nodes are infected with Epstein-Barr virus (a virus linked to nasopharyngeal cancer). Cancer was not found in the nasopharynx.
  • Cancer has spread to the parapharyngeal space and/or nearby muscles. Cancer has also spread to one or more lymph nodes on both sides of the neck. The affected lymph nodes are 6 centimeters or smaller.
  • Cancer has spread to the bones at the bottom of the skull, the bones in the neck, jaw muscles, and/or the sinuses around the nose and eyes. Cancer may have also spread to one or more lymph nodes on one or both sides of the neck and/or the back of the throat. The affected lymph nodes are 6 centimeters or smaller.

Stage IV nasopharyngeal cancer

Stage IV is subdivided based on whether the cancer has spread to other tissue.

  • In stage IVA:
    • Cancer has spread to the brain, the cranial nerves, the hypopharynx, the salivary gland in front of the ear, the bone around the eye, and/or the soft tissues of the jaw. Cancer may have also spread to one or more lymph nodes on one or both sides of the neck and/or the back of the throat. The affected lymph nodes are 6 centimeters or smaller; or
    • Cancer has spread to one or more lymph nodes on one or both sides of the neck. The affected lymph nodes are larger than 6 centimeters and/or are found in the lowest part of the neck.
  • In stage IVB: Cancer has spread beyond the lymph nodes in the neck to distant lymph nodes, such as those between the lungs, below the collarbone, or in the armpit or groin, or to other parts of the body, such as the lung, bone, or liver.

Stage IV nasopharyngeal cancer is also called metastatic nasopharyngeal 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 nasopharyngeal cancer spreads to the lung, the cancer cells in the lung are actually nasopharyngeal cancer cells. The disease is called metastatic nasopharyngeal cancer, not lung cancer. Learn more in Metastatic Cancer: When Cancer Spreads. 

Recurrent nasopharyngeal cancer

Recurrent nasopharyngeal cancer is cancer that has come back after it has been treated. Nasopharyngeal cancer may come back in the nasopharynx, or in other parts of the body, such as the bone, lung, or liver. Tests will be done to help determine where the cancer has returned in the body, if it has spread, and how far. The type of treatment that your child will have for recurrent nasopharyngeal cancer will depend on where it has come back.  

Learn more in Recurrent Cancer: When Cancer Comes Back.

Types of treatment for childhood nasopharyngeal cancer

Who treats children with nasopharyngeal cancer?

A pediatric oncologist, a doctor who specializes in treating children with cancer, oversees treatment of nasopharyngeal cancer. The pediatric oncologist works with other health care providers who are experts in treating children with cancer and who specialize in certain areas of medicine. Other specialists may include:

Treatment options

There are different types of treatment for children and adolescents with nasopharyngeal cancer. You and your child’s care team will work together to decide treatment. Many factors will be considered, such as your child’s overall health and whether the cancer is newly diagnosed or has come back.

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 care team before treatment begins about what to expect. For help every step of the way, visit our booklet, Children with Cancer: A Guide for Parents.

The types of treatment your child might have include:

Chemotherapy

Chemotherapy (also called chemo) uses drugs to stop the growth of cancer cells. Chemotherapy either kills the cancer cells or stops them from dividing. Chemotherapy may be given alone or with other types of treatment, such as radiation therapy.

Chemotherapy for nasopharyngeal cancer is injected into a vein. When given this way, the drugs enter the bloodstream to reach cancer cells throughout the body. Chemotherapy used alone or in combination to treat nasopharyngeal cancer in children include:

Other chemotherapy not listed here may also be used. 

Learn more about how chemotherapy works, how it is given, common side effects, and more at Chemotherapy to Treat Cancer.

Radiation therapy

Radiation therapy uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. Nasopharyngeal cancer is treated with external beam radiation therapy. This type of radiation therapy uses a machine outside the body to send radiation toward the area of the body with cancer. Radiation therapy may be given alone or with other treatments, such as chemotherapy.

Learn more about External Beam Radiation Therapy for Cancer and Radiation Therapy Side Effects. 

Surgery

Surgery to remove the tumor is done if the tumor has not spread throughout the nasal cavity and throat at the time of diagnosis.

Immunotherapy

Immunotherapy uses a person’s immune system to fight cancer. Interferon may be used to treat nasopharyngeal cancer.

Learn more about Immunotherapy to Treat Cancer.

Clinical trials

For some children, joining a clinical trial may be an option. There are different types of clinical trials for childhood 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 child’s age, and where the trials are being done. Clinical trials supported by other organizations can be found on the ClinicalTrials.gov website.

Learn more about clinical trials, including how to find and join one, at Clinical Trials Information for Patients and Caregivers.

Treatment of childhood nasopharyngeal cancer

Treatment of newly diagnosed nasopharyngeal cancer in children may include:

  • Chemotherapy followed by chemotherapy and radiation therapy given at the same time. Interferon may also be given.
  • Surgery to remove the tumor.

Treatment of refractory or recurrent nasopharyngeal cancer is 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.

Prognostic factors for childhood nasopharyngeal cancer

If your child has been diagnosed with nasopharyngeal cancer, 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 can be affected by whether the cancer has spread to other parts of the body at the time of diagnosis and whether the cancer 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.

Side effects and late effects of 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.

To learn more about side effects that begin during treatment for cancer, visit Side Effects.

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:

  • problems with the thyroid gland
  • decreased mouth function
  • hearing loss or chronic ear infections
  • dental cavities
  • chronic sinusitis
  • changes in mood, feelings, thinking, learning, or memory
  • second cancers (new types of cancer) or other problems

Some late effects may be treated or controlled. It is important to talk with your child’s doctors about the possible late effects caused by some treatments. Learn more about Late Effects of Treatment for Childhood Cancer.

Follow-up care

As your child goes through treatment, they 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 child’s condition has changed or if the cancer has recurred (come back).

Coping with your child's cancer

When your child has cancer, every member of the family needs support. Taking care of yourself during this difficult time is important. Reach out to your child’s treatment team and to people in your family and community for support. To learn more, visit Support for Families: Childhood Cancer and the booklet Children with Cancer: A Guide for Parents. 

Related Resources

About This PDQ Summary

About PDQ

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 nasopharyngeal 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 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 Nasopharyngeal Cancer. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/patient/child/nasopharyngeal-treatment-pdq. Accessed <MM/DD/YYYY>.

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.

Childhood Laryngeal Tumors (PDQ®)–Patient Version

Childhood Laryngeal Tumors (PDQ®)–Patient Version

What are childhood laryngeal tumors?

Childhood laryngeal tumors form in the voice box, also called the larynx. These tumors may be benign (which means they are not cancer) or cancer. Most laryngeal tumors in children are not cancer and do not spread to other tissues. Both types of tumors need treatment.

The most common type of benign laryngeal tumor is papillomatosis of the larynx. In this condition, papillomas (benign tumors that look like warts) form in the lining of the larynx. These tumors may block the airway and cause trouble breathing. They often come back after treatment and, in rare cases, may turn into cancer in the larynx or the lung.

The larynx is a part of the throat, between the base of the tongue and the trachea (windpipe). The larynx contains the vocal cords, which vibrate and make sound when air is directed against them. The sound echoes through the pharynx, mouth, and nose to make a person’s voice.

There are three main parts of the larynx:

  • The supraglottis is the upper part of the larynx above the vocal cords, including the epiglottis.
  • The glottis is the middle part of the larynx where the vocal cords are located.
  • The subglottis is the lower part of the larynx between the vocal cords and the trachea.
EnlargeAnatomy of the larynx; drawing shows the epiglottis, supraglottis, vocal cord, glottis, and subglottis. Also shown are the tongue, trachea, and esophagus.
Anatomy of the larynx. The larynx is a part of the throat, between the base of the tongue and the trachea. The three main parts of the larynx are the supraglottis (including the epiglottis), the glottis (including the vocal cords), and the subglottis.

Causes of childhood laryngeal tumors

Laryngeal cancer in children is caused by certain changes to the way cells in the larynx function, especially how they grow and divide into new cells. Often, the exact cause of these cell changes is unknown. Learn more about how cancer develops at What Is Cancer?

Laryngeal papillomatosis is caused by infection with low-risk HPV (human papillomavirus), most often types 6 and 11. Children can get the HPV infection that causes laryngeal papillomatosis from an infected mother during birth. Most children with an HPV infection do not develop laryngeal papillomatosis. Talk with your child’s doctor if you think your child may be at risk.

Getting the HPV vaccine can help protect against HPV infection and reduce the risk of transmitting it during childbirth. Learn more about HPV Vaccines.

Symptoms of childhood laryngeal tumors

Children may not have symptoms of a laryngeal tumor until the tumor has grown bigger. It’s important to check with your child’s doctor if your child has:

  • hoarseness or a change in the voice
  • trouble or pain when swallowing
  • trouble breathing
  • a high-pitched sound with breathing
  • a lump in the neck or throat
  • a sore throat
  • a cough that does not go away

Infants and young children with these tumors may grow slowly and not eat well or meet developmental milestones such as sitting, walking, and talking in sentences.

These symptoms may be caused by problems other than a laryngeal tumor. The only way to know is to see your child’s doctor.

Tests to diagnose childhood laryngeal tumors

If your child has symptoms that suggest a laryngeal tumor, the doctor will need to find out if these are due to cancer or some other problem. The doctor will ask when the symptoms started and how often your child has been having them. They will also ask about your child’s personal and family medical history and do a physical exam. Depending on these results, they may recommend other tests. If your child is diagnosed with a laryngeal tumor, the results of these tests will help you and your child’s doctor plan treatment.

The tests used to diagnose laryngeal tumors may include:

Oral exam

In an oral exam, a doctor or dentist checks the mouth for abnormal areas. This exam may be done while under anesthesia. The doctor or dentist will feel the entire inside of the mouth with a gloved finger and examine the area with a small long-handled mirror and lights or a fiberoptic device.

Neck and chest x-ray

An x-ray is a type of radiation that can go through the body and make pictures of the inside of the body. A neck and chest x-ray makes pictures of the organs and bones inside the neck and chest.

Magnetic resonance imaging (MRI)

MRI uses a magnet, radio waves, and a computer to make a series of detailed pictures of areas in the body, such as the head and neck. This procedure is also called nuclear magnetic resonance imaging (NMRI).

CT scan

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. 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. Learn more about Computed Tomography (CT) Scans and Cancer.

EnlargeComputed tomography (CT) scan of the head and neck; drawing shows a child lying on a table that slides through the CT scanner, which takes a series of detailed x-ray pictures of the inside of the head and neck.
Computed tomography (CT) scan of the head and neck. The child lies on a table that slides through the CT scanner, which takes a series of detailed x-ray pictures of the inside of the head and neck.

PET scan

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 sugar is being used by the body. Cancer cells show up brighter in the pictures because they are more active and take up more sugar than normal cells do.

EnlargePositron emission tomography (PET) scan; drawing shows a child lying on table that slides through the PET scanner.
Positron emission tomography (PET) scan. The child lies on a table that slides through the PET scanner. The head rest and white strap help the child lie still. A small amount of radioactive glucose (sugar) is injected into the child’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.

Ultrasound

Ultrasound uses high-energy sound waves (ultrasound) that bounce off internal tissues or organs and make echoes. The echoes form a picture of body tissues called a sonogram.

Barium swallow

Barium swallow is a series of x-rays of the esophagus and stomach. For this procedure, the patient drinks a liquid that contains barium (a silver-white metallic compound). The barium coats the esophagus and stomach which helps them show up more clearly in x-rays. This procedure is also called an upper GI series.

Biopsy

A biopsy is a procedure in which a sample of tissue is removed from the tumor so that a pathologist can view it under a microscope to check for signs of disease, such as cancer or laryngeal papillomatosis.

The sample of tissue may be removed during a procedure, such as:

  • Laryngoscopy is a procedure in which the doctor checks the larynx (voice box) with a mirror or a laryngoscope to check for abnormal areas. A laryngoscope is a thin, tube-like instrument with a light and a lens for viewing the inside of the throat and voice box. It may also have a tool to remove tissue samples, which are checked under a microscope for signs of disease.
  • Endoscopy is a procedure to look at organs and tissues inside the body, such as the throat, esophagus, and trachea to check for abnormal areas. An endoscope is a thin, tube-like instrument with a light and a lens for viewing that is inserted through an opening in the body, such as the mouth. A special tool on the endoscope may be used to remove samples of tissue.

Human papillomavirus (HPV) test

An HPV test checks tissue samples from the biopsy for certain types of HPV infection that may be linked to laryngeal papillomatosis.

Immunohistochemistry

Immunohistochemistry uses antibodies to check for certain antigens (markers) in a sample of a patient’s cells or tissue. The antibodies are usually linked to an enzyme or 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 help tell one type of cancer from another type.

Getting a second opinion

You may want to get a second opinion to confirm your child’s 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. This doctor may agree with the first doctor, suggest changes to the treatment plan, or provide more information about your child’s cancer.

To learn more about choosing a doctor and getting a second opinion, visit 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, visit Questions to Ask Your Doctor About Cancer.

Treatment of childhood laryngeal tumors

Who treats children with laryngeal tumors?

A pediatric oncologist, a doctor who specializes in treating children with cancer, oversees treatment of laryngeal tumors. The pediatric oncologist works with other health care providers who are experts in treating children with cancer and who specialize in certain areas of medicine. Other specialists may include:

There are different types of treatment for children with laryngeal tumors. You and your child’s care team will work together to decide on treatment. Many factors will be considered, such as your child’s overall health and whether the tumor is newly diagnosed or has come back.

Your child’s treatment plan will include information about the tumor, the goals of treatment, treatment options, and the possible side effects. It will be helpful to talk with your child’s care team before treatment begins about what to expect. For help every step of the way, see our booklet, Children with Cancer: A Guide for Parents.

Treatment of childhood laryngeal cancer

Laser surgery uses a laser beam (a narrow beam of intense light) to destroy the cancer cells.

For laryngeal cancer that is likely to spread, external radiation therapy is also used. This treatment uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. Learn more about External Beam Radiation Therapy for Cancer and Radiation Therapy Side Effects.

If the cancer comes back after treatment, your child’s doctor will talk with you about what to expect and possible next steps. There might be treatment options that may shrink the cancer or control its growth. If there are no treatments, your child can receive care to control symptoms from cancer so they can be as comfortable as possible.

Treatment of childhood laryngeal papillomatosis

Laser surgery, which uses a laser beam (a narrow beam of intense light) to destroy cancer cells, may be used to treat newly diagnosed papillomatosis or other benign tumors.

For laryngeal papillomatosis that comes back after being removed by laser surgery four times in one year, treatment may include:

  • Immunotherapy. Immunotherapy uses the patient’s immune system to fight disease.
  • Laser surgery combined with 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.

Clinical trials

For some children, joining a clinical trial may be an option. There are different types of clinical trials for childhood 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 child’s age, and where the trials are being done. Clinical trials supported by other organizations can be found on the ClinicalTrials.gov website.

Learn more about clinical trials, including how to find and join one, at Clinical Trials Information for Patients and Caregivers.

Follow-up care

As your child goes through treatment, they will have follow-up tests or check-ups. Some of the tests that were done to diagnose 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 child’s condition has changed or if the cancer has recurred (come back).

Coping with your child's cancer

When your child has cancer, every member of the family needs support. Taking care of yourself during this difficult time is important. Reach out to your child’s treatment team and to people in your family and community for support. To learn more, visit Support for Families: Childhood Cancer and the booklet Children with Cancer: A Guide for Parents.

Related Resources

About This PDQ Summary

About PDQ

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 laryngeal cancer and papillomatosis. 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 Laryngeal Tumors. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/patient/child/laryngeal-treatment-pdq. Accessed <MM/DD/YYYY>.

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.

Paranasal Sinus and Nasal Cavity Cancer Treatment (PDQ®)–Patient Version

Paranasal Sinus and Nasal Cavity Cancer Treatment (PDQ®)–Patient Version

General Information About Paranasal Sinus and Nasal Cavity
Cancer

Key Points

  • Paranasal sinus and nasal cavity cancer is a disease in which malignant (cancer) cells form in the tissues of the paranasal sinuses and nasal cavity.
  • Different types of cells in the paranasal sinus and nasal cavity may become malignant.
  • Being exposed to certain chemicals or dust in the workplace can increase the risk of paranasal sinus and nasal cavity cancer.
  • Signs and symptoms of paranasal sinus and nasal cavity cancer include sinus problems and nosebleeds.
  • Tests that examine the sinuses and nasal cavity are used to diagnose paranasal sinus and nasal cavity cancer.
  • Certain factors affect prognosis (chance of recovery) and treatment options.

Paranasal sinus and nasal cavity cancer is a disease in which malignant (cancer) cells form in the tissues of the paranasal sinuses and nasal cavity.

Paranasal sinuses

“Paranasal” means near the nose. The para sinuses are hollow, air-filled spaces in the bones around the nose. The sinuses are lined with cells that make mucus, which keeps the inside of the nose from drying out during breathing.

EnlargeAnatomy of the paranasal sinuses; drawing shows front and side views of the frontal sinus, ethmoid sinus, maxillary sinus, and sphenoid sinus. The nasal cavity and pharynx (throat) are also shown.
Anatomy of the paranasal sinuses (spaces between the bones around the nose).

There are several para sinuses named after the bones that surround them:

Nasal cavity

The nose opens into the nasal cavity, which is divided into two nasal passages. Air moves through these passages during breathing. The nasal cavity lies above the bone that forms the roof of the mouth and curves down at the back to join the throat. The area just inside the nostrils is called the nasal vestibule. A small area of special cells in the roof of each nasal passage sends signals to the brain to give the sense of smell.

Together the paranasal sinuses and the nasal cavity filter and warm the air, and make it moist before it goes into the lungs. The movement of air through the sinuses and other parts of the respiratory system help make sounds for talking.

Paranasal sinus and nasal cavity cancer is a type of head and neck cancer.

Different types of cells in the paranasal sinus and nasal cavity may become malignant.

The most common type of paranasal sinus and nasal cavity cancer is squamous cell carcinoma. This type of cancer forms in the thin, flat cells lining the inside of the paranasal sinuses and the nasal cavity.

Other types of paranasal sinus and nasal cavity cancer include:

Being exposed to certain chemicals or dust in the workplace can increase the risk of paranasal sinus and nasal cavity cancer.

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 paranasal sinus and nasal cavity cancer, and it will develop in people who don’t have any known risk factors. Talk with your doctor if you think you may be at risk. Risk factors for paranasal sinus and nasal cavity cancer include:

  • Being exposed to certain workplace chemicals or dust, such as those found in the following jobs:
    • Furniture-making.
    • Sawmill work.
    • Woodworking (carpentry).
    • Shoemaking.
    • Metal-plating.
    • Flour mill or bakery work.
  • Being infected with human papillomavirus (HPV).
  • Being male and older than 40 years.
  • Smoking.

Signs and symptoms of paranasal sinus and nasal cavity cancer include sinus problems and nosebleeds.

These and other signs and symptoms may be caused by paranasal sinus and nasal cavity cancer or by other conditions. There may be no signs or symptoms in the early stages. Signs and symptoms may appear as the tumor grows. Check with your doctor if you have:

  • Blocked sinuses that do not clear, or sinus pressure.
  • Headaches or pain in the sinus areas.
  • A runny nose.
  • Nosebleeds.
  • A lump or sore inside the nose that does not heal.
  • A lump on the face or roof of the mouth.
  • Numbness or tingling in the face.
  • Swelling or other trouble with the eyes, such as double vision or the eyes pointing in different directions.
  • Pain in the upper teeth, loose teeth, or dentures that no longer fit well.
  • Pain or pressure in the ear.

Tests that examine the sinuses and nasal cavity are used to diagnose paranasal sinus and nasal cavity cancer.

In addition to asking about your personal and family health history and doing a physical exam, your doctor may perform the following tests and procedures:

  • Physical exam of the nose, face, and neck: An exam in which the doctor looks into the nose with a small, long-handled mirror to check for abnormal areas and checks the face and neck for lumps or swollen lymph nodes.
  • X-rays of the head and neck: An x-ray is a type of energy beam that can go through the body and onto film, making a picture of areas inside the body.
  • MRI (magnetic resonance imaging): A procedure that 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).
  • 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.
  • Biopsy: The removal of cells or tissues so they can be viewed under a microscope by a pathologist to check for signs of cancer. There are three types of biopsy:
  • Nasoscopy: A procedure to look inside the nose for abnormal areas. A nasoscope is inserted into the nose. A nasoscope is a thin, tube-like instrument with a light and a lens for viewing. A special tool on the nasoscope may be used to remove samples of tissue. The tissues samples are viewed under a microscope by a pathologist to check for signs of cancer.
  • Laryngoscopy: A procedure in which the doctor checks the larynx (voice box) with a mirror or a laryngoscope to check for abnormal areas. A laryngoscope is a thin, tube-like instrument with a light and a lens for viewing the inside of the throat and voice box. It may also have a tool to remove tissue samples, which are checked under a microscope for signs of cancer.

Certain factors affect prognosis (chance of recovery) and treatment options.

The prognosis and treatment options depend on:

  • Where the tumor is in the paranasal sinus or nasal cavity and whether it has spread.
  • The size of the tumor.
  • The type of cancer.
  • The patient’s age and general health.
  • Whether the cancer has just been diagnosed or has recurred (come back).

Paranasal sinus and nasal cavity cancers often have spread by the time they are diagnosed and are hard to cure. After treatment, a lifetime of frequent and careful follow-up is important because there is an increased risk of developing a second kind of cancer in the head or neck.

Stages of Paranasal Sinus and Nasal Cavity Cancer

Key Points

  • After paranasal sinus and nasal cavity cancer has been diagnosed, tests are done to find out if cancer cells have spread within the paranasal sinuses and nasal cavity or to other parts of the body.
  • There are three ways that cancer spreads in the body.
  • Cancer may spread from where it began to other parts of the body.
  • There is no standard staging system for cancer of the sphenoid and frontal sinuses.
  • The following stages are used for maxillary sinus cancer:
    • Stage 0 (carcinoma in situ)
    • Stage I
    • Stage II
    • Stage III
    • Stage IV
  • The following stages are used for nasal cavity and ethmoid sinus cancer:
    • Stage 0 (carcinoma in situ)
    • Stage I
    • Stage II
    • Stage III
    • Stage IV
  • After surgery, the stage of the cancer may change and more treatment may be needed.
  • Paranasal sinus and nasal cavity cancer can recur (come back) after it has been treated.

After paranasal sinus and nasal cavity cancer has been diagnosed, tests are done to find out if cancer cells have spread within the paranasal sinuses and nasal cavity or to other parts of the body.

The process used to find out if cancer has spread within the paranasal sinuses and nasal cavity or to other parts of the body is called staging. The information gathered from the staging process determines the stage of the disease. It is important to know the stage in order to plan treatment. The following tests and procedures may be used in the staging process:

  • Endoscopy: A procedure to look at organs and tissues inside the body to check for abnormal areas. An endoscope is inserted through an opening in the body, such as the nose or mouth. An endoscope is a thin, tube-like instrument with a light and a lens for viewing. It may also have a tool to remove tissue or lymph node samples, which are checked under a microscope for signs of disease.
  • 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.
  • Chest x-ray: An x-ray of the organs and bones inside the chest. An x-ray is a type of energy beam that can go through the body and onto film, making a picture of areas inside the body.
  • 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 areas inside the body. Sometimes 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).
  • 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 body. Malignant tumor cells show up brighter in the picture because they are more active and take up more glucose than normal cells do.
  • Bone scan: 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.

There are three ways that cancer spreads in the body.

Cancer can spread through tissue, the lymph system, and the blood:

  • Tissue. The cancer spreads from where it began by growing into nearby areas.
  • Lymph system. The cancer spreads from where it began by getting into the lymph system. The cancer travels through the lymph vessels to other parts of the body.
  • Blood. The cancer spreads from where it began by getting into the blood. The cancer travels through the blood vessels to other parts of the body.

Cancer may spread from where it began to other parts of the body.

When cancer spreads to another part of the body, it is called metastasis. Cancer cells break away from where they began (the primary tumor) and travel through the lymph system or blood.

  • Lymph system. The cancer gets into the lymph system, travels through the lymph vessels, and forms a tumor (metastatic tumor) in another part of the body.
  • Blood. The cancer gets into the blood, travels through the blood vessels, and forms a tumor (metastatic tumor) in another part of the body.

The metastatic tumor is the same type of cancer as the primary tumor. For example, if nasal cavity cancer spreads to the lung, the cancer cells in the lung are actually nasal cavity cancer cells. The disease is metastatic nasal cavity cancer, not lung cancer.

Many cancer deaths are caused when cancer moves from the original tumor and spreads to other tissues and organs. This is called metastatic cancer. This animation shows how cancer cells travel from the place in the body where they first formed to other parts of the body.

There is no standard staging system for cancer of the sphenoid and frontal sinuses.

The staging described below for the maxillary and ethmoid sinuses and the nasal cavity is only used for patients who have not had lymph nodes in the neck removed and checked for signs of cancer.

EnlargeDrawing shows different sizes of a tumor in centimeters (cm) compared to the size of a pea (1 cm), a peanut (2 cm), a grape (3 cm), a walnut (4 cm), a lime (5 cm), an egg (6 cm), a peach (7 cm), and a grapefruit (10 cm). Also shown is a 10-cm ruler and a 4-inch ruler.
Tumor 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).

The following stages are used for maxillary sinus cancer:

Stage 0 (carcinoma in situ)

In stage 0, abnormal cells are found in the mucous membranes lining the maxillary sinus. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called carcinoma in situ.

Stage I

In stage I, cancer has formed in the mucous membranes of the maxillary sinus.

Stage II

In stage II, cancer has spread to bone around the maxillary sinus, including the roof of the mouth and the nose, but not to bone at the back of the maxillary sinus or the part of the sphenoid bone behind the upper jaw.

Stage III

In stage III, cancer has spread to any of the following:

or

Cancer is found in the maxillary sinus and may have spread to any of the following:

  • The bones around the maxillary sinus, including the roof of the mouth and the nose.
  • The tissues under the skin.
  • The part of the eye socket near the nose or the bottom of the eye socket.
  • The area behind the cheek bone.
  • The ethmoid sinus.

Cancer has also spread to one lymph node on the same side of the neck as the cancer, and the lymph node is 3 centimeters or smaller.

Stage IV

Stage IV is divided into stages IVA, IVB, and IVC.

Stage IVA

In stage IVA, cancer has spread to any of the following:

Cancer may have also spread to one lymph node on the same side of the neck as the cancer, and the lymph node is 3 centimeters or smaller.

or

Cancer is found in the maxillary sinus and may have spread to any of the following:

  • The bones around the maxillary sinus, including the roof of the mouth and the nose.
  • The bone between the eyes.
  • The tissues under the skin.
  • The skin of the cheek.
  • The eye, the part of the eye socket near the nose, or the bottom of the eye socket.
  • The area behind the cheek bone.
  • The part of the sphenoid bone behind the upper jaw.
  • The area behind the upper jaw.
  • The ethmoid, sphenoid, or frontal sinuses.

Cancer has also spread to one of the following:

  • one lymph node on the same side of the neck as the cancer and the lymph node is larger than 3 centimeters but not larger than 6 centimeters; or
  • more than one lymph node on the same side of the neck as the cancer and the lymph nodes are not larger than 6 centimeters; or
  • lymph nodes on the opposite side of the neck as the cancer or on both sides of the neck, and the lymph nodes are not larger than 6 centimeters.

Stage IVB

In stage IVB, cancer has spread to any of the following:

  • The area behind the eye.
  • The brain.
  • The middle parts of the skull.
  • The nerves that begin in the brain and go to the face, neck, and other parts of the brain (cranial nerves).
  • The upper part of the throat behind the nose.
  • The base of the skull near the spinal cord.

Cancer may have also spread to one or more lymph nodes of any size, anywhere in the neck.

or

Cancer may be found anywhere in or near the maxillary sinus. Cancer has spread to a lymph node that is larger than 6 centimeters or has spread through the outside covering of a lymph node into nearby connective tissue.

Stage IVC

In stage IVC, cancer may be found anywhere in or near the maxillary sinus, may have spread to lymph nodes, and has spread to organs far away from the maxillary sinus, such as the lungs.

The following stages are used for nasal cavity and ethmoid sinus cancer:

Stage 0 (carcinoma in situ)

In stage 0, abnormal cells are found in the mucous membranes lining the nasal cavity or ethmoid sinus. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called carcinoma in situ.

Stage I

In stage I, cancer has formed and is found in only one area of either the nasal cavity or the ethmoid sinus and may have spread into bone.

Stage II

In stage II, cancer is found in two areas of either the nasal cavity or the ethmoid sinus that are near each other, or cancer has spread to an area next to the sinuses. Cancer may also have spread into bone.

Stage III

In stage III, cancer has spread to any of the following:

  • The part of the eye socket near the nose or the bottom of the eye socket.
  • The maxillary sinus.
  • The roof of the mouth.
  • The bone between the eyes.

or

Cancer is found in the nasal cavity or ethmoid sinus and may have spread to any of the following:

  • The part of the eye socket near the nose or the bottom of the eye socket.
  • The maxillary sinus.
  • The roof of the mouth.
  • The bone between the eyes.

Cancer has also spread to one lymph node on the same side of the neck as the cancer, and the lymph node is 3 centimeters or smaller.

Stage IV

Stage IV is divided into stages IVA, IVB, and IVC.

Stage IVA

In stage IVA, cancer has spread to any of the following:

Cancer may have also spread to one lymph node on the same side of the neck as the cancer, and the lymph node is 3 centimeters or smaller.

or

Cancer is found in the nasal cavity or ethmoid sinus and may have spread to any of the following:

  • The eye, the part of the eye socket near the nose, or the bottom of the eye socket.
  • The skin of the nose or cheek.
  • The front parts of the skull.
  • The part of the sphenoid bone behind the upper jaw.
  • The sphenoid or frontal sinuses.

Cancer has also spread to one of the following:

  • one lymph node on the same side of the neck as the cancer and the lymph node is larger than 3 centimeters but not larger than 6 centimeters; or
  • more than one lymph node on the same side of the neck as the cancer and the lymph nodes are not larger than 6 centimeters; or
  • lymph nodes on the opposite side of the neck as the cancer or on both sides of the neck, and the lymph nodes are not larger than 6 centimeters.

Stage IVB

In stage IVB, cancer has spread to any of the following:

  • The area behind the eye.
  • The brain.
  • The middle parts of the skull.
  • The nerves that begin in the brain and go to the face, neck, and other parts of the brain (cranial nerves).
  • The upper part of the throat behind the nose.
  • The base of the skull near the spinal cord.

Cancer may have also spread to one or more lymph nodes of any size, anywhere in the neck.

or

Cancer may be found anywhere in or near the nasal cavity and ethmoid sinus. Cancer has spread to a lymph node that is larger than 6 centimeters or has spread through the outside covering of a lymph node into nearby connective tissue.

Stage IVC

In stage IVC, cancer may be found anywhere in or near the nasal cavity and ethmoid sinus, may have spread to lymph nodes, and has spread to organs far away from the nasal cavity and ethmoid sinus, such as the lungs.

After surgery, the stage of the cancer may change and more treatment may be needed.

If the cancer is removed by surgery, a pathologist will examine a sample of the cancer tissue under a microscope. Sometimes, the pathologist’s review results in a change to the stage of the cancer and more treatment is needed after surgery.

Paranasal sinus and nasal cavity cancer can recur (come back) after it has been treated.

The cancer may come back in the paranasal sinuses and nasal cavity or in other parts of the body.

Treatment Option Overview

Key Points

  • There are different types of treatment for patients with paranasal sinus and nasal cavity cancer.
  • Patients with paranasal sinus and nasal cavity cancer should have their treatment planned by a team of doctors with expertise in treating head and neck cancer.
  • The following types of treatment are used:
    • Surgery
    • Radiation therapy
    • Chemotherapy
  • New types of treatment are being tested in clinical trials.
  • Treatment for paranasal sinus and nasal cavity cancer 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 paranasal sinus and nasal cavity cancer.

Different types of treatment are available for patients with paranasal sinus and nasal cavity cancer. 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.

Patients with paranasal sinus and nasal cavity cancer should have their treatment planned by a team of doctors with expertise in treating head and neck cancer.

Treatment will be overseen by a medical oncologist, a doctor who specializes in treating people with cancer. The medical oncologist works with other doctors who are experts in treating patients with head and neck cancer and who specialize in certain areas of medicine and rehabilitation. Patients who have paranasal sinus and nasal cavity cancer may need special help adjusting to breathing problems or other side effects of the cancer and its treatment. If a large amount of tissue or bone around the paranasal sinuses or nasal cavity is taken out, plastic surgery may be done to repair or rebuild the area. The treatment team may include the following specialists:

The following types of treatment are used:

Surgery

Surgery (removing the cancer in an operation) is a common treatment for all stages of paranasal sinus and nasal cavity cancer. A doctor may remove the cancer and some of the healthy tissue and bone around the cancer. If the cancer has spread, the doctor may remove lymph nodes and other tissues in the neck.

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. There are two types of radiation therapy:

  • External radiation therapy uses a machine outside the body to send radiation toward the area of the body with cancer. The total dose of radiation therapy is sometimes divided into several smaller, equal doses delivered over a period of several days. This is called fractionation.
    EnlargeExternal-beam radiation therapy of the head and neck; drawing shows a patient lying on a table under a machine that is used to aim high-energy radiation at the cancer. An inset shows a mesh mask that helps keep the patient's head and neck from moving during treatment. The mask has pieces of white tape with small ink marks on it. The ink marks are used to line up the radiation machine in the same position before each treatment.
    External-beam radiation therapy of the head and neck. A machine is used to aim high-energy radiation at the cancer. The machine can rotate around the patient, delivering radiation from many different angles to provide highly conformal treatment. A mesh mask helps keep the patient’s head and neck 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.
  • Internal radiation therapy uses a radioactive substance sealed in needles, seeds, wires, or catheters that are placed directly into or near the cancer.

The way the radiation therapy is given depends on the type and stage of the cancer being treated. External and internal radiation therapy are used to treat paranasal sinus and nasal cavity cancer.

External radiation therapy to the thyroid or the pituitary gland may change the way the thyroid gland works. The thyroid hormone levels in the blood may be tested before and after treatment.

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). When chemotherapy is placed directly into the cerebrospinal fluid, an organ, or a body cavity such as the abdomen, the drugs mainly affect cancer cells in those areas (regional chemotherapy). Combination chemotherapy is treatment using more than one anticancer drug.

The way the chemotherapy is given depends on the type and stage of the cancer being treated.

For more information, see Drugs Approved for Head and Neck Cancer. (Paranasal sinus and nasal cavity cancer is a type of head and neck cancer.)

New types of treatment are being tested in clinical trials.

Information about clinical trials is available from the NCI website.

Treatment for paranasal sinus and nasal cavity cancer may cause side effects.

For information about side effects caused by treatment for cancer, visit our Side Effects page.

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).

Treatment of Stage I Paranasal Sinus and Nasal Cavity Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of stage I paranasal sinus and nasal cavity cancer depends on where cancer is found in the paranasal sinuses and nasal cavity:

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 Stage II Paranasal Sinus and Nasal Cavity Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of stage II paranasal sinus and nasal cavity cancer depends on where cancer is found in the paranasal sinuses and nasal cavity:

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 Stage III Paranasal Sinus and Nasal Cavity Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of stage III paranasal sinus and nasal cavity cancer depends on where cancer is found in the paranasal sinuses and nasal cavity.

If cancer is in the maxillary sinus, treatment may include:

If cancer is in the ethmoid sinus, treatment may include:

  • Surgery followed by radiation therapy.
  • A clinical trial of combination chemotherapy before surgery or radiation therapy.
  • A clinical trial of combination chemotherapy after surgery or other cancer treatment.

If cancer is in the sphenoid sinus, treatment is the same as for nasopharyngeal cancer, usually radiation therapy with or without chemotherapy. For more information, see Nasopharyngeal Cancer Treatment.

If cancer is in the nasal cavity, treatment may include:

  • Surgery and/or radiation therapy.
  • Chemotherapy and radiation therapy.
  • A clinical trial of combination chemotherapy before surgery or radiation therapy.
  • A clinical trial of combination chemotherapy after surgery or other cancer treatment.

For inverting papillomas, treatment is usually surgery with or without radiation therapy.

For melanomas and sarcomas, treatment may include:

  • Surgery.
  • Radiation therapy.
  • Surgery, radiation therapy, and chemotherapy.

For midline granulomas, treatment is usually radiation therapy.

If cancer is in the nasal vestibule, treatment may include:

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 Stage IV Paranasal Sinus and Nasal Cavity Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of stage IV paranasal sinus and nasal cavity cancer depends on where cancer is found in the paranasal sinuses and nasal cavity.

If cancer is in the maxillary sinus, treatment may include:

If cancer is in the ethmoid sinus, treatment may include:

  • Radiation therapy before or after surgery.
  • Chemotherapy and radiation therapy.
  • A clinical trial of chemotherapy before surgery or radiation therapy.
  • A clinical trial of chemotherapy after surgery or other cancer treatment.
  • A clinical trial of chemotherapy and radiation therapy.

If cancer is in the sphenoid sinus, treatment is the same as for nasopharyngeal cancer, usually radiation therapy with or without chemotherapy. For more information, see Nasopharyngeal Cancer Treatment.

If cancer is in the nasal cavity, treatment may include:

  • Surgery and/or radiation therapy.
  • Chemotherapy and radiation therapy.
  • A clinical trial of combination chemotherapy before surgery or radiation therapy.
  • A clinical trial of combination chemotherapy after surgery or other cancer treatment.

For inverting papillomas, treatment is usually surgery with or without radiation therapy.

For melanomas and sarcomas, treatment may include:

  • Surgery.
  • Radiation therapy.
  • Chemotherapy.

For midline granulomas, treatment is usually radiation therapy.

If cancer is in the nasal vestibule, treatment may include:

  • External radiation therapy and/or internal radiation therapy with or without surgery.
  • A clinical trial of chemotherapy before surgery or radiation therapy.
  • A clinical trial of chemotherapy after surgery or other cancer treatment.
  • A clinical trial of chemotherapy 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.

Treatment of Recurrent Paranasal Sinus and Nasal Cavity
Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of recurrent paranasal sinus and nasal cavity cancer depends on where cancer is found in the paranasal sinuses and nasal cavity.

If cancer is in the maxillary sinus, treatment may include:

If cancer is in the ethmoid sinus, treatment may include:

  • Surgery and/or radiation therapy.
  • Chemotherapy as palliative therapy to relieve symptoms and improve the quality of life.
  • A clinical trial of chemotherapy.

If cancer is in the sphenoid sinus, treatment is the same as for nasopharyngeal cancer and may include radiation therapy with or without chemotherapy. For more information, see Nasopharyngeal Cancer Treatment.

If cancer is in the nasal cavity, treatment may include:

  • Surgery and/or radiation therapy.
  • Chemotherapy as palliative therapy to relieve symptoms and improve the quality of life.
  • A clinical trial of chemotherapy.

For inverting papillomas, treatment is usually surgery with or without radiation therapy.

For melanomas and sarcomas, treatment may include:

  • Surgery.
  • Chemotherapy as palliative therapy to relieve symptoms and improve the quality of life.

For midline granulomas, treatment is usually radiation therapy.

If cancer is in the nasal vestibule, treatment may include:

  • Surgery and/or radiation therapy.
  • Chemotherapy as palliative therapy to relieve symptoms and improve the quality of life.
  • A clinical trial of 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.

To Learn More About Paranasal Sinus and Nasal Cavity Cancer

About This PDQ Summary

About PDQ

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 paranasal sinus and nasal cavity 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 Paranasal Sinus and Nasal Cavity Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/patient/adult/paranasal-sinus-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389439]

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.

Salivary Gland Cancer Treatment (PDQ®)–Patient Version

Salivary Gland Cancer Treatment (PDQ®)–Patient Version

General Information About Salivary Gland Cancer

Key Points

  • Salivary gland cancer is a rare disease in which malignant (cancer) cells form in the tissues of the salivary glands.
  • Being exposed to certain types of radiation may increase the risk of salivary gland cancer.
  • Signs and symptoms of salivary gland cancer include a lump or trouble swallowing.
  • Tests that examine the head, neck, and the inside of the mouth are used to diagnose salivary gland cancer.
  • Certain factors affect prognosis (chance of recovery) and treatment options.

Salivary gland cancer is a rare disease in which malignant (cancer) cells form in the tissues of the salivary glands.

The salivary glands make saliva and release it into the mouth. Saliva has enzymes that help digest food and antibodies that help protect against infections of the mouth and throat. There are 3 pairs of major salivary glands:

  • Parotid glands: These are the largest salivary glands and are found in front of and just below each ear. Most major salivary gland tumors begin in this gland.
  • Sublingual glands: These glands are found under the tongue in the floor of the mouth.
  • Submandibular glands: These glands are found below the jawbone.
EnlargeAnatomy of the salivary glands; drawing shows a cross section of the head and the three main pairs of salivary glands. The parotid glands are in front of and just below each ear; the sublingual glands are under the tongue in the floor of the mouth; the submandibular glands are below each side of the jawbone. The tongue and lymph nodes are also shown.
Anatomy of the salivary glands. The three main pairs of salivary glands are the parotid glands, the sublingual glands, and the submandibular glands.

There are also hundreds of small (minor) salivary glands lining parts of the mouth, nose, and larynx that can be seen only with a microscope. Most small salivary gland tumors begin in the palate (roof of the mouth).

More than half of all salivary gland tumors are benign (not cancerous) and do not spread to other tissues.

Salivary gland cancer is a type of head and neck cancer.

Being exposed to certain types of radiation may increase the risk of salivary gland cancer.

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 salivary gland cancer, and it can develop in people who don’t have any known risk factors. Talk with your doctor if you think you may be at risk. Although the cause of most salivary gland cancers is not known, risk factors include the following:

  • Older age.
  • Treatment with radiation therapy to the head and neck.
  • Being exposed to certain substances at work.

Signs and symptoms of salivary gland cancer include a lump or trouble swallowing.

Salivary gland cancer may not cause any symptoms. It may be found during a regular dental check-up or physical exam. Signs and symptoms may be caused by salivary gland cancer or by other conditions. Check with your doctor if you have any of the following symptoms that do not go away:

  • A lump (usually painless) in the area of the ear, cheek, jaw, lip, or inside the mouth.
  • Fluid draining from the ear.
  • Trouble swallowing or opening the mouth widely.
  • Numbness or weakness in the face.
  • Pain in the face that does not go away.

Tests that examine the head, neck, and the inside of the mouth are used to diagnose salivary gland cancer.

In addition to asking about your personal and family health history and doing a physical exam, your doctor may perform the following tests and procedures:

  • MRI (magnetic resonance imaging): A procedure that 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).
  • 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 head and neck; drawing shows a patient lying on a table that slides through the CT scanner, which takes x-ray pictures of the inside of the head and neck.
    Computed tomography (CT) scan of the head and neck. The patient lies on a table that slides through the CT scanner, which takes x-ray pictures of the inside of the head and neck.
  • 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 body. Malignant tumor cells show up brighter in the picture because they are more active and take up more glucose than normal cells do.
  • Endoscopy: A procedure to look at organs and tissues inside the body to check for abnormal areas. For salivary gland cancer, an endoscope is inserted into the mouth to look at the mouth, throat, and larynx. An endoscope is a thin, tube-like instrument with a light and a lens for viewing.
  • Biopsy: The removal of cells or tissues so they can be viewed under a microscope by a pathologist to check for signs of cancer.
    • Fine needle aspiration (FNA) biopsy: The removal of tissue or fluid using a thin needle. An FNA is the most common type of biopsy used for salivary gland cancer.
    • Incisional biopsy: The removal of part of a lump or a sample of tissue that doesn’t look normal.
    • Surgery: If cancer cannot be diagnosed from the sample of tissue removed during an FNA biopsy or an incisional biopsy, the mass may be removed and checked for signs of cancer.

Because salivary gland cancer can be hard to diagnose, patients should ask to have the tissue samples checked by a pathologist who has experience in diagnosing salivary gland cancer.

Certain factors affect prognosis (chance of recovery) and treatment options.

The prognosis and treatment options depend on the following:

  • The stage of the cancer (especially the size of the tumor).
  • The type of salivary gland the cancer is in.
  • The type of cancer cells (how they look under a microscope).
  • The patient’s age and general health.

Stages of Salivary Gland Cancer

Key Points

  • After salivary gland cancer has been diagnosed, tests are done to find out if cancer cells have spread within the salivary gland or to other parts of the body.
  • There are three ways that cancer spreads in the body.
  • Cancer may spread from where it began to other parts of the body.
  • The following stages are used for salivary gland cancers that affect the parotid, submandibular, and sublingual glands:
    • Stage 0 (carcinoma in situ)
    • Stage I
    • Stage II
    • Stage III
    • Stage IV
  • Minor salivary glands are staged differently from the parotid, submandibular, and sublingual glands.
  • Salivary gland cancer can recur (come back) after it has been treated.

After salivary gland cancer has been diagnosed, tests are done to find out if cancer cells have spread within the salivary gland or to other parts of the body.

The process used to find out if cancer has spread within the salivary glands or to other parts of the body is called staging. The information gathered from the staging process determines the stage of the disease. It is important to know the stage in order to plan treatment. The following procedures may be used in the staging process:

  • MRI (magnetic resonance imaging): A procedure that 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).
  • 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 head and neck; drawing shows a patient lying on a table that slides through the CT scanner, which takes x-ray pictures of the inside of the head and neck.
    Computed tomography (CT) scan of the head and neck. The patient lies on a table that slides through the CT scanner, which takes x-ray pictures of the inside of the head and neck.

There are three ways that cancer spreads in the body.

Cancer can spread through tissue, the lymph system, and the blood:

  • Tissue. The cancer spreads from where it began by growing into nearby areas.
  • Lymph system. The cancer spreads from where it began by getting into the lymph system. The cancer travels through the lymph vessels to other parts of the body.
  • Blood. The cancer spreads from where it began by getting into the blood. The cancer travels through the blood vessels to other parts of the body.

Cancer may spread from where it began to other parts of the body.

When cancer spreads to another part of the body, it is called metastasis. Cancer cells break away from where they began (the primary tumor) and travel through the lymph system or blood.

  • Lymph system. The cancer gets into the lymph system, travels through the lymph vessels, and forms a tumor (metastatic tumor) in another part of the body.
  • Blood. The cancer gets into the blood, travels through the blood vessels, and forms a tumor (metastatic tumor) in another part of the body.

The metastatic tumor is the same type of cancer as the primary tumor. For example, if salivary gland cancer spreads to the lung, the cancer cells in the lung are actually salivary gland cancer cells. The disease is metastatic salivary gland cancer, not lung cancer.

Many cancer deaths are caused when cancer moves from the original tumor and spreads to other tissues and organs. This is called metastatic cancer. This animation shows how cancer cells travel from the place in the body where they first formed to other parts of the body.

The following stages are used for salivary gland cancers that affect the parotid, submandibular, and sublingual glands:

EnlargeDrawing shows different sizes of a tumor in centimeters (cm) compared to the size of a pea (1 cm), a peanut (2 cm), a grape (3 cm), a walnut (4 cm), a lime (5 cm), an egg (6 cm), a peach (7 cm), and a grapefruit (10 cm). Also shown is a 10-cm ruler and a 4-inch ruler.
Tumor 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 0 (carcinoma in situ)

In stage 0, abnormal cells are found in the lining of the salivary ducts or the small sacs that make up the salivary gland. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called carcinoma in situ.

Stage I

In stage I, cancer has formed. The tumor is in the salivary gland only and is 2 centimeters or smaller.

Stage II

In stage II, the tumor is in the salivary gland only and is larger than 2 centimeters but not larger than 4 centimeters.

Stage III

In stage III, one of the following is true:

  • The tumor is larger than 4 centimeters, and/or cancer has spread to soft tissue around the salivary gland; or
  • The tumor is any size, and cancer may have spread to soft tissue around the salivary gland. Cancer has spread to one lymph node on the same side of the head or neck as the tumor. The lymph node is 3 centimeters or smaller, and cancer has not grown outside the lymph node.

Stage IV

Stage IV is divided into stages IVA, IVB, and IVC as follows:

  • Stage IVA:
    • Cancer has spread to the skin, jawbone, ear canal, and/or facial nerve. Cancer may have spread to one lymph node on the same side of the head or neck as the tumor. The lymph node is 3 centimeters or smaller, and cancer has not grown outside the lymph node; or
    • The tumor is any size, and cancer may have spread to soft tissue around the salivary gland or to the skin, jawbone, ear canal, and/or facial nerve. Cancer has spread:
      • to one lymph node on the same side of the head or neck as the tumor or on the side opposite the primary tumor; the lymph node is 3 centimeters or smaller, and cancer has grown outside the lymph node; or
      • to one lymph node on the same side of the head or neck as the tumor; the lymph node is larger than 3 centimeters but not larger than 6 centimeters, and cancer has not grown outside the lymph node; or
      • to more than one lymph node on the same side of the head or neck as the tumor; the lymph nodes are 6 centimeters or smaller, and cancer has not grown outside the lymph nodes; or
      • to lymph nodes on both sides of the head or neck or on the side opposite the primary tumor; the lymph nodes are 6 centimeters or smaller, and cancer has not grown outside the lymph nodes.
  • Stage IVB:
    • The tumor is any size, and cancer may have spread to soft tissue around the salivary gland or to the skin, jawbone, ear canal, and/or facial nerve. Cancer has spread:
      • to one lymph node larger than 6 centimeters, and cancer has not grown outside the lymph node; or
      • to one lymph node on the same side of the head or neck as the tumor; the lymph node is larger than 3 centimeters, and cancer has grown outside the lymph node; or
      • to more than one lymph node on the same side of the head or neck as the tumor, on the side opposite the primary tumor, or on both sides of the head or neck; cancer has grown outside any of the lymph nodes; or
      • to one lymph node of any size on the side of the head or neck opposite the primary tumor; cancer has grown outside the lymph node;

        or

    • Cancer has spread to the bottom of the skull and/or surrounds the carotid artery. Cancer may have spread to one or more lymph nodes of any size on either or both sides of the head or neck and may have grown outside the lymph nodes.
  • Stage IVC:
    • Cancer has spread to other parts of the body, such as the lungs.

Minor salivary glands are staged differently from the parotid, submandibular, and sublingual glands.

Minor salivary gland (small salivary glands lining parts of the mouth, nose, and larynx) cancers are staged according to where they were first formed, such as the oral cavity or sinuses.

Salivary gland cancer can recur (come back) after it has been treated.

The cancer may come back in the salivary glands or in other parts of the body.

Treatment Option Overview

Key Points

  • There are different types of treatment for patients with salivary gland cancer.
  • Patients with salivary gland cancer should have their treatment planned by a team of health care providers who are experts in treating head and neck cancer.
  • The following types of treatment are used:
    • Surgery
    • Radiation therapy
    • Chemotherapy
  • New types of treatment are being tested in clinical trials.
    • Radiosensitizers
  • Treatment for salivary gland cancer 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 salivary gland cancer.

Different types of treatment are available for patients with salivary gland cancer. 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.

Patients with salivary gland cancer should have their treatment planned by a team of health care providers who are experts in treating head and neck cancer.

Treatment will be overseen by a medical oncologist, a doctor who specializes in treating cancer. Because the salivary glands help in eating and digesting food, patients may need special help adjusting to the side effects of the cancer and its treatment. The medical oncologist may refer you to other health care providers who have experience and expertise in treating patients with head and neck cancer and who specialize in certain areas of medicine. These may include the following specialists:

The following types of treatment are used:

Surgery

Surgery (removing the cancer in an operation) is a common treatment for salivary gland cancer. A doctor may remove the cancer and some of the healthy tissue around the cancer. In some cases, a lymphadenectomy (surgery in which lymph nodes are removed) will also be done.

After the doctor removes all the cancer that can be seen at the time of the surgery, some patients may be given radiation therapy after surgery to kill any cancer 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 head and neck; drawing shows a patient lying on a table under a machine that is used to aim high-energy radiation at the cancer. An inset shows a mesh mask that helps keep the patient's head and neck from moving during treatment. The mask has pieces of white tape with small ink marks on it. The ink marks are used to line up the radiation machine in the same position before each treatment.
    External-beam radiation therapy of the head and neck. A machine is used to aim high-energy radiation at the cancer. The machine can rotate around the patient, delivering radiation from many different angles to provide highly conformal treatment. A mesh mask helps keep the patient’s head and neck 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.

    Special types of external radiation may be used to treat some salivary gland tumors. These include:

    • Fast-neutron radiation therapy: Fast-neutron radiation therapy is a type of high-energy external radiation therapy. A radiation therapy machine aims neutrons (tiny, invisible particles) at the cancer cells to kill them. Fast-neutron radiation therapy uses a higher-energy radiation than the x-ray type of radiation therapy. This allows the radiation therapy to be given in fewer treatments.
    • Photon-beam radiation therapy: Photon-beam radiation therapy is a type of external radiation therapy that reaches deep tumors with high-energy x-rays made by a machine called a linear accelerator. This can be delivered as hyperfractionated radiation therapy, in which the total dose of radiation is divided into small doses, and the treatments are given more than once a day.

External radiation therapy is used to treat salivary gland cancer and may also be used as palliative therapy to relieve symptoms and improve quality of life.

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).

Learn more about Drugs Approved for Head and Neck Cancer. (Salivary gland cancer is a type of head and neck cancer.)

New types of treatment are being tested in clinical trials.

This summary section describes treatments that are being studied in clinical trials. It may not mention every new treatment being studied. Information about clinical trials is available from the NCI website.

Radiosensitizers

Radiosensitizers are drugs that make tumor cells more sensitive to radiation therapy. Combining radiation therapy with radiosensitizers may kill more tumor cells.

Treatment for salivary gland cancer may cause side effects.

For information about side effects caused by treatment for cancer, visit our Side Effects page.

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).

Treatment of Stage I Salivary Gland Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment for stage I salivary gland cancer depends on whether the cancer is low-grade (slow growing) or high-grade (fast growing).

If the cancer is low-grade, treatment may include the following:

If the cancer is high-grade, treatment 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.

Treatment of Stage II Salivary Gland Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment for stage II salivary gland cancer depends on whether the cancer is low-grade (slow growing) or high-grade (fast growing).

If the cancer is low-grade, treatment may include the following:

If the cancer is high-grade, treatment 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.

Treatment of Stage III Salivary Gland Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment for stage III salivary gland cancer depends on whether the cancer is low-grade (slow growing) or high-grade (fast growing).

If the cancer is low-grade, treatment may include the following:

If the cancer is high-grade, treatment may include the following:

  • Surgery with or without lymphadenectomy. Radiation therapy may also be given after surgery.
  • Fast-neutron radiation therapy.
  • Radiation therapy as palliative therapy to relieve symptoms and improve quality of life.
  • A clinical trial of radiation therapy and/or radiosensitizers.
  • A clinical trial of 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.

Treatment of Stages IVA, IVB, and IVC Salivary Gland Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of stage IVA, stage IVB, and stage IVC salivary gland cancer 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.

Treatment of Recurrent Salivary Gland Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of recurrent salivary gland cancer 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.

To Learn More About Salivary Gland Cancer

About This PDQ Summary

About PDQ

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 salivary gland 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 Salivary Gland Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/patient/adult/salivary-gland-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389192]

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.

Paranasal Sinus and Nasal Cavity Cancer Treatment (PDQ®)–Health Professional Version

Paranasal Sinus and Nasal Cavity Cancer Treatment (PDQ®)–Health Professional Version

General Information About Paranasal Sinus and Nasal Cavity Cancer

Incidence and Mortality

Most tumors of the paranasal sinuses present with advanced disease, and cure rates are generally poor (≤50%). Squamous cell carcinoma (SCC) is the most frequent type of malignant tumor in the nose and paranasal sinuses (70%–80%). Papillomas are distinct entities that may undergo malignant degeneration. The cancers grow within the bony confines of the sinuses and are often asymptomatic until they erode and invade adjacent structures.[13]

Nodal involvement is infrequent. Metastases from both the nasal cavity and paranasal sinuses may occur, and distant metastases are found in 20% to 40% of patients who do not respond to treatment. However, locoregional recurrence accounts for most cancer deaths because most patients die of direct extension into vital areas of the skull or of rapidly recurring local disease.

Cancers of the maxillary sinus are the most common of the paranasal sinus cancers. Tumors of the ethmoid sinuses, nasal vestibule, and nasal cavity are less common, and tumors of the sphenoid and frontal sinuses are rare.

Anatomy

The major lymphatic drainage route of the maxillary antrum is through the lateral and inferior collecting trunks to the first station submandibular, parotid, and jugulodigastric nodes and through the superoposterior trunk to retropharyngeal and jugular nodes.

Clinical Evaluation and Follow-Up

Pretreatment evaluation and staging, as well as the need for multidisciplinary planning of treatment, is very important. Generally, the first opportunity to treat patients with head and neck cancers is the most effective, although salvage surgery or salvage radiation therapy, as appropriate, may occasionally be successful.

Because most treatment failures occur within 2 years, patients must be monitored frequently and meticulously during this period. Lifetime follow-up is essential because nearly 33% of these patients develop second primary cancers in the aerodigestive tract.

Carcinogenesis and Risk Factors

Data indicate that various industrial exposures may be related to cancer of the paranasal sinus and nasal cavity. The risk of a second primary head and neck tumor is considerably increased.[4] A study has shown that a subgroup of paranasal sinus and nasal cavity SCCs are associated with human papilloma virus (HPV) infection and that HPV-positive patients may have a better prognosis than those who are HPV negative.[5]

References
  1. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  2. Laramore GE, ed.: Radiation Therapy of Head and Neck Cancer. Springer-Verlag, 1989.
  3. Thawley SE, Panje WR, Batsakis JG, et al., eds.: Comprehensive Management of Head and Neck Tumors. 2nd ed. WB Saunders, 1999.
  4. Johns ME, Kaplan MJ: Advances in the management of paranasal sinus tumors. In: Wolf GT, ed.: Head and Neck Oncology. Martinus Nijhoff Publishers, 1984, pp 27-52.
  5. Alos L, Moyano S, Nadal A, et al.: Human papillomaviruses are identified in a subgroup of sinonasal squamous cell carcinomas with favorable outcome. Cancer 115 (12): 2701-9, 2009. [PUBMED Abstract]

Cellular Classification of Paranasal Sinus and Nasal Cavity Cancer

The most common cell type for paranasal sinus and nasal cavity cancers is squamous cell carcinoma. Minor salivary gland tumors comprise 10% to 15% of these neoplasms. Malignant melanoma presents in less than 1% of neoplasms in this region. Some 5% of cases are malignant lymphomas.[1,2]

Esthesioneuroepithelioma, sometimes confused with undifferentiated carcinoma or undifferentiated lymphoma, arises from the olfactory nerves.[3]

Chondrosarcoma, osteosarcoma, Ewing sarcoma, and most soft tissue sarcomas have been reported for this region.

Inverting papilloma is considered a low-grade benign tumor with a tendency to recur and, in a small percentage of cases, to transform into a malignant tumor.

Midline granuloma, a progressively destructive condition, also involves this region.

References
  1. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  2. Goldenberg D, Golz A, Fradis M, et al.: Malignant tumors of the nose and paranasal sinuses: a retrospective review of 291 cases. Ear Nose Throat J 80 (4): 272-7, 2001. [PUBMED Abstract]
  3. Jethanamest D, Morris LG, Sikora AG, et al.: Esthesioneuroblastoma: a population-based analysis of survival and prognostic factors. Arch Otolaryngol Head Neck Surg 133 (3): 276-80, 2007. [PUBMED Abstract]

Stage Information for Paranasal Sinus and Nasal Cavity Cancer

The staging systems are clinical estimates of the extent of disease. The assessment of the tumor is based on inspection, palpation, and direct endoscopy when necessary. The tumor must be confirmed histologically, and any other pathological data obtained on biopsy may be included. The appropriate nodal drainage areas are examined by careful palpation. Computed tomographic and/or magnetic resonance imaging studies are generally required to adequately evaluate tumor extent before surgical resection or definitive radiation therapy. If a patient’s disease relapses, complete restaging must be done to select the appropriate additional therapy.[1,2]

American Joint Committee on Cancer (AJCC) Stage Groupings and TNM Definitions

Staging for nasal cavity and paranasal sinus carcinomas is not as well established as staging for other head and neck tumors. For cancer of the maxillary sinus, the nasal cavity, and the ethmoid sinus, the AJCC has designated staging by TNM (tumor, node, metastasis) classification. Lymphomas, sarcomas, and mucosal melanomas of the paranasal sinuses and nasal cavity are not staged using this system.[3] The staging described below is used only for patients who have not had a lymph node dissection of the neck.

Table 1. Definition of Primary Tumor (T)a
T Category Maxillary Sinus T Criteria Nasal Cavity and Ethmoid Sinus T Criteria
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
TX Primary tumor cannot be assessed. Primary tumor cannot be assessed.
Tis Carcinoma in situ. Carcinoma in situ.
T1 Tumor limited to maxillary sinus mucosa with no erosion or destruction of bone. Tumor restricted to any one subsite, with or without bony invasion.
T2 Tumor causing bone erosion or destruction including extension into the hard palate and/or middle nasal meatus, except extension to posterior wall of maxillary sinus and pterygoid plates. Tumor invading two subsites in a single region or extending to involve an adjacent region within the nasoethmoidal complex, with or without bony invasion.
T3 Tumor invades any of the following: bone of the posterior wall of maxillary sinus, subcutaneous tissues, floor or medial wall of orbit, pterygoid fossa, ethmoid sinuses. Tumor extends to invade the medial wall or floor of the orbit, maxillary sinus, palate, or cribriform plate.
T4 Moderately advanced or very advanced local disease. Moderately advanced or very advanced local disease.
–T4a Moderately advanced local disease. Tumor invades anterior orbital contents, skin of cheek, pterygoid plates, infratemporal fossa, cribriform plate, sphenoid or frontal sinuses. Moderately advanced local disease. Tumor invades any of the following: anterior orbital contents, skin of nose or cheek, minimal extension to anterior cranial fossa, pterygoid plates, sphenoid or frontal sinuses.
–T4b Very advanced local disease. Tumor invades any of the following: orbital apex, dura, brain, middle cranial fossa, cranial nerves other than maxillary division of trigeminal nerve (V2), nasopharynx, or clivus. Very advanced local disease. Tumor invades any of the following: orbital apex, dura, brain, middle cranial fossa, cranial nerves other than V2, nasopharynx, or clivus.
Table 2. Definition of Regional Lymph Node (N)a
N Category Clinical Node (cN) Criteria
ENE = extranodal extension.
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
Note: A designation of “U” or “L” may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L).
NX Regional lymph nodes cannot be assessed.
N0 No regional lymph node metastasis.
N1 Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(‒).
N2 Metastasis in a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(‒); or metastases in multiple ipsilateral lymph nodes, none >6 cm in greatest dimension and ENE(‒); or in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(‒).
–N2a Metastasis in a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(‒).
–N2b Metastases in multiple ipsilateral nodes, none >6 cm in greatest dimension and ENE(‒).
–N2c Metastases in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(‒).
N3 Metastasis in a lymph node >6 cm in greatest dimension and ENE(‒); or metastasis in any node(s) with clinically overt ENE(+).
–N3a Metastasis in a lymph node >6 cm in greatest dimension and ENE(‒).
–N3b Metastasis in any node(s) with clinically overt ENE (ENEc).
Table 3. Definition of Distant Metastasis (M)a
M Category M Criteria
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
M0 No distant metastasis (no pathologic M0; use clinical M to complete stage group).
M1 Distant metastasis.
Table 4. Definition of TNM Stage 0a
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
0 Tis, N0, M0 Tis = See Table 1.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
Table 5. Definition of TNM Stage Ia
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
I T1, N0, M0 T1 = See Table 1.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
Table 6. Definition of TNM Stage IIa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
II T2, N0, M0 T2 = See Table 1.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
Table 7. Definitions of TNM Stage IIIa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis; ENE = extranodal extension.
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
III T3, N0, M0 T3 = See Table 1.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
T1, T2, T3; N1, M0 T1, T2, T3 = See Table 1.
N1 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(‒).
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
Table 8. Definitions of TNM Stage IVA, IVB, and IVCa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 137–47.
IVA T4a; N0, N1; M0 T4a = See Table 1.
N0, N1 = See Table 2.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
T1, T2, T3, T4a; N2, M0 T1, T2, T3, T4a = See Table 1.
N2 = See Table 2.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
IVB Any T, N3, M0 Any T = See Table 1.
N3 = See Table 2.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
T4b, Any N, M0 T4b = See Table 1.
Any N = See Table 2.
M0 = No distant metastasis (no pathological M0; use clinical M to complete stage group).
IVC Any T, Any N, M1 Any T = See Table 1.
Any N = See Table 2.
M1 = Distant metastasis.
References
  1. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  2. Laramore GE, ed.: Radiation Therapy of Head and Neck Cancer. Springer-Verlag, 1989.
  3. Nasal cavity and paranasal sinuses. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp 137-47.

Treatment Option Overview for Paranasal Sinus and Nasal Cavity Cancer

Except for patients with T1 mucosal carcinomas, the accepted method of treatment is a combination of radiation therapy and surgery. The incidence of lymph node metastases is generally low (approximately 20% of cases). Thus, routine radical neck dissection or elective neck radiation therapy is recommended only for patients presenting with positive nodes.

For patients with operable tumors, radical surgery is generally performed first to remove the bulk of the tumor and to establish drainage of the affected sinus(es). This is followed by postoperative radiation therapy. Some institutions continue to give a full dose of radiation therapy preoperatively for all patients with stage II and stage III tumors and operate 4 to 6 weeks later.[13] A review of published clinical results of radical radiation therapy for head and neck cancer suggested a significant loss of local control with prolonged radiation therapy; therefore, lengthening of standard treatment schedules should be avoided whenever possible.[4]

Surgery

Surgical exploration may be required to determine operability.

Relative contraindications to surgery include destruction of the base of the skull (i.e., anterior cranial fossa), cavernous sinus, or the pterygoid process; infiltration of the mucous membranes of the nasopharynx; or nonresectable lymph node metastases. Surgical approaches include fenestration with removal of the bulk tumor, which is usually followed by radiation therapy or block resection of the upper jaw. A combined craniofacial approach, including resection of the floor of the anterior cranial fossa, has been used with success in selected patients.[5] Removal of the eye is performed if the orbit is extensively invaded by cancer. Clinically positive nodes, if resectable, may be treated with radical neck dissection.

Radiation Therapy

Radiation therapy must be carried to high doses for any significant probability of permanent control. The treatment volume must include all of the maxillary antrum and involved hemiparanasal sinus and contiguous areas. The orbit and its contents are excluded except under unusual circumstances. Lymph nodes of the neck, when palpable, should be treated in conjunction with treatment of advanced carcinomas of the antrum. This may be unnecessary for early tumors.

Accumulating evidence has demonstrated a high incidence (>30%–40%) of hypothyroidism in patients who have received external-beam radiation therapy to the entire thyroid gland or to the pituitary gland. Thyroid function testing of patients should be considered prior to therapy and as part of posttreatment follow-up.[6,7]

Recurrent Disease

Patients with recurrent disease should consider chemotherapy clinical trials. Chemotherapy for recurrent squamous cell cancer of the head and neck has been shown to be efficacious as palliation and may improve a patient’s quality of life and length of survival. Various drug combinations, including cisplatin, fluorouracil, and methotrexate, are effective.[8,9]

Treatment of tumors of the paranasal sinuses and of the nasal cavity should be planned on an individual basis because of the complexity involved.

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.[10,11] 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.[1012] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[1315] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[16] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[17]

References
  1. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  2. Laramore GE, ed.: Radiation Therapy of Head and Neck Cancer. Springer-Verlag, 1989.
  3. Thawley SE, Panje WR, Batsakis JG, et al., eds.: Comprehensive Management of Head and Neck Tumors. 2nd ed. WB Saunders, 1999.
  4. Fowler JF, Lindstrom MJ: Loss of local control with prolongation in radiotherapy. Int J Radiat Oncol Biol Phys 23 (2): 457-67, 1992. [PUBMED Abstract]
  5. Ganly I, Patel SG, Singh B, et al.: Craniofacial resection for malignant paranasal sinus tumors: Report of an International Collaborative Study. Head Neck 27 (7): 575-84, 2005. [PUBMED Abstract]
  6. Turner SL, Tiver KW, Boyages SC: Thyroid dysfunction following radiotherapy for head and neck cancer. Int J Radiat Oncol Biol Phys 31 (2): 279-83, 1995. [PUBMED Abstract]
  7. Constine LS: What else don’t we know about the late effects of radiation in patients treated for head and neck cancer? Int J Radiat Oncol Biol Phys 31 (2): 427-9, 1995. [PUBMED Abstract]
  8. Jacobs C, Lyman G, Velez-García E, et al.: A phase III randomized study comparing cisplatin and fluorouracil as single agents and in combination for advanced squamous cell carcinoma of the head and neck. J Clin Oncol 10 (2): 257-63, 1992. [PUBMED Abstract]
  9. Schornagel JH, Verweij J, de Mulder PH, et al.: Randomized phase III trial of edatrexate versus methotrexate in patients with metastatic and/or recurrent squamous cell carcinoma of the head and neck: a European Organization for Research and Treatment of Cancer Head and Neck Cancer Cooperative Group study. J Clin Oncol 13 (7): 1649-55, 1995. [PUBMED Abstract]
  10. 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]
  11. Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
  12. 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]
  13. 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]
  14. 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]
  15. 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]
  16. 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]
  17. 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 I Paranasal Sinus and Nasal Cavity Cancer

Stage I disease includes small lesions.

Maxillary Sinus Tumors

Maxillary sinus tumors are small lesions of the infrastructure.

Treatment options for stage I maxillary sinus tumors include the following:

  1. Surgical resection.
  2. Postoperative radiation therapy should be considered for close margins (particularly in tumors of the suprastructure).

Ethmoid Sinus Tumors

Ethmoid sinus tumors are usually extensive when diagnosed.[13]

Treatment options for stage I ethmoid sinus tumors include the following:

  1. External-beam radiation therapy alone is generally used for unresectable lesions.
  2. Well-localized lesions can be resected, but resection of the ethmoids, maxilla, and orbit, with consideration for a craniofacial approach, is generally required.
  3. If surgery can be done with good functional and cosmetic results, postoperative radiation therapy should be given even with clear surgical margins.

Sphenoid Sinus Tumors

Treatment options for stage I sphenoid sinus tumors include the following:

  1. Treatment is the same as for nasopharyngeal cancers, primarily radiation therapy. For more information, see the Treatment of Stage I Nasopharyngeal Carcinoma section in Nasopharyngeal Carcinoma Treatment.

Nasal Cavity Tumors

For nasal cavity tumors (squamous cell carcinomas), treatment preferences are either surgery or radiation therapy, which have equal cure rates.

Treatment options for stage I nasal cavity tumors include the following:

  1. Surgery for tumors of the septum.
  2. Radiation therapy for tumors of the lateral and superior walls.[4]
  3. Surgery plus radiation therapy for tumors of the septal and lateral walls.[5]

Inverting Papillomas

Treatment options for stage I inverting papillomas include the following:

  1. Surgical excision.
  2. Re-excision for surgery failures.
  3. Radical surgery may eventually be necessary.
  4. Radiation therapy has been used successfully for surgical failures.

Melanomas and Sarcomas

Treatment options for stage I melanomas and sarcomas include the following:

  1. Surgical excision if possible.
  2. Combined surgery, radiation therapy, and chemotherapy are recommended for rhabdomyosarcoma.

Midline Granulomas

Treatment options for stage I midline granulomas include the following:

  1. Radiation therapy to nasal cavity and paranasal sinuses.

Nasal Vestibule Tumors

Treatment options for stage I nasal vestibule tumors include the following:

  1. Surgery or radiation therapy may be performed. If lesions are extremely small, surgery is preferred, provided that no deformity is expected and a need for reconstruction is not anticipated. Radiation therapy is preferred for other small lesions.[6,7] Treatment of the ipsilateral neck should be considered.

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
  1. Kraus DH, Sterman BM, Levine HL, et al.: Factors influencing survival in ethmoid sinus cancer. Arch Otolaryngol Head Neck Surg 118 (4): 367-72, 1992. [PUBMED Abstract]
  2. Shah JP: Surgery of the anterior skull base for malignant tumors. Acta Otorhinolaryngol Belg 53 (3): 191-4, 1999. [PUBMED Abstract]
  3. Cantù G, Solero CL, Mariani L, et al.: Anterior craniofacial resection for malignant ethmoid tumors–a series of 91 patients. Head Neck 21 (3): 185-91, 1999. [PUBMED Abstract]
  4. Hawkins RB, Wynstra JH, Pilepich MV, et al.: Carcinoma of the nasal cavity–results of primary and adjuvant radiotherapy. Int J Radiat Oncol Biol Phys 15 (5): 1129-33, 1988. [PUBMED Abstract]
  5. Ang KK, Jiang GL, Frankenthaler RA, et al.: Carcinomas of the nasal cavity. Radiother Oncol 24 (3): 163-8, 1992. [PUBMED Abstract]
  6. Levendag PC, Pomp J: Radiation therapy of squamous cell carcinoma of the nasal vestibule. Int J Radiat Oncol Biol Phys 19 (6): 1363-7, 1990. [PUBMED Abstract]
  7. Wong CS, Cummings BJ: The place of radiation therapy in the treatment of squamous cell carcinoma of the nasal vestibule. A review. Acta Oncol 27 (3): 203-8, 1988. [PUBMED Abstract]

Treatment of Stage II Paranasal Sinus and Nasal Cavity Cancer

Stage II disease includes small and moderately advanced lesions.

Maxillary Sinus Tumors

Treatment options for stage II maxillary sinus tumors include the following:

  1. Surgical resection with high-dose preoperative or postoperative radiation therapy.

Ethmoid Sinus Tumors

Ethmoid sinus tumors are usually extensive when diagnosed.[13]

Treatment options for stage II ethmoid sinus tumors include the following:

  1. External-beam radiation therapy alone is generally used and produces better overall results than surgery.
  2. Well-localized lesions can be resected, but resection of the ethmoids, maxilla, and orbit, often with a combined neurosurgical sinus craniofacial approach, is generally required.
  3. If surgery can be done with good functional and cosmetic results, postoperative radiation therapy should be given, even when surgical margins are clear.

Sphenoid Sinus Tumors

Treatment options for stage II sphenoid sinus tumors include the following:

  1. Treatment is the same as for nasopharyngeal cancers, primarily radiation therapy. Concurrent chemotherapy and radiation therapy may be considered. For more information, see the Treatment of Stages II, III, and IV Nonmetastatic Nasopharyngeal Carcinoma section in Nasopharyngeal Carcinoma Treatment.

Nasal Cavity Tumors

For nasal cavity tumors (squamous cell carcinomas), treatment preferences are either surgery or radiation therapy, which have equal cure rates.[4]

Treatment options for stage II nasal cavity tumors include the following:

  1. Surgery or radiation therapy for tumors of the septum.
  2. Radiation therapy for tumors of the lateral and superior walls. Concurrent chemotherapy and radiation therapy may be considered.
  3. Surgery plus radiation therapy for tumors of the septal and lateral walls.[5]

Inverting Papillomas

Treatment options for stage II inverting papillomas include the following:

  1. Surgical excision.
  2. Re-excision for surgery failures.
  3. Radiation therapy for radical surgery failures may eventually be necessary.

Melanomas and Sarcomas

Treatment options for stage II melanomas and sarcomas include the following:

  1. Surgical excision if possible.
  2. Combined surgery, radiation therapy, and chemotherapy are recommended for rhabdomyosarcoma.

Midline Granulomas

Treatment options for stage II midline granulomas include the following:

  1. Radiation therapy to nasal cavity and paranasal sinuses.

Nasal Vestibule Tumors

Treatment options for stage II nasal vestibule tumors include the following:

  1. Surgery or radiation therapy may be performed. If tumors are extremely small, surgery is preferred, provided that no deformity is expected and a need for reconstruction is not anticipated. Radiation therapy is preferred for other small lesions.[6,7] Treatment of the neck should be considered.

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
  1. Kraus DH, Sterman BM, Levine HL, et al.: Factors influencing survival in ethmoid sinus cancer. Arch Otolaryngol Head Neck Surg 118 (4): 367-72, 1992. [PUBMED Abstract]
  2. Cantù G, Solero CL, Mariani L, et al.: Anterior craniofacial resection for malignant ethmoid tumors–a series of 91 patients. Head Neck 21 (3): 185-91, 1999. [PUBMED Abstract]
  3. Shah JP: Surgery of the anterior skull base for malignant tumors. Acta Otorhinolaryngol Belg 53 (3): 191-4, 1999. [PUBMED Abstract]
  4. Hawkins RB, Wynstra JH, Pilepich MV, et al.: Carcinoma of the nasal cavity–results of primary and adjuvant radiotherapy. Int J Radiat Oncol Biol Phys 15 (5): 1129-33, 1988. [PUBMED Abstract]
  5. Ang KK, Jiang GL, Frankenthaler RA, et al.: Carcinomas of the nasal cavity. Radiother Oncol 24 (3): 163-8, 1992. [PUBMED Abstract]
  6. Levendag PC, Pomp J: Radiation therapy of squamous cell carcinoma of the nasal vestibule. Int J Radiat Oncol Biol Phys 19 (6): 1363-7, 1990. [PUBMED Abstract]
  7. Wong CS, Cummings BJ: The place of radiation therapy in the treatment of squamous cell carcinoma of the nasal vestibule. A review. Acta Oncol 27 (3): 203-8, 1988. [PUBMED Abstract]

Treatment of Stage III Paranasal Sinus and Nasal Cavity Cancer

Stage III disease includes small and moderately advanced lesions.

Maxillary Sinus Tumors

Treatment options for stage III maxillary sinus tumors include the following:

  1. Surgical resection with high-dose preoperative or postoperative radiation therapy.
  2. Superfractionated preoperative or postoperative radiation therapy (under clinical evaluation).[1]

Ethmoid Sinus Tumors

Treatment options for stage III ethmoid sinus tumors include the following:

  1. Generally, a craniofacial resection in combination with postoperative radiation therapy.[24]
  2. Clinical trials using new drug combinations for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.

Sphenoid Sinus Tumors

Treatment options for stage III sphenoid sinus tumors include the following:

  1. Treatment is the same as for nasopharyngeal cancers, primarily radiation therapy. For more information, see the Treatment of Stages II, III, and IV Nonmetastatic Nasopharyngeal Carcinoma section in Nasopharyngeal Carcinoma Treatment.
  2. Concurrent chemotherapy and radiation therapy may be considered.

Nasal Cavity Tumors

Nasal cavity tumors are squamous cell carcinomas.

Treatment options for stage III nasal cavity tumors include the following:

  1. Surgery alone.
  2. Radiation therapy alone.[5] Concurrent chemotherapy and radiation therapy may be considered.
  3. Combined surgery and radiation therapy (postoperative radiation therapy is preferred).[5,6]
  4. Clinical trials using new drug combinations for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.

Inverting Papillomas

Treatment options for stage III inverting papillomas include the following:

  1. Surgical excision.
  2. Re-excision for surgery failures.
  3. Radiation therapy or radical surgery may eventually be necessary.

Melanomas and Sarcomas

Treatment options for stage III melanomas and sarcomas include the following:

  1. Surgical excision if possible; otherwise, consider radiation therapy.
  2. Combined surgery, radiation therapy, and chemotherapy are recommended for rhabdomyosarcoma.

Midline Granulomas

Treatment options for stage III midline granulomas include the following:

  1. Radiation therapy to nasal cavity and paranasal sinuses.

Nasal Vestibule Tumors

Treatment options for stage III nasal vestibule tumors include the following:

  1. Generally, radiation is preferred to minimize deformity.[7] External-beam (photons or electrons) and/or interstitial implantation can be used. Surgery is reserved for salvage.
  2. Clinical trials using new drug combinations for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.

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
  1. Johnson CR, Schmidt-Ullrich RK, Wazer DE: Concomitant boost technique using accelerated superfractionated radiation therapy for advanced squamous cell carcinoma of the head and neck. Cancer 69 (11): 2749-54, 1992. [PUBMED Abstract]
  2. Kraus DH, Sterman BM, Levine HL, et al.: Factors influencing survival in ethmoid sinus cancer. Arch Otolaryngol Head Neck Surg 118 (4): 367-72, 1992. [PUBMED Abstract]
  3. Cantù G, Solero CL, Mariani L, et al.: Anterior craniofacial resection for malignant ethmoid tumors–a series of 91 patients. Head Neck 21 (3): 185-91, 1999. [PUBMED Abstract]
  4. Shah JP: Surgery of the anterior skull base for malignant tumors. Acta Otorhinolaryngol Belg 53 (3): 191-4, 1999. [PUBMED Abstract]
  5. Hawkins RB, Wynstra JH, Pilepich MV, et al.: Carcinoma of the nasal cavity–results of primary and adjuvant radiotherapy. Int J Radiat Oncol Biol Phys 15 (5): 1129-33, 1988. [PUBMED Abstract]
  6. Ang KK, Jiang GL, Frankenthaler RA, et al.: Carcinomas of the nasal cavity. Radiother Oncol 24 (3): 163-8, 1992. [PUBMED Abstract]
  7. Wong CS, Cummings BJ: The place of radiation therapy in the treatment of squamous cell carcinoma of the nasal vestibule. A review. Acta Oncol 27 (3): 203-8, 1988. [PUBMED Abstract]

Treatment of Stage IV Paranasal Sinus and Nasal Cavity Cancer

Stage IV disease includes advanced lesions.

Maxillary Sinus Tumors

Treatment options for stage IV maxillary sinus tumors include the following:

  1. High-dose radiation therapy is used because extension to the base of the skull and nasopharynx is a potential, but not absolute, contraindication to surgery. If radiation therapy is to be used alone, localized drainage of the sinus(es) must be established before radiation therapy treatments are initiated.
  2. Superfractionated radiation therapy (under clinical evaluation).[1]
  3. Clinical trials for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.
  4. Concurrent chemotherapy and radiation therapy may be considered.

Ethmoid Sinus Tumors

Treatment options for stage IV ethmoid sinus tumors include the following:

  1. Generally, a craniofacial resection in combination with preoperative or postoperative radiation therapy is performed.[24]
  2. Concurrent chemotherapy and radiation therapy may be considered for patients with inoperable tumors.
  3. Clinical trials for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.

Sphenoid Sinus Tumors

Treatment options for stage IV sphenoid sinus tumors include the following:

  1. Treatment is the same as for nasopharyngeal cancers, primarily radiation therapy. For more information, see the Treatment of Stages II, III, and IV Nonmetastatic Nasopharyngeal Carcinoma and Treatment of Metastatic and Recurrent Nasopharyngeal Carcinoma sections in Nasopharyngeal Carcinoma Treatment.
  2. Concurrent chemotherapy and radiation therapy may be considered.

Nasal Cavity Tumors

Nasal cavity tumors are squamous cell carcinomas.

Treatment options for stage IV nasal cavity tumors include the following:

  1. Surgery alone.
  2. Radiation therapy alone.[5] Concurrent chemotherapy and radiation therapy may be considered.
  3. Combined surgery and radiation therapy (postoperative radiation therapy is preferred).[5]
  4. Clinical trials for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.

Inverting Papillomas

Treatment options for stage IV inverting papillomas include the following:

  1. Surgical excision.
  2. Re-excision for surgery failures.
  3. Radiation therapy or radical surgery may eventually be necessary.

Melanomas and Sarcomas

Treatment options for stage IV melanomas and sarcomas include the following:

  1. Surgical excision if possible.
  2. Appropriate radiation therapy and various chemotherapy agents should be considered.

Midline Granulomas

Treatment options for stage IV midline granulomas include the following:

  1. Radiation therapy to nasal cavity and paranasal sinuses.

Nasal Vestibule Tumors

Treatment options for stage IV nasal vestibule tumors include the following:

  1. Generally, radiation therapy is preferred to minimize deformity. External-beam (i.e., photons or electrons) and/or interstitial implantation can be used. Surgery is reserved for salvage. Treatment of the neck should be considered.
  2. Clinical trials for advanced tumors should be considered to evaluate chemotherapy preoperatively or before radiation therapy. Adjuvant therapy after surgery or after combined-modality therapy should also be considered.
  3. Concurrent chemotherapy and radiation therapy may be considered.

Neoadjuvant chemotherapy as used in clinical trials has been used to shrink tumors and to render them more definitively treatable with either surgery or radiation therapy. This chemotherapy is given before the other modalities; therefore, the designation of neoadjuvant is used to distinguish it from standard adjuvant therapy, which is given after or during definitive therapy with radiation or after surgery. Many drug combinations have been used in neoadjuvant chemotherapy.[68]

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
  1. Johnson CR, Schmidt-Ullrich RK, Wazer DE: Concomitant boost technique using accelerated superfractionated radiation therapy for advanced squamous cell carcinoma of the head and neck. Cancer 69 (11): 2749-54, 1992. [PUBMED Abstract]
  2. Kraus DH, Sterman BM, Levine HL, et al.: Factors influencing survival in ethmoid sinus cancer. Arch Otolaryngol Head Neck Surg 118 (4): 367-72, 1992. [PUBMED Abstract]
  3. Cantù G, Solero CL, Mariani L, et al.: Anterior craniofacial resection for malignant ethmoid tumors–a series of 91 patients. Head Neck 21 (3): 185-91, 1999. [PUBMED Abstract]
  4. Shah JP: Surgery of the anterior skull base for malignant tumors. Acta Otorhinolaryngol Belg 53 (3): 191-4, 1999. [PUBMED Abstract]
  5. Hawkins RB, Wynstra JH, Pilepich MV, et al.: Carcinoma of the nasal cavity–results of primary and adjuvant radiotherapy. Int J Radiat Oncol Biol Phys 15 (5): 1129-33, 1988. [PUBMED Abstract]
  6. Stupp R, Weichselbaum RR, Vokes EE: Combined modality therapy of head and neck cancer. Semin Oncol 21 (3): 349-58, 1994. [PUBMED Abstract]
  7. Al-Sarraf M: Head and neck cancer: chemotherapy concepts. Semin Oncol 15 (1): 70-85, 1988. [PUBMED Abstract]
  8. Dimery IW, Hong WK: Overview of combined modality therapies for head and neck cancer. J Natl Cancer Inst 85 (2): 95-111, 1993. [PUBMED Abstract]

Treatment of Recurrent Paranasal Sinus and Nasal Cavity Cancer

Chemotherapy for recurrent head and neck squamous cell cancer has shown promise. Chemotherapy may be indicated when there is recurrence in either distant or local disease after primary surgery or radiation therapy, and when there is residual disease after primary treatment.[1,2] Survival may be improved in those achieving a complete response to chemotherapy.[3] Combined-modality therapy with platinum and radiation therapy has been used in clinical trials such as UMCC-8810.[4]

Maxillary Sinus Tumors

Treatment options for recurrent maxillary sinus tumors include the following:

  1. After surgery, radiation therapy or craniofacial resection with postoperative radiation therapy.
  2. After radiation therapy, craniofacial resection if indicated.
  3. Chemotherapy should be considered for patients with disease that does not respond to other treatments.
  4. Clinical trials using chemotherapy should be considered.[5,6]

Ethmoid Sinus Tumors

Treatment options for recurrent ethmoid sinus tumors include the following:

  1. After limited surgery, craniofacial resection, radiation therapy, or both.[79]
  2. After radiation therapy, craniofacial resection.
  3. Chemotherapy should be considered for patients with disease that does not respond to other treatments.
  4. Clinical trials using chemotherapy should be considered.[5,6]

Sphenoid Sinus Tumors

Treatment options for recurrent sphenoid sinus tumors include the following:

  1. Treatment is the same as for nasopharyngeal cancers, primarily radiation therapy. For more information, see the Treatment of Metastatic and Recurrent Nasopharyngeal Carcinoma section in Nasopharyngeal Carcinoma Treatment.
  2. Chemotherapy should be considered for patients with disease that does not respond to other treatments.

Nasal Cavity Tumors

For nasal cavity tumors (squamous cell carcinomas), salvage is possible in approximately 25% of patients.

Treatment options for recurrent nasal cavity tumors include the following:

  1. For disease that does not respond to radiation therapy, craniofacial resection.
  2. For disease that does not respond to surgery, radiation therapy.
  3. Chemotherapy should be considered for patients with disease that does not respond to radiation therapy or surgery.
  4. Clinical trials using chemotherapy should be considered.[5,6]

Inverting Papillomas

Treatment options for recurrent inverting papillomas include the following:

  1. Surgical excision.
  2. Re-excision for surgery failures.
  3. Radical surgery or radiation therapy may eventually be necessary.

Melanomas and Sarcomas

Treatment options for recurrent melanomas and sarcomas include the following:

  1. Surgical excision if possible.
  2. Appropriate chemotherapy geared specifically to cell type. For more information, see the Treatment of Metastatic and Recurrent Nasopharyngeal Carcinoma section in Nasopharyngeal Carcinoma Treatment and the Treatment of Recurrent Major Salivary Gland Cancer section in Salivary Gland Cancer Treatment.

Midline Granulomas

Treatment options for recurrent midline granulomas include the following:

  1. Radiation therapy to nasal cavity and paranasal sinuses.

Nasal Vestibule Tumors

Treatment options for recurrent nasal vestibule tumors include the following:

  1. For disease that does not respond to radiation therapy, surgery.
  2. For disease that does not respond to surgery, radiation therapy or a combination of surgery and radiation therapy.
  3. Chemotherapy should be considered for patients with disease that does not respond to radiation therapy or surgery.
  4. Clinical trials using chemotherapy should be considered.[5,6]

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
  1. Kies MS, Levitan N, Hong WK: Chemotherapy of head and neck cancer. Otolaryngol Clin North Am 18 (3): 533-41, 1985. [PUBMED Abstract]
  2. LoRusso P, Tapazoglou E, Kish JA, et al.: Chemotherapy for paranasal sinus carcinoma. A 10-year experience at Wayne State University. Cancer 62 (1): 1-5, 1988. [PUBMED Abstract]
  3. Al-Kourainy K, Kish J, Ensley J, et al.: Achievement of superior survival for histologically negative versus histologically positive clinically complete responders to cisplatin combination in patients with locally advanced head and neck cancer. Cancer 59 (2): 233-8, 1987. [PUBMED Abstract]
  4. Al-Sarraf M, Pajak TF, Marcial VA, et al.: Concurrent radiotherapy and chemotherapy with cisplatin in inoperable squamous cell carcinoma of the head and neck. An RTOG Study. Cancer 59 (2): 259-65, 1987. [PUBMED Abstract]
  5. Brasnu D, Laccourreye O, Bassot V, et al.: Cisplatin-based neoadjuvant chemotherapy and combined resection for ethmoid sinus adenocarcinoma reaching and/or invading the skull base. Arch Otolaryngol Head Neck Surg 122 (7): 765-8, 1996. [PUBMED Abstract]
  6. Licitra L, Locati LD, Cavina R, et al.: Primary chemotherapy followed by anterior craniofacial resection and radiotherapy for paranasal cancer. Ann Oncol 14 (3): 367-72, 2003. [PUBMED Abstract]
  7. Kraus DH, Sterman BM, Levine HL, et al.: Factors influencing survival in ethmoid sinus cancer. Arch Otolaryngol Head Neck Surg 118 (4): 367-72, 1992. [PUBMED Abstract]
  8. Cantù G, Solero CL, Mariani L, et al.: Anterior craniofacial resection for malignant ethmoid tumors–a series of 91 patients. Head Neck 21 (3): 185-91, 1999. [PUBMED Abstract]
  9. Shah JP: Surgery of the anterior skull base for malignant tumors. Acta Otorhinolaryngol Belg 53 (3): 191-4, 1999. [PUBMED Abstract]

Latest Updates to This Summary (07/05/2024)

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.

Treatment Option Overview for Paranasal Sinus and Nasal Cavity Cancer

Added Fluorouracil dosing as a new subsection.

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 paranasal sinus and nasal cavity 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 Paranasal Sinus and Nasal Cavity Cancer Treatment are:

  • Andrea Bonetti, MD (Pederzoli Hospital)
  • 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 Paranasal Sinus and Nasal Cavity Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/hp/adult/paranasal-sinus-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389272]

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.

Salivary Gland Cancer Treatment (PDQ®)–Health Professional Version

Salivary Gland Cancer Treatment (PDQ®)–Health Professional Version

General Information About Salivary Gland Cancer

Incidence and Mortality

Salivary gland tumors are a morphologically and clinically diverse group of neoplasms, which may present significant diagnostic and management challenges. These tumors are rare. From 2018 to 2022, the age-adjusted incidence of salivary gland cancer in the United States was 1.3 cases per 100,000 people per year. The age-adjusted mortality rate from 2019 to 2023 was 0.3 deaths per 100,000 people per year.[1] Malignant salivary gland neoplasms account for more than 0.5% of all malignancies and approximately 3% to 5% of all head and neck cancers.[2,3] Most patients with malignant salivary gland tumors are in their sixth or seventh decade of life.[4,5]

Carcinogenesis and Risk Factors

Although exposure to ionizing radiation has been implicated as a cause of salivary gland cancer, the etiology of most salivary gland cancers cannot be determined.[3,4,6,7] Occupations associated with an increased risk for salivary gland cancers include rubber products manufacturing, asbestos mining, plumbing, and some types of woodworking.[4]

Anatomy

Tumors of the salivary glands comprise those in the major glands (e.g., parotid, submandibular, and sublingual) and the minor glands (e.g., oral mucosa, palate, uvula, floor of mouth, posterior tongue, retromolar area and peritonsillar area, pharynx, larynx, and paranasal sinuses).[3,8] Minor salivary gland lesions are most frequently seen in the oral cavity.[3]

Of salivary gland neoplasms, more than 50% are benign, and approximately 70% to 80% originate in the parotid gland.[2,3,9] The palate is the most common site of minor salivary gland tumors. The frequency of malignant lesions varies by site. Approximately 20% to 25% of parotid tumors, 35% to 40% of submandibular tumors, 50% of palate tumors, and more than 90% of sublingual gland tumors are malignant.[2,10]

Histopathology

Histologically, salivary gland tumors represent the most heterogenous group of tumors of any tissue in the body.[11] Although almost 40 histological types of epithelial tumors of the salivary glands exist, some are exceedingly rare and may be the subject of only a few case reports.[2,12] The most common benign major and minor salivary gland tumor is pleomorphic adenoma, which makes up about 50% of all salivary gland tumors and 65% of parotid gland tumors.[2] The most common malignant major and minor salivary gland tumor is mucoepidermoid carcinoma, which represents about 10% of all salivary gland neoplasms and approximately 35% of malignant salivary gland neoplasms.[2,13] This neoplasm occurs most often in the parotid gland.[3,13,14] For more information about this type and other histological types of salivary gland neoplasms, see the Cellular Classification of Salivary Gland Cancer section.

Clinical Presentation

Most patients with benign tumors of the major or minor salivary glands present with painless swelling of the parotid, submandibular, or sublingual glands. Neurological signs, such as numbness or weakness caused by nerve involvement, typically indicate a malignancy.[3] Facial nerve weakness associated with a parotid or submandibular tumor is an ominous sign. Persistent facial pain is highly suggestive of malignancy. Approximately 10% to 15% of malignant parotid neoplasms present with pain.[9,15] However, most parotid tumors, both benign and malignant, present as an asymptomatic mass in the gland.[3,9] For more information, see Cancer Pain.

Prognostic Factors

Early-stage, low-grade, malignant salivary gland tumors are usually curable by adequate surgical resection alone. The prognosis is more favorable when the tumor is in a major salivary gland. The parotid gland is most favorable followed by the submandibular gland. The least favorable primary sites are the sublingual and minor salivary glands. Large bulky tumors or high-grade tumors carry a poorer prognosis and may best be treated by surgical resection combined with postoperative radiation therapy.[16] The prognosis also depends on the following factors:[17,18]

  • Gland in which the tumor arises.
  • Histology.
  • Grade (i.e., degree of malignancy).
  • Extent of primary tumor (i.e., stage).
  • Whether the tumor involves the facial nerve, has fixation to the skin or deep structures, or has spread to lymph nodes or distant sites.

Follow-Up and Survivorship

Overall, clinical stage, particularly tumor size, may be the crucial factor that determines the outcome of salivary gland cancer and may be more important than histological grade.[19]

Treatment management

Perineural invasion can occur, particularly in high-grade adenoid cystic carcinoma, and should be specifically identified and treated.[20] Radiation therapy may increase the chance of local control and increase the survival of patients when adequate margins cannot be achieved.[21][Level of evidence C2] Unresectable or recurrent tumors may respond to chemotherapy.[2224] Fast neutron-beam radiation therapy or accelerated hyperfractionated photon-beam schedules have been effective in the treatment of inoperable, unresectable, and recurrent tumors.[2527]

Follow-up after treatment

Complications of surgical treatment for parotid neoplasms include facial nerve dysfunction and Frey syndrome (also known as gustatory flushing and sweating and auriculotemporal syndrome).[9] Frey syndrome has been successfully treated with injections of botulinum toxin A.[2830]

References
  1. 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.
  2. Speight PM, Barrett AW: Salivary gland tumours. Oral Dis 8 (5): 229-40, 2002. [PUBMED Abstract]
  3. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  4. Ellis GL, Auclair PL: Tumors of the Salivary Glands. Armed Forces Institute of Pathology, 1996. Atlas of Tumor Pathology, 3.
  5. Wahlberg P, Anderson H, Biörklund A, et al.: Carcinoma of the parotid and submandibular glands–a study of survival in 2465 patients. Oral Oncol 38 (7): 706-13, 2002. [PUBMED Abstract]
  6. Scanlon EF, Sener SF: Head and neck neoplasia following irradiation for benign conditions. Head Neck Surg 4 (2): 139-45, 1981 Nov-Dec. [PUBMED Abstract]
  7. van der Laan BF, Baris G, Gregor RT, et al.: Radiation-induced tumours of the head and neck. J Laryngol Otol 109 (4): 346-9, 1995. [PUBMED Abstract]
  8. Spiro RH, Thaler HT, Hicks WF, et al.: The importance of clinical staging of minor salivary gland carcinoma. Am J Surg 162 (4): 330-6, 1991. [PUBMED Abstract]
  9. Gooden E, Witterick IJ, Hacker D, et al.: Parotid gland tumours in 255 consecutive patients: Mount Sinai Hospital’s quality assurance review. J Otolaryngol 31 (6): 351-4, 2002. [PUBMED Abstract]
  10. Theriault C, Fitzpatrick PJ: Malignant parotid tumors. Prognostic factors and optimum treatment. Am J Clin Oncol 9 (6): 510-6, 1986. [PUBMED Abstract]
  11. Brandwein MS, Ferlito A, Bradley PJ, et al.: Diagnosis and classification of salivary neoplasms: pathologic challenges and relevance to clinical outcomes. Acta Otolaryngol 122 (7): 758-64, 2002. [PUBMED Abstract]
  12. Seifert G, Sobin LH: Histological Typing of Salivary Gland Tumours. 2nd ed. Springer-Verlag, 1991.
  13. Guzzo M, Andreola S, Sirizzotti G, et al.: Mucoepidermoid carcinoma of the salivary glands: clinicopathologic review of 108 patients treated at the National Cancer Institute of Milan. Ann Surg Oncol 9 (7): 688-95, 2002. [PUBMED Abstract]
  14. Goode RK, Auclair PL, Ellis GL: Mucoepidermoid carcinoma of the major salivary glands: clinical and histopathologic analysis of 234 cases with evaluation of grading criteria. Cancer 82 (7): 1217-24, 1998. [PUBMED Abstract]
  15. Spiro RH, Huvos AG, Strong EW: Cancer of the parotid gland. A clinicopathologic study of 288 primary cases. Am J Surg 130 (4): 452-9, 1975. [PUBMED Abstract]
  16. Parsons JT, Mendenhall WM, Stringer SP, et al.: Management of minor salivary gland carcinomas. Int J Radiat Oncol Biol Phys 35 (3): 443-54, 1996. [PUBMED Abstract]
  17. Vander Poorten VL, Balm AJ, Hilgers FJ, et al.: The development of a prognostic score for patients with parotid carcinoma. Cancer 85 (9): 2057-67, 1999. [PUBMED Abstract]
  18. Terhaard CH, Lubsen H, Van der Tweel I, et al.: Salivary gland carcinoma: independent prognostic factors for locoregional control, distant metastases, and overall survival: results of the Dutch head and neck oncology cooperative group. Head Neck 26 (8): 681-92; discussion 692-3, 2004. [PUBMED Abstract]
  19. Spiro RH: Factors affecting survival in salivary gland cancers. In: McGurk M, Renehan AG, eds.: Controversies in the Management of Salivary Gland Disease. Oxford University Press, 2001, pp 143-50.
  20. Gormley WB, Sekhar LN, Wright DC, et al.: Management and long-term outcome of adenoid cystic carcinoma with intracranial extension: a neurosurgical perspective. Neurosurgery 38 (6): 1105-12; discussion 1112-3, 1996. [PUBMED Abstract]
  21. Hosokawa Y, Shirato H, Kagei K, et al.: Role of radiotherapy for mucoepidermoid carcinoma of salivary gland. Oral Oncol 35 (1): 105-11, 1999. [PUBMED Abstract]
  22. Borthne A, Kjellevold K, Kaalhus O, et al.: Salivary gland malignant neoplasms: treatment and prognosis. Int J Radiat Oncol Biol Phys 12 (5): 747-54, 1986. [PUBMED Abstract]
  23. Spiro RH: Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head Neck Surg 8 (3): 177-84, 1986 Jan-Feb. [PUBMED Abstract]
  24. Licitra L, Cavina R, Grandi C, et al.: Cisplatin, doxorubicin and cyclophosphamide in advanced salivary gland carcinoma. A phase II trial of 22 patients. Ann Oncol 7 (6): 640-2, 1996. [PUBMED Abstract]
  25. Wang CC, Goodman M: Photon irradiation of unresectable carcinomas of salivary glands. Int J Radiat Oncol Biol Phys 21 (3): 569-76, 1991. [PUBMED Abstract]
  26. Buchholz TA, Laramore GE, Griffin BR, et al.: The role of fast neutron radiation therapy in the management of advanced salivary gland malignant neoplasms. Cancer 69 (11): 2779-88, 1992. [PUBMED Abstract]
  27. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]
  28. Naumann M, Zellner M, Toyka KV, et al.: Treatment of gustatory sweating with botulinum toxin. Ann Neurol 42 (6): 973-5, 1997. [PUBMED Abstract]
  29. Arad-Cohen A, Blitzer A: Botulinum toxin treatment for symptomatic Frey’s syndrome. Otolaryngol Head Neck Surg 122 (2): 237-40, 2000. [PUBMED Abstract]
  30. von Lindern JJ, Niederhagen B, Bergé S, et al.: Frey syndrome: treatment with type A botulinum toxin. Cancer 89 (8): 1659-63, 2000. [PUBMED Abstract]

Cellular Classification of Salivary Gland Cancer

Salivary gland neoplasms are remarkable for their histological diversity. These neoplasms include benign and malignant tumors of epithelial, mesenchymal, and lymphoid origin. Salivary gland tumors pose a particular challenge to the surgical pathologist. Differentiating benign from malignant tumors may be difficult, primarily because of the complexity of the classification and the rarity of several entities, which may exhibit a broad spectrum of morphological diversity in individual lesions.[1] In some cases, hybrid lesions may be seen.[2] The key guiding principle to establish the malignant nature of a salivary gland tumor is the demonstration of an infiltrative margin.[1]

The following cellular classification scheme draws heavily from a scheme published by the Armed Forces Institute of Pathology (AFIP).[3] Malignant nonepithelial neoplasms are included in the scheme because these neoplasms comprise a significant proportion of salivary gland neoplasms seen in the clinical setting. For completeness, malignant secondary tumors are also included in the scheme.

Where AFIP statistics regarding the incidence, or relative frequency, of particular histopathologies are cited, some bias may exist because of the AFIP methods of case accrual as a pathology reference service. When possible, other sources are cited for incidence data. Notwithstanding the AFIP data, the incidence of a particular histopathology has been found to vary considerably depending upon the study cited. This variability in reporting may be partially caused by the rare incidence of many salivary gland neoplasms.

Epithelial Neoplasms

The clinician should be aware that several benign epithelial salivary gland neoplasms have malignant counterparts, which are discussed below:[3]

Histological grading of salivary gland carcinomas is important to determine the proper treatment approach, although it is not an independent indicator of the clinical course and must be considered in the context of the clinical stage. Clinical stage, particularly tumor size, may be the critical factor to determine the outcome of salivary gland cancer and may be more important than histological grade.[1] For example, stage I intermediate-grade or high-grade mucoepidermoid carcinomas can be successfully treated, whereas low-grade mucoepidermoid carcinomas that present as stage III disease may have a very aggressive clinical course.[4]

Grading is used primarily for mucoepidermoid carcinomas, adenocarcinomas, not otherwise specified (NOS), adenoid cystic carcinomas, and squamous cell carcinomas.[1,3] Various other salivary gland carcinomas can also be categorized according to histological grade as follows:[3,58]

Low grade

Low grade, intermediate grade, and high grade

Intermediate grade and high grade

High grade

*[Note: Some investigators consider mucoepidermoid carcinoma to be of only two grades: low grade and high grade.5]

Mucoepidermoid carcinoma

Mucoepidermoid carcinoma is a malignant epithelial tumor that is composed of various proportions of mucous, epidermoid (e.g., squamous), intermediate, columnar, and clear cells and often demonstrates prominent cystic growth. It is the most common malignant neoplasm observed in the major and minor salivary glands.[1,9] Mucoepidermoid carcinoma represents 29% to 34% of malignant tumors originating in both major and minor salivary glands.[3,5,10,11] In two large retrospective series, 84% to 93% of cases originated in the parotid gland.[12,13] With regard to malignant tumors of the minor salivary glands, mucoepidermoid carcinoma shows a strong predilection for the lower lip.[3,14] In an AFIP review of civilian cases, the mean age of patients was 47 years (range, 8–92 years).[3] Prior exposure to ionizing radiation appears to substantially increase the risk of developing malignant neoplasms of the major salivary glands, particularly mucoepidermoid carcinoma.[3,13]

Most patients are asymptomatic and present with solitary, painless masses. Symptoms include pain, drainage from the ipsilateral ear, dysphagia, trismus, and facial paralysis.[3] For more information, see Cancer Pain.

Microscopic grading of mucoepidermoid carcinoma is important to determine the prognosis.[1,12,15] Mucoepidermoid carcinomas are graded as low grade, intermediate grade, and high grade. Grading parameters with point values include the following:

  • Intracystic component (+2).
  • Neural invasion present (+2).
  • Necrosis present (+3).
  • Mitosis (≥4 per 10 high-power field [+3]).
  • Anaplasia present (+4).

Total point scores are 0 to 4 for low grade, 5 to 6 for intermediate grade, and 7 to 14 for high grade.

In a retrospective review of 243 cases of mucoepidermoid carcinoma of the major salivary glands, a statistically significant correlation was shown between this point-based grading system and outcome for parotid tumors but not for submandibular tumors.[12] Another retrospective study that used this histological grading system indicated that tumor grade correlated well with prognosis for mucoepidermoid carcinoma of the major salivary glands, excluding submandibular tumors, and minor salivary glands.[13] A modification of this grading system placed more emphasis on features of tumor invasion.[16] Nonetheless, though tumor grade may be useful, stage appears to be a better indicator of prognosis.[3,16]

Cytogenetically, mucoepidermoid carcinoma is characterized by a t(11;19)(q14–21;p12–13) translocation, which is occasionally the sole cytogenetic alteration.[1719] This translocation creates a novel MECT1::MAML2 gene fusion that disrupts a Notch signaling pathway.[20] Notch signaling plays a key role in the normal development of many tissues and cell types, through diverse effects on cellular differentiation, survival, and/or proliferation, and may be involved in a wide variety of human neoplasms.[21]

Rarely, mucoepidermoid carcinoma may originate within the jaws. This tumor type is known as central mucoepidermoid carcinoma.[3] The mandibular to maxillary predilection is approximately 3:1.[22]

Adenoid cystic carcinoma

Adenoid cystic carcinoma, formerly known as cylindroma, is a slow growing but aggressive neoplasm with a remarkable capacity for recurrence.[23] Morphologically, three growth patterns have been described: cribriform, or classic pattern; tubular; and solid, or basaloid pattern. The tumors are categorized according to the predominant pattern.[3,2325] The cribriform pattern shows epithelial cell nests that form cylindrical patterns. The lumina of these spaces contain periodic acid-Schiff (PAS)-positive mucopolysaccharide secretions. The tubular pattern reveals tubular structures that are lined by stratified cuboidal epithelium. The solid pattern shows solid groups of cuboidal cells. The cribriform pattern is the most common, and the solid pattern is the least common.[26] Solid adenoid cystic carcinoma is a high-grade lesion with reported recurrence rates of as much as 100%, compared with 50% to 80% for the tubular and cribriform variants.[25]

In a review of its case files, the AFIP found adenoid cystic carcinoma to be the fifth most common malignant epithelial tumor of the salivary glands after mucoepidermoid carcinomas; adenocarcinomas, NOS; acinic cell carcinomas; and PLGA.[3] Other series, however, reported adenoid cystic carcinoma to be the second most common malignant tumor, with an incidence or relative frequency of approximately 20%.[1] In the AFIP data, this neoplasm constitutes approximately 7.5% of all epithelial malignancies and 4% of all benign and malignant epithelial salivary gland tumors. The peak incidence for this tumor is reported to be in the fourth through sixth decades of life.[3]

This neoplasm typically develops as a slow-growing swelling in the preauricular or submandibular region. Pain and facial paralysis develop frequently during the course of the disease and are likely related to the associated high incidence of nerve invasion.[3] For more information, see Cancer Pain. Regardless of histological grade, adenoid cystic carcinomas, with their unusually slow biological growth, tend to have a protracted course and ultimately a poor outcome, with 10-year survival rates reported to be less than 50% for all grades.[1,27] These carcinomas typically show frequent recurrences and late distant metastases.[1,28] Clinical stage may be a better prognostic indicator than histological grade.[28,29] In a retrospective review of 92 cases, tumors larger than 4 cm were associated with an unfavorable clinical course in all cases.[30]

Adenocarcinomas
Acinic cell carcinoma

Acinic cell carcinoma, also known as acinic cell adenocarcinoma, is a malignant epithelial neoplasm in which the neoplastic cells express acinar differentiation. By conventional use, the term acinic cell carcinoma is defined by cytological differentiation toward serous acinar cells, as opposed to mucous acinar cells, whose characteristic feature is cytoplasmic PAS-positive zymogen-type secretory granules.[3] In AFIP data of salivary gland neoplasms, acinic cell carcinoma is the third most common salivary gland epithelial neoplasm after mucoepidermoid carcinoma and adenocarcinoma, NOS.[3] Acinic cell carcinoma accounted for 17% of primary malignant salivary gland tumors or about 6% of all salivary gland neoplasms. More than 80% of these tumors occur in the parotid gland, women were affected more than men, and the mean age was 44 years. Other studies have reported a relative frequency of acinic cell carcinoma from 0% to 19% of malignant salivary gland neoplasms.[3]

Clinically, patients typically present with a slowly enlarging mass in the parotid region. Pain is a symptom in more than 33% of patients. For acinic cell carcinoma, staging is likely a better predictor of outcome than histological grading.[3] In a retrospective review of 90 cases, poor prognostic features included pain or fixation; gross invasion; and microscopic features of desmoplasia, atypia, or increased mitotic activity. Neither morphological pattern nor cell composition was a predictive feature.[31] For more information, see Cancer Pain.

PLGA

PLGA is a malignant epithelial tumor that is essentially limited to occurrence in minor salivary gland sites and is characterized by bland, uniform nuclear features; diverse but characteristic architecture; infiltrative growth; and perineural infiltration.[3] In a series of 426 patients with minor salivary gland tumors, PLGA represented 11% of all tumors and 26% of those that were malignant.[32] In minor gland sites, PLGA is twice as frequent as adenoid cystic carcinoma, and among all benign and malignant salivary gland neoplasms, only pleomorphic adenoma and mucoepidermoid carcinoma are more common.[3] In the AFIP case files, more than 60% of tumors occurred in the mucosa of either the soft or hard palates, approximately 16% occurred in the buccal mucosa, and 12% occurred in the upper lip. The average age of patients is 59 years, with 70% of patients between the ages of 50 and 79 years.[3] The female-to-male ratio is about 2:1, a proportion greater than for malignant salivary gland tumors in general.[3,33]

PLGA typically presents as a firm, nontender swelling involving the mucosa of the hard and soft palates (i.e., it is often found at their junction), the cheek, or the upper lip. Discomfort, bleeding, telangiectasia, or ulceration of the overlying mucosa may occasionally occur.[3] This salivary gland neoplasm typically runs a moderately indolent course. In a study of 40 cases with long-term follow-up, the overall survival rate was 80% at 25 years.[34] Because of the unpredictable behavior of the tumor, some investigators consider the qualifying term, low grade, to be misleading and instead prefer the term polymorphous adenocarcinoma.[1]

Adenocarcinoma, NOS

Adenocarcinoma, NOS, is a salivary gland carcinoma that shows glandular or ductal differentiation but lacks the prominence of any of the morphological features that characterize the other, more specific carcinoma types. The diagnosis of adenocarcinoma, NOS, is essentially one of exclusion. In an AFIP review of cases, adenocarcinoma, NOS, was second only to mucoepidermoid carcinoma in frequency among malignant salivary gland neoplasms.[3] Other series have reported an incidence of 4% to 10%.[1] In AFIP files, the mean patient age was 58 years.[3] Approximately 40% and 60% of tumors occurred in the major and minor salivary glands, respectively. Among the major salivary gland tumors, 90% occurred in the parotid gland. Adenocarcinoma, NOS is graded in a similar way to extrasalivary lesions according to the degree of differentiation.[1] Tumor grades include low-grade, intermediate-grade, and high-grade categories.[3]

Patients with tumors in the major salivary glands typically present with solitary, painless masses.[35] Two retrospective studies indicate that survival is better for patients with tumors of the oral cavity than for those with tumors of the parotid and submandibular glands.[35,36] These studies differ regarding the prognostic significance of tumor grade.

Rare adenocarcinomas
Basal cell adenocarcinoma

Basal cell adenocarcinoma, also known as basaloid salivary carcinoma, carcinoma ex monomorphic adenoma, malignant basal cell adenoma, malignant basal cell tumor, or basal cell carcinoma, is an epithelial neoplasm that is cytologically similar to basal cell adenoma but is infiltrative and has a small potential for metastasis.[3] In AFIP case files spanning almost 11 years, basal cell carcinoma made up 1.6% of all salivary gland neoplasms and 2.9% of salivary gland malignancies.[3] Nearly 90% of tumors occurred in the parotid gland.[3,37] The average age of patients was 60 years.[3]

Similar to most salivary gland neoplasms, swelling is typically the only sign or symptom.[37] A sudden increase in size may occur in a few patients.[38] Basal cell carcinomas are low-grade carcinomas that are infiltrative, locally destructive, and tend to recur. The carcinomas occasionally metastasize. In a retrospective series that included 29 patients, there were recurrences in 7 patients and metastases in 3 patients.[37] In another retrospective review that included 72 patients, 37% of the patients experienced local recurrences.[38] The overall prognosis for patients with this tumor is good.[37,38]

Clear cell carcinoma

Clear cell carcinoma, also known as clear cell adenocarcinoma, is a very rare malignant epithelial neoplasm composed of a monomorphous population of cells that have optically clear cytoplasm with standard hematoxylin and eosin stains and lack features of other specific neoplasms. Because of inconsistencies in the methods of reporting salivary gland neoplasms, meaningful incidence rates for this tumor are difficult to derive from the literature.[3] Most cases involve the minor salivary glands.[1,3,3941] In the AFIP case files, the mean age of patients was approximately 58 years.[3]

In most patients, swelling is the only symptom. Clear cell adenocarcinoma is a low-grade neoplasm. As of 1996, the AFIP reported that no patient had died of this tumor.[3]

Cystadenocarcinoma

Cystadenocarcinoma is a rare malignant epithelial tumor characterized histologically by prominent cystic and, frequently, papillary growth but lacking features that characterize cystic variants of several more common salivary gland neoplasms. It is also known as malignant papillary cystadenoma, mucus-producing adenopapillary, or nonepidermoid, carcinoma; low-grade papillary adenocarcinoma of the palate; or papillary adenocarcinoma. Cystadenocarcinoma is the malignant counterpart of cystadenoma.[3]

In a review that included 57 patients, the AFIP found that men and women are affected equally. The average patient age was approximately 59 years. Approximately 65% of the tumors occurred in the major salivary glands, primarily in the parotid.[3] Most patients present with a slow-growing asymptomatic mass. Clinically, this neoplasm is rarely associated with pain or facial paralysis. Cystadenocarcinoma is considered to be a low-grade neoplasm.[3]

Sebaceous adenocarcinoma

Sebaceous adenocarcinoma is a rare malignant epithelial tumor composed of islands and sheets of cells that have morphologically atypical nuclei, an infiltrative growth pattern, and focal sebaceous differentiation. This is a very rare tumor, as few cases have been reported in the literature.[3] Almost all cases occur in the parotid gland.[3] The average age of patients is reported to be 69 years.[42]

An equal number of patients present with a painless, slow-growing, asymptomatic swelling or pain. A few experience facial paralysis.[3] Most sebaceous adenocarcinomas are probably intermediate-grade malignancies. The tumor recurs in about 33% of cases.[43,44]

Sebaceous lymphadenocarcinoma

Sebaceous lymphadenocarcinoma is an extremely rare malignant tumor that represents carcinomatous transformation of sebaceous lymphadenoma. The carcinoma element may be sebaceous adenocarcinoma or some other specific or nonspecific form of salivary gland cancer.[3] Only three cases have been reported in the literature.[43,45] The three cases occurred in or around the parotid gland. All patients were in their seventh decade of life. Two of the three patients were asymptomatic. One had tenderness on palpation. Case reports suggest that this is a low-grade malignancy with a good prognosis.[44,45]

Oncocytic carcinoma

Oncocytic carcinoma, also known as oncocytic adenocarcinoma, is a rare, predominantly oncocytic neoplasm whose malignant nature is reflected both by its abnormal morphological features and infiltrative growth. Oncocytic carcinoma represented less than 1% of almost 3,100 salivary gland tumors accessioned to the AFIP files during a 10-year period.[3] Most cases occurred in the parotid gland. The average age of patients in the AFIP series was 63 years.[3]

Approximately 33% of the patients usually develop parotid masses that cause pain or paralysis.[46] Oncocytic carcinoma is a high-grade carcinoma. Patients with tumors smaller than 2 cm have a better prognosis than patients with larger tumors.[6]

Salivary duct carcinoma

Salivary duct carcinoma, also known as salivary duct adenocarcinoma, is a rare, typically high-grade malignant epithelial neoplasm composed of structures that resemble expanded salivary gland ducts. A low-grade variant exists.[47] Incidence rates vary depending on the study cited.[3] In the AFIP files, salivary duct carcinomas represented only 0.2% of all epithelial salivary gland neoplasms. More than 85% of cases involved the parotid gland, and approximately 75% of patients were men. The peak incidence was reported to be in the seventh and eighth decades of life.[3]

Clinically, parotid swelling is the most common sign. Facial nerve dysfunction or paralysis occur in more than 25% of patients and may be the initial manifestation.[3] The high-grade variant of this neoplasm is one of the most aggressive types of salivary gland carcinoma and is typified by local invasion, lymphatic and hematogenous spread, and poor prognosis.[3,7] In a retrospective review of 104 cases, 33% of patients developed local recurrence, and 46% of patients developed distant metastasis.[48]

Mucinous adenocarcinoma

Mucinous adenocarcinoma is a rare malignant neoplasm characterized by large amounts of extracellular epithelial mucin that contains cords, nests, and solitary epithelial cells. The incidence is unknown. Limited data indicate that most, if not all, occur in the major salivary glands, with the submandibular gland as the predominant site.[3,49] These tumors may be associated with dull pain and tenderness.[3,49] This neoplasm may be considered low grade.[3]

Malignant mixed tumors

The classification of malignant mixed tumors includes three distinct clinicopathological entities: carcinoma ex pleomorphic adenoma, carcinosarcoma, and metastasizing mixed tumor. Carcinoma ex pleomorphic adenoma accounts for most cases, whereas carcinosarcoma, a true malignant mixed tumor, and metastasizing mixed tumor are extremely rare.[3]

Carcinoma ex pleomorphic adenoma

Carcinoma ex pleomorphic adenoma, also known as carcinoma ex mixed tumor, shows histological evidence of arising from or in a benign pleomorphic adenoma.[50] Diagnosis requires the identification of benign tumor in the tissue sample.[51] The incidence or relative frequency of this tumor varies considerably depending on the study cited.[1] A review of material at the AFIP showed that carcinoma ex pleomorphic adenoma made up 8.8% of all mixed tumors and 4.6% of all malignant salivary gland tumors. It is the sixth most common malignant salivary gland tumor after mucoepidermoid carcinoma; adenocarcinoma, NOS; acinic cell carcinoma; polymorphous low-grade adenocarcinoma; and adenoid cystic carcinoma.[3] The neoplasm occurs primarily in the major salivary glands.[52]

The most common clinical presentation is a painless mass.[3] Approximately 33% of patients may experience facial paralysis.[53] Depending on the series cited, survival rates vary significantly: 25% to 65% at 5 years, 24% to 50% at 10 years, 10% to 35% at 15 years, and 0% to 38% at 20 years.[3] In addition to tumor stage, histological grade and degree of invasion help to determine prognosis.[54]

Carcinosarcoma

Carcinosarcoma, also known as true malignant mixed tumor, is a rare malignant salivary gland neoplasm that contains both carcinoma and sarcoma components. Either or both components are expressed in metastatic foci. Some carcinosarcomas develop de novo, while others develop in association with benign mixed tumor. This neoplasm is rare; only eight cases exist in the AFIP case files.[3] At one facility, only 11 cases were recorded over a 32-year period.[8] Most of these tumors occur in the major salivary glands.

Swelling, pain, nerve palsy, and ulceration have been frequent clinical findings. Carcinosarcoma is an aggressive, high-grade malignancy. In the largest series reported, which consisted of 12 cases, the average survival period was 3.6 years.[8]

Metastasizing mixed tumor

Metastasizing mixed tumor is a very rare, histologically benign salivary gland neoplasm that inexplicably metastasizes. Often, a long interval occurs between the diagnosis of the primary tumor and the metastases. The histological features are within the spectrum of features that typify pleomorphic adenoma.[3] Most of these tumors occur in the major salivary glands. The primary neoplasm is typically a single, well-defined mass. Recurrences, which may be multiple, have occurred as many as 26 years after excision of the primary neoplasm.[55]

Rare carcinomas
Primary squamous cell carcinoma

Primary squamous cell carcinoma, also known as primary epidermoid carcinoma, is a malignant epithelial neoplasm of the major salivary glands that is composed of squamous (i.e., epidermoid) cells. Diagnosis requires the exclusion of primary disease located in some other head and neck site; indeed, most squamous cell carcinomas of the major salivary glands represent metastatic disease.[3] This diagnosis is not made in minor salivary glands because distinction from the more common mucosal squamous cell carcinoma is not possible.[3] Previous exposure to ionizing radiation appears to increase the risk of developing this neoplasm.[11,56,57] The median time between radiation therapy and diagnosis of the neoplasm is approximately 15.5 years.[11] The reported frequency of this tumor among all major salivary gland tumors has varied from 0.9% to 4.7%.[3,10] In AFIP major salivary gland accessions from 1985 to 1996, primary squamous cell carcinoma accounted for 2.7% of all tumors; 5.4% of malignant tumors; and 2.5% and 2.8%, respectively, of all parotid and submandibular tumors.[3] The average age in the AFIP registry was 64 years.[3] This neoplasm occurs in the parotid gland almost nine times more often than in the submandibular gland.[3,57] There is a strong male predilection.[3,11,5759] This tumor is graded in a similar way to extrasalivary lesions according to the degree of differentiation, namely, low grade, intermediate grade, and high grade.[1]

Most patients present with an asymptomatic mass in the parotid region. Other symptoms may include a painful mass and facial nerve palsy.[57] The prognosis for this neoplasm is poor. In a 30-year retrospective analysis of 50 cases of squamous cell carcinoma of the salivary glands, survival rates at 5 years and 10 years were 24% and 18%, respectively.[57]

Epithelial-myoepithelial carcinoma

Epithelial-myoepithelial carcinoma is an uncommon, low-grade epithelial neoplasm composed of variable proportions of ductal and large, clear-staining, differentiated myoepithelial cells. It is also known as adenomyoepithelioma, clear cell adenoma, tubular solid adenoma, monomorphic clear cell tumor, glycogen-rich adenoma, glycogen-rich adenocarcinoma, clear cell carcinoma, or salivary duct carcinoma. The tumor represents approximately 1% of all epithelial salivary gland neoplasms.[3,60] It is predominantly a tumor of the parotid gland. In the AFIP case files, the mean age of patients was about 60 years, and about 60% of the patients were female.[3]

Localized swelling is commonly the only symptom, but occasionally patients experience facial weakness or pain.[61,62] Overall, epithelial-myoepithelial carcinoma is a low-grade carcinoma that recurs frequently, has a tendency to metastasize to periparotid and cervical lymph nodes, and occasionally results in distant metastasis and death.[60,6264]

Anaplastic small cell carcinoma

Anaplastic small cell carcinoma of the salivary glands was first described in 1972.[65] Subsequent histochemical and electron microscopic studies have supported the neuroendocrine nature of this tumor.[66,67] Microscopically, the tumor cells have oval, hyperchromatic nuclei and a scant amount of cytoplasm and are organized in sheets, strands, and nests. The mitotic rate is high. Neuroendocrine carcinomas are more frequently found in the minor salivary glands. These patients have a better survival rate than patients with small cell carcinomas of the lung.[68] The undifferentiated counterpart of this neoplasm is the small cell undifferentiated carcinoma.

Undifferentiated carcinomas

Undifferentiated carcinomas of salivary glands are a group of uncommon malignant epithelial neoplasms that lack the specific light-microscopic morphological features of other types of salivary gland carcinomas. These carcinomas are histologically similar to undifferentiated carcinomas that arise in other organs and tissues. Accordingly, metastatic carcinoma is a primary concern in the differential diagnosis of these neoplasms.[3]

Small cell undifferentiated carcinoma

Small cell undifferentiated carcinoma, also known as extrapulmonary oat cell carcinoma, is a rare, primary malignant tumor. With conventional light microscopy, it is composed of undifferentiated cells and, with ultrastructural or immunohistochemical studies, does not demonstrate neuroendocrine differentiation. This is the undifferentiated counterpart of anaplastic small cell carcinoma. For more information, see the Anaplastic small cell carcinoma section.

In an AFIP review of case files, small cell carcinoma represented 1.8% of all major salivary gland malignancies; the mean age of patients was 56 years.[3] In 50% of the cases, patients present with an asymptomatic parotid mass of 3 months’ or less duration.[6870] This is a high-grade neoplasm. In a retrospective review of 12 cases, a tumor size of more than 4 cm was found to be the most important predictor of behavior. In another small retrospective series, estimated survival rates at 2 and 5 years were 70% and 46%, respectively.[68]

Large cell undifferentiated carcinoma

Large cell undifferentiated carcinoma is a tumor in which features of acinar, ductal, epidermoid, or myoepithelial differentiation are absent under light microscopy, though occasionally, poorly formed duct-like structures are found. This neoplasm accounts for approximately 1% of all epithelial salivary gland neoplasms.[3,53,71,72] Most of these tumors occur in the parotid gland.[70,72] In AFIP data, the peak incidence is in the seventh to eighth decades of life.[3]

Rapid growth of a parotid swelling is a common clinical presentation.[59] This is a high-grade neoplasm that frequently metastasizes and has a poor prognosis. Patients with neoplasms of 4 cm or larger may have a particularly poor outcome.[70,72]

Lymphoepithelial carcinoma

Lymphoepithelial carcinoma, also known as undifferentiated carcinoma with lymphoid stroma and carcinoma ex lymphoepithelial lesion, is an undifferentiated tumor that is associated with a dense lymphoid stroma. An exceptionally high incidence of this tumor is found in the Inuit population.[3,73] This neoplasm has been associated with Epstein-Barr virus infection.[74,75] Of the occurrences, 80% are in the parotid gland.[3]

In addition to the presence of a parotid or submandibular mass, pain is a frequent symptom, and facial nerve palsy occurs in as many as 20% of patients.[76] Of the patients, more than 40% have metastases to cervical lymph nodes at initial presentation, 20% develop local recurrences or lymph node metastases, and 20% develop distant metastases within 3 years following therapy.[73,7678] For more information, see Cancer Pain.

Myoepithelial carcinoma

Myoepithelioma carcinoma is a rare, malignant salivary gland neoplasm in which the tumor cells almost exclusively manifest myoepithelial differentiation. This neoplasm represents the malignant counterpart of benign myoepithelioma.[3] The largest series reported involved 25 cases.[79] Approximately 66% of the tumors occur in the parotid gland.[3,74] The mean age of patients is 55 years.[79]

Most patients present with the primary complaint of a painless mass.[79] This is an intermediate grade to high-grade carcinoma.[3,79] Histological grade does not appear to correlate well with clinical behavior. For instance, tumors with a low-grade histological appearance may behave aggressively.[79]

Adenosquamous carcinoma

Adenosquamous carcinoma is an extremely rare malignant neoplasm that simultaneously arises from surface mucosal epithelium and salivary gland ductal epithelium. The carcinoma shows histopathological features of both squamous cell carcinoma and adenocarcinoma. Only a handful of reports describe this tumor.[3]

In addition to swelling, adenosquamous carcinoma produces visible changes in the mucosa, including erythema, ulceration, and induration. Pain frequently accompanies ulceration. Limited data indicate that this is a highly aggressive neoplasm with a poor prognosis.[3]

Nonepithelial Neoplasms

Lymphomas and benign lymphoepithelial lesion

Lymphomas of the major salivary glands are characteristically of the non-Hodgkin type. In an AFIP review of case files, non-Hodgkin lymphoma accounted for 16.3% of all malignant tumors that occurred in the major salivary glands. Disease in the parotid gland accounted for about 80% of all cases.[3]

Patients with benign lymphoepithelial lesion (e.g., Mikulicz disease), which is a manifestation of the autoimmune disease Sjögren syndrome, are at an increased risk of developing non-Hodgkin lymphoma.[8084] Benign lymphoepithelial lesion is clinically characterized by diffuse and bilateral enlargement of the salivary and lacrimal glands.[23] Morphologically, a salivary gland lesion is composed of prominent myoepithelial islands surrounded by a lymphocytic infiltrate. Germinal centers are often present in the lymphocytic infiltrate.[23] Immunophenotypically and genotypically, the lymphocytic infiltrate is composed of B-lymphocytes and T-lymphocytes, which are polyclonal. In some instances, the B-cell lymphocytic infiltrate can undergo clonal expansion and evolve into frank non-Hodgkin lymphoma. Most of the non-Hodgkin lymphomas arising in a background of benign lymphoepithelial lesions are marginal zone lymphomas of mucosa-associated lymphoid tissue (MALT).[8184] MALT lymphomas of the salivary glands, like their counterparts in other anatomical sites, typically display relatively indolent clinical behavior.[3,85]

Primary non-MALT lymphomas of the salivary glands may also occur and appear to have a prognosis similar to those in patients who have histologically identical nodal lymphomas.[86,87] Unlike non-Hodgkin lymphoma, involvement of the major salivary glands by Hodgkin lymphoma is rare. Most tumors occur in the parotid gland.[3] The most common histological types encountered are the nodular sclerosing and lymphocyte-predominant variants.[88,89]

Mesenchymal neoplasms

Mesenchymal neoplasms account for 1.9% to 5% of all neoplasms that occur within the major salivary glands.[90,91] These cellular classifications pertain to major salivary gland tumors. Because the minor salivary glands are small and embedded within fibrous connective tissue, fat, and skeletal muscle, the origin of a mesenchymal neoplasm from stroma cannot be determined.[3] The types of benign mesenchymal salivary gland neoplasms include hemangiomas, lipomas, and lymphangiomas.

Malignant mesenchymal salivary gland neoplasms include malignant schwannomas, hemangiopericytomas, malignant fibrous histiocytomas, rhabdomyosarcomas, and fibrosarcomas, among others. In the major salivary glands, these neoplasms represent approximately 0.5% of all benign and malignant salivary gland tumors and approximately 1.5% of all malignant tumors.[90,92,93] It is important to establish a primary salivary gland origin for these tumors by excluding the possibilities of metastasis and direct extension from other sites. In addition, the diagnosis of salivary gland carcinosarcoma should be excluded.[3] Primary salivary gland sarcomas behave like their soft tissue counterparts, in which prognosis is related to sarcoma type, histological grade, tumor size, and stage.[93,94] For more information, see Soft Tissue Sarcoma Treatment. A comprehensive review of salivary gland mesenchymal neoplasms can be found elsewhere.[95]

Malignant Secondary Neoplasms

Malignant neoplasms whose origins lie outside the salivary glands may involve the major salivary glands by the following routes:[3]

  1. Direct invasion from cancers that lie adjacent to the salivary glands.
  2. Hematogenous metastases from distant primary tumors.
  3. Lymphatic metastases to lymph nodes within the salivary gland.

Direct invasion of nonsalivary gland tumors into the major salivary glands is principally from squamous cell and basal cell carcinomas of the overlying skin.

Approximately 80% of metastases to the major salivary glands may be from primary tumors elsewhere in the head and neck; the remaining 20% may be from infraclavicular sites.[96,97] The parotid gland is the site of 80% to 90% of the metastases, and the remainder involve the submandibular gland.[97,98] In a decade-long AFIP experience, metastatic tumors constituted approximately 10% of malignant neoplasms in the major salivary glands, exclusive of malignant lymphomas.[3] Most metastatic primary tumors to the major salivary glands are squamous cell carcinomas and melanomas from the head and neck that presumably reach the parotid gland via the lymphatic system. Infraclavicular primary tumors, such as those in the lung, kidney, and breast, reach the salivary glands by a hematogenous route.[9799] The peak incidence for metastatic tumors in the salivary glands is reported to be in the seventh decade of life.[3]

References
  1. Speight PM, Barrett AW: Salivary gland tumours. Oral Dis 8 (5): 229-40, 2002. [PUBMED Abstract]
  2. Seifert G, Donath K: Hybrid tumours of salivary glands. Definition and classification of five rare cases. Eur J Cancer B Oral Oncol 32B (4): 251-9, 1996. [PUBMED Abstract]
  3. Ellis GL, Auclair PL: Tumors of the Salivary Glands. Armed Forces Institute of Pathology, 1996. Atlas of Tumor Pathology, 3.
  4. Cheuk W, Chan JKC: Salivary gland tumors. In: Fletcher CDM, ed.: Diagnostic Histopathology of Tumors. 3rd ed. Churchill Livingstone, 2007, pp 239-326.
  5. Spiro RH, Huvos AG, Berk R, et al.: Mucoepidermoid carcinoma of salivary gland origin. A clinicopathologic study of 367 cases. Am J Surg 136 (4): 461-8, 1978. [PUBMED Abstract]
  6. Goode RK, Corio RL: Oncocytic adenocarcinoma of salivary glands. Oral Surg Oral Med Oral Pathol 65 (1): 61-6, 1988. [PUBMED Abstract]
  7. Guzzo M, Di Palma S, Grandi C, et al.: Salivary duct carcinoma: clinical characteristics and treatment strategies. Head Neck 19 (2): 126-33, 1997. [PUBMED Abstract]
  8. Stephen J, Batsakis JG, Luna MA, et al.: True malignant mixed tumors (carcinosarcoma) of salivary glands. Oral Surg Oral Med Oral Pathol 61 (6): 597-602, 1986. [PUBMED Abstract]
  9. Auclair PL, Ellis GL, Gnepp DR, et al.: Salivary gland neoplasms: general considerations. In: Ellis GL, Auclair PL, Gnepp DR, eds.: Surgical Pathology of the Salivary Glands. Saunders, 1991, pp 135-64.
  10. Eveson JW, Cawson RA: Salivary gland tumours. A review of 2410 cases with particular reference to histological types, site, age and sex distribution. J Pathol 146 (1): 51-8, 1985. [PUBMED Abstract]
  11. Spitz MR, Batsakis JG: Major salivary gland carcinoma. Descriptive epidemiology and survival of 498 patients. Arch Otolaryngol 110 (1): 45-9, 1984. [PUBMED Abstract]
  12. Goode RK, Auclair PL, Ellis GL: Mucoepidermoid carcinoma of the major salivary glands: clinical and histopathologic analysis of 234 cases with evaluation of grading criteria. Cancer 82 (7): 1217-24, 1998. [PUBMED Abstract]
  13. Guzzo M, Andreola S, Sirizzotti G, et al.: Mucoepidermoid carcinoma of the salivary glands: clinicopathologic review of 108 patients treated at the National Cancer Institute of Milan. Ann Surg Oncol 9 (7): 688-95, 2002. [PUBMED Abstract]
  14. Neville BW, Damm DD, Weir JC, et al.: Labial salivary gland tumors. Cancer 61 (10): 2113-6, 1988. [PUBMED Abstract]
  15. Auclair PL, Goode RK, Ellis GL: Mucoepidermoid carcinoma of intraoral salivary glands. Evaluation and application of grading criteria in 143 cases. Cancer 69 (8): 2021-30, 1992. [PUBMED Abstract]
  16. Brandwein MS, Ivanov K, Wallace DI, et al.: Mucoepidermoid carcinoma: a clinicopathologic study of 80 patients with special reference to histological grading. Am J Surg Pathol 25 (7): 835-45, 2001. [PUBMED Abstract]
  17. Nordkvist A, Gustafsson H, Juberg-Ode M, et al.: Recurrent rearrangements of 11q14-22 in mucoepidermoid carcinoma. Cancer Genet Cytogenet 74 (2): 77-83, 1994. [PUBMED Abstract]
  18. Horsman DE, Berean K, Durham JS: Translocation (11;19)(q21;p13.1) in mucoepidermoid carcinoma of salivary gland. Cancer Genet Cytogenet 80 (2): 165-6, 1995. [PUBMED Abstract]
  19. El-Naggar AK, Lovell M, Killary AM, et al.: A mucoepidermoid carcinoma of minor salivary gland with t(11;19)(q21;p13.1) as the only karyotypic abnormality. Cancer Genet Cytogenet 87 (1): 29-33, 1996. [PUBMED Abstract]
  20. Tonon G, Modi S, Wu L, et al.: t(11;19)(q21;p13) translocation in mucoepidermoid carcinoma creates a novel fusion product that disrupts a Notch signaling pathway. Nat Genet 33 (2): 208-13, 2003. [PUBMED Abstract]
  21. Allenspach EJ, Maillard I, Aster JC, et al.: Notch signaling in cancer. Cancer Biol Ther 1 (5): 466-76, 2002 Sep-Oct. [PUBMED Abstract]
  22. Brookstone MS, Huvos AG: Central salivary gland tumors of the maxilla and mandible: a clinicopathologic study of 11 cases with an analysis of the literature. J Oral Maxillofac Surg 50 (3): 229-36, 1992. [PUBMED Abstract]
  23. Major and minor salivary glands. In: Rosai J, ed.: Ackerman’s Surgical Pathology. 8th ed. Mosby, 1996, pp 815-56.
  24. Batsakis JG, Luna MA, el-Naggar A: Histopathologic grading of salivary gland neoplasms: III. Adenoid cystic carcinomas. Ann Otol Rhinol Laryngol 99 (12): 1007-9, 1990. [PUBMED Abstract]
  25. Tomich CE: Adenoid cystic carcinoma. In: Ellis GL, Auclair PL, Gnepp DR, eds.: Surgical Pathology of the Salivary Glands. Saunders, 1991, pp 333-49.
  26. Perzin KH, Gullane P, Clairmont AC: Adenoid cystic carcinomas arising in salivary glands: a correlation of histologic features and clinical course. Cancer 42 (1): 265-82, 1978. [PUBMED Abstract]
  27. Spiro RH: The controversial adenoid cystic carcinoma. Clinical considerations. In: McGurk M, Renehan AG, eds.: Controversies in the Management of Salivary Gland Disease. Oxford University Press, 2001, pp 207-11.
  28. Friedrich RE, Bleckmann V: Adenoid cystic carcinoma of salivary and lacrimal gland origin: localization, classification, clinical pathological correlation, treatment results and long-term follow-up control in 84 patients. Anticancer Res 23 (2A): 931-40, 2003 Mar-Apr. [PUBMED Abstract]
  29. Spiro RH, Huvos AG: Stage means more than grade in adenoid cystic carcinoma. Am J Surg 164 (6): 623-8, 1992. [PUBMED Abstract]
  30. Hamper K, Lazar F, Dietel M, et al.: Prognostic factors for adenoid cystic carcinoma of the head and neck: a retrospective evaluation of 96 cases. J Oral Pathol Med 19 (3): 101-7, 1990. [PUBMED Abstract]
  31. Lewis JE, Olsen KD, Weiland LH: Acinic cell carcinoma. Clinicopathologic review. Cancer 67 (1): 172-9, 1991. [PUBMED Abstract]
  32. Waldron CA, el-Mofty SK, Gnepp DR: Tumors of the intraoral minor salivary glands: a demographic and histologic study of 426 cases. Oral Surg Oral Med Oral Pathol 66 (3): 323-33, 1988. [PUBMED Abstract]
  33. Vincent SD, Hammond HL, Finkelstein MW: Clinical and therapeutic features of polymorphous low-grade adenocarcinoma. Oral Surg Oral Med Oral Pathol 77 (1): 41-7, 1994. [PUBMED Abstract]
  34. Evans HL, Luna MA: Polymorphous low-grade adenocarcinoma: a study of 40 cases with long-term follow up and an evaluation of the importance of papillary areas. Am J Surg Pathol 24 (10): 1319-28, 2000. [PUBMED Abstract]
  35. Spiro RH, Huvos AG, Strong EW: Adenocarcinoma of salivary origin. Clinicopathologic study of 204 patients. Am J Surg 144 (4): 423-31, 1982. [PUBMED Abstract]
  36. Matsuba HM, Mauney M, Simpson JR, et al.: Adenocarcinomas of major and minor salivary gland origin: a histopathologic review of treatment failure patterns. Laryngoscope 98 (7): 784-8, 1988. [PUBMED Abstract]
  37. Muller S, Barnes L: Basal cell adenocarcinoma of the salivary glands. Report of seven cases and review of the literature. Cancer 78 (12): 2471-7, 1996. [PUBMED Abstract]
  38. Ellis GL, Wiscovitch JG: Basal cell adenocarcinomas of the major salivary glands. Oral Surg Oral Med Oral Pathol 69 (4): 461-9, 1990. [PUBMED Abstract]
  39. Simpson RH, Sarsfield PT, Clarke T, et al.: Clear cell carcinoma of minor salivary glands. Histopathology 17 (5): 433-8, 1990. [PUBMED Abstract]
  40. Ogawa I, Nikai H, Takata T, et al.: Clear cell tumors of minor salivary gland origin. An immunohistochemical and ultrastructural analysis. Oral Surg Oral Med Oral Pathol 72 (2): 200-7, 1991. [PUBMED Abstract]
  41. Milchgrub S, Gnepp DR, Vuitch F, et al.: Hyalinizing clear cell carcinoma of salivary gland. Am J Surg Pathol 18 (1): 74-82, 1994. [PUBMED Abstract]
  42. Ellis GL, Auclair PL, Gnepp DR, et al.: Other malignant epithelial neoplasms. In: Ellis GL, Auclair PL, Gnepp DR, eds.: Surgical Pathology of the Salivary Glands. Saunders, 1991, pp 455-88.
  43. Gnepp DR: Sebaceous neoplasms of salivary gland origin: a review. Pathol Annu 18 Pt 1: 71-102, 1983. [PUBMED Abstract]
  44. Gnepp DR, Brannon R: Sebaceous neoplasms of salivary gland origin. Report of 21 cases. Cancer 53 (10): 2155-70, 1984. [PUBMED Abstract]
  45. Linhartová A: Sebaceous glands in salivary gland tissue. Arch Pathol 98 (5): 320-4, 1974. [PUBMED Abstract]
  46. Sugimoto T, Wakizono S, Uemura T, et al.: Malignant oncocytoma of the parotid gland: a case report with an immunohistochemical and ultrastructural study. J Laryngol Otol 107 (1): 69-74, 1993. [PUBMED Abstract]
  47. Delgado R, Klimstra D, Albores-Saavedra J: Low grade salivary duct carcinoma. A distinctive variant with a low grade histology and a predominant intraductal growth pattern. Cancer 78 (5): 958-67, 1996. [PUBMED Abstract]
  48. Barnes L, Rao U, Krause J, et al.: Salivary duct carcinoma. Part I. A clinicopathologic evaluation and DNA image analysis of 13 cases with review of the literature. Oral Surg Oral Med Oral Pathol 78 (1): 64-73, 1994. [PUBMED Abstract]
  49. Osaki T, Hirota J, Ohno A, et al.: Mucinous adenocarcinoma of the submandibular gland. Cancer 66 (8): 1796-801, 1990. [PUBMED Abstract]
  50. Röijer E, Nordkvist A, Ström AK, et al.: Translocation, deletion/amplification, and expression of HMGIC and MDM2 in a carcinoma ex pleomorphic adenoma. Am J Pathol 160 (2): 433-40, 2002. [PUBMED Abstract]
  51. LiVolsi VA, Perzin KH: Malignant mixed tumors arising in salivary glands. I. Carcinomas arising in benign mixed tumors: a clinicopathologic study. Cancer 39 (5): 2209-30, 1977. [PUBMED Abstract]
  52. Gnepp DR: Malignant mixed tumors of the salivary glands: a review. Pathol Annu 28 Pt 1: 279-328, 1993. [PUBMED Abstract]
  53. Seifert G: [Diseases of the Salivary Glands: Pathology, Diagnosis, Treatment, Facial Nerve Surgery]; translated by Philip M. Stell. Thieme, 1986.
  54. Brandwein MS, Ferlito A, Bradley PJ, et al.: Diagnosis and classification of salivary neoplasms: pathologic challenges and relevance to clinical outcomes. Acta Otolaryngol 122 (7): 758-64, 2002. [PUBMED Abstract]
  55. Wenig BM, Hitchcock CL, Ellis GL, et al.: Metastasizing mixed tumor of salivary glands. A clinicopathologic and flow cytometric analysis. Am J Surg Pathol 16 (9): 845-58, 1992. [PUBMED Abstract]
  56. Schneider AB, Favus MJ, Stachura ME, et al.: Salivary gland neoplasms as a late consequence of head and neck irradiation. Ann Intern Med 87 (2): 160-4, 1977. [PUBMED Abstract]
  57. Shemen LJ, Huvos AG, Spiro RH: Squamous cell carcinoma of salivary gland origin. Head Neck Surg 9 (4): 235-40, 1987 Mar-Apr. [PUBMED Abstract]
  58. Sterman BM, Kraus DH, Sebek BA, et al.: Primary squamous cell carcinoma of the parotid gland. Laryngoscope 100 (2 Pt 1): 146-8, 1990. [PUBMED Abstract]
  59. Gaughan RK, Olsen KD, Lewis JE: Primary squamous cell carcinoma of the parotid gland. Arch Otolaryngol Head Neck Surg 118 (8): 798-801, 1992. [PUBMED Abstract]
  60. Batsakis JG, el-Naggar AK, Luna MA: Epithelial-myoepithelial carcinoma of salivary glands. Ann Otol Rhinol Laryngol 101 (6): 540-2, 1992. [PUBMED Abstract]
  61. Daley TD, Wysocki GP, Smout MS, et al.: Epithelial-myoepithelial carcinoma of salivary glands. Oral Surg Oral Med Oral Pathol 57 (5): 512-9, 1984. [PUBMED Abstract]
  62. Collina G, Gale N, Visonà A, et al.: Epithelial-myoepithelial carcinoma of the parotid gland: a clinico-pathologic and immunohistochemical study of seven cases. Tumori 77 (3): 257-63, 1991. [PUBMED Abstract]
  63. Simpson RH, Clarke TJ, Sarsfield PT, et al.: Epithelial-myoepithelial carcinoma of salivary glands. J Clin Pathol 44 (5): 419-23, 1991. [PUBMED Abstract]
  64. Noel S, Brozna JP: Epithelial-myoepithelial carcinoma of salivary gland with metastasis to lung: report of a case and review of the literature. Head Neck 14 (5): 401-6, 1992 Sep-Oct. [PUBMED Abstract]
  65. Koss LG, Spiro RH, Hajdu S: Small cell (oat cell) carcinoma of minor salivary gland origin. Cancer 30 (3): 737-41, 1972. [PUBMED Abstract]
  66. Gnepp DR, Wick MR: Small cell carcinoma of the major salivary glands. An immunohistochemical study. Cancer 66 (1): 185-92, 1990. [PUBMED Abstract]
  67. Perez-Ordonez B, Caruana SM, Huvos AG, et al.: Small cell neuroendocrine carcinoma of the nasal cavity and paranasal sinuses. Hum Pathol 29 (8): 826-32, 1998. [PUBMED Abstract]
  68. Gnepp DR, Corio RL, Brannon RB: Small cell carcinoma of the major salivary glands. Cancer 58 (3): 705-14, 1986. [PUBMED Abstract]
  69. Scher RL, Feldman PS, Levine PA: Small-cell carcinoma of the parotid gland with neuroendocrine features. Arch Otolaryngol Head Neck Surg 114 (3): 319-21, 1988. [PUBMED Abstract]
  70. Hui KK, Luna MA, Batsakis JG, et al.: Undifferentiated carcinomas of the major salivary glands. Oral Surg Oral Med Oral Pathol 69 (1): 76-83, 1990. [PUBMED Abstract]
  71. Spiro RH: Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head Neck Surg 8 (3): 177-84, 1986 Jan-Feb. [PUBMED Abstract]
  72. Batsakis JG, Luna MA: Undifferentiated carcinomas of salivary glands. Ann Otol Rhinol Laryngol 100 (1): 82-4, 1991. [PUBMED Abstract]
  73. Bosch JD, Kudryk WH, Johnson GH: The malignant lymphoepithelial lesion of the salivary glands. J Otolaryngol 17 (4): 187-90, 1988. [PUBMED Abstract]
  74. Hamilton-Dutoit SJ, Therkildsen MH, Neilsen NH, et al.: Undifferentiated carcinoma of the salivary gland in Greenlandic Eskimos: demonstration of Epstein-Barr virus DNA by in situ nucleic acid hybridization. Hum Pathol 22 (8): 811-5, 1991. [PUBMED Abstract]
  75. Leung SY, Chung LP, Yuen ST, et al.: Lymphoepithelial carcinoma of the salivary gland: in situ detection of Epstein-Barr virus. J Clin Pathol 48 (11): 1022-7, 1995. [PUBMED Abstract]
  76. Borg MF, Benjamin CS, Morton RP, et al.: Malignant lympho-epithelial lesion of the salivary gland: a case report and review of the literature. Australas Radiol 37 (3): 288-91, 1993. [PUBMED Abstract]
  77. Saw D, Lau WH, Ho JH, et al.: Malignant lymphoepithelial lesion of the salivary gland. Hum Pathol 17 (9): 914-23, 1986. [PUBMED Abstract]
  78. Cleary KR, Batsakis JG: Undifferentiated carcinoma with lymphoid stroma of the major salivary glands. Ann Otol Rhinol Laryngol 99 (3 Pt 1): 236-8, 1990. [PUBMED Abstract]
  79. Savera AT, Sloman A, Huvos AG, et al.: Myoepithelial carcinoma of the salivary glands: a clinicopathologic study of 25 patients. Am J Surg Pathol 24 (6): 761-74, 2000. [PUBMED Abstract]
  80. Kassan SS, Thomas TL, Moutsopoulos HM, et al.: Increased risk of lymphoma in sicca syndrome. Ann Intern Med 89 (6): 888-92, 1978. [PUBMED Abstract]
  81. Abbondanzo SL: Extranodal marginal-zone B-cell lymphoma of the salivary gland. Ann Diagn Pathol 5 (4): 246-54, 2001. [PUBMED Abstract]
  82. Ihrler S, Baretton GB, Menauer F, et al.: Sjögren’s syndrome and MALT lymphomas of salivary glands: a DNA-cytometric and interphase-cytogenetic study. Mod Pathol 13 (1): 4-12, 2000. [PUBMED Abstract]
  83. Harris NL: Lymphoid proliferations of the salivary glands. Am J Clin Pathol 111 (1 Suppl 1): S94-103, 1999. [PUBMED Abstract]
  84. DiGiuseppe JA, Corio RL, Westra WH: Lymphoid infiltrates of the salivary glands: pathology, biology and clinical significance. Curr Opin Oncol 8 (3): 232-7, 1996. [PUBMED Abstract]
  85. Harris NL: Extranodal lymphoid infiltrates and mucosa-associated lymphoid tissue (MALT). A unifying concept. Am J Surg Pathol 15 (9): 879-84, 1991. [PUBMED Abstract]
  86. Burke JS: Waldeyer’s ring, sinonasal region, salivary gland, thyroid gland, central nervous system, and other extranodal lymphomas and lymphoid hyperplasias. In: Knowles DM, ed.: Neoplastic Hematopathology. Williams & Wilkins, 1992, pp 1047-79.
  87. Salhany KE, Pietra GG: Extranodal lymphoid disorders. Am J Clin Pathol 99 (4): 472-85, 1993. [PUBMED Abstract]
  88. Schmid U, Helbron D, Lennert K: Primary malignant lymphomas localized in salivary glands. Histopathology 6 (6): 673-87, 1982. [PUBMED Abstract]
  89. Gleeson MJ, Bennett MH, Cawson RA: Lymphomas of salivary glands. Cancer 58 (3): 699-704, 1986. [PUBMED Abstract]
  90. Seifert G, Oehne H: [Mesenchymal (non-epithelial) salivary gland tumors. Analysis of 167 tumor cases of the salivary gland register] Laryngol Rhinol Otol (Stuttg) 65 (9): 485-91, 1986. [PUBMED Abstract]
  91. Auclair PL, Ellis GL, Gnepp DR, et al.: Salivary gland neoplasms: general considerations. In: Ellis GL, Auclair PL, Gnepp DR, eds.: Surgical Pathology of the Salivary Glands. Saunders, 1991, pp 135-64.
  92. Auclair PL, Ellis GL: Nonlymphoid sarcomas of the major salivary glands. In: Ellis GL, Auclair PL, Gnepp DR, eds.: Surgical Pathology of the Salivary Glands. Saunders, 1991, pp 514-27.
  93. Luna MA, Tortoledo ME, Ordóñez NG, et al.: Primary sarcomas of the major salivary glands. Arch Otolaryngol Head Neck Surg 117 (3): 302-6, 1991. [PUBMED Abstract]
  94. Auclair PL, Langloss JM, Weiss SW, et al.: Sarcomas and sarcomatoid neoplasms of the major salivary gland regions. A clinicopathologic and immunohistochemical study of 67 cases and review of the literature. Cancer 58 (6): 1305-15, 1986. [PUBMED Abstract]
  95. Weiss SW, Goldblum JR: Enzinger and Weiss’s Soft Tissue Tumors. 4th ed. Mosby, 2001.
  96. Cantera JM, Hernandez AV: Bilateral parotid gland metastasis as the initial presentation of a small cell lung carcinoma. J Oral Maxillofac Surg 47 (11): 1199-201, 1989. [PUBMED Abstract]
  97. Gnepp DR: Metastatic disease to the major salivary glands. In: Ellis GL, Auclair PL, Gnepp DR, eds.: Surgical Pathology of the Salivary Glands. Saunders, 1991, pp 560-9.
  98. Seifert G, Hennings K, Caselitz J: Metastatic tumors to the parotid and submandibular glands–analysis and differential diagnosis of 108 cases. Pathol Res Pract 181 (6): 684-92, 1986. [PUBMED Abstract]
  99. Batsakis JG, Bautina E: Metastases to major salivary glands. Ann Otol Rhinol Laryngol 99 (6 Pt 1): 501-3, 1990. [PUBMED Abstract]

Stage Information for Salivary Gland Cancer

In general, tumors of the major salivary glands are staged according to size, extraparenchymal extension, lymph node involvement (in parotid tumors, whether or not the facial nerve is involved), and presence of metastases.[14] Tumors arising in the minor salivary glands are staged according to the anatomical site of origin (e.g., oral cavity and sinuses).

Clinical stage, particularly tumor size, may be the critical factor in determining the outcome of salivary gland cancer and may be more important than histological grade.[5,6] Diagnostic imaging studies may be used in staging. With excellent spatial resolution and superior soft tissue contrast, magnetic resonance imaging (MRI) offers advantages over computed tomographic scanning in the detection and localization of head and neck tumors. Overall, MRI is the preferred modality for evaluation of suspected neoplasms of the salivary glands.[7]

American Joint Committee on Cancer (AJCC) Stage Groupings and TNM Definitions

The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define salivary gland cancer.[5]

Table 1. Definitions of TNM Stage 0a
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Major salivary glands (parotid, submandibular, and sublingual). In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 95–101.
bA designation of U or L may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L). Similarly, clinical and pathological extranodal extension (ENE) should be recorded as ENE(–) or ENE(+).
0 Tis, N0, M0 Tis = Carcinoma in situ.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 2. Definitions of TNM Stage Ia
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Major salivary glands. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 95–101.
bA designation of U or L may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L). Similarly, clinical and pathological extranodal extension (ENE) should be recorded as ENE(–) or ENE(+).
cExtraparenchymal extension is clinical or macroscopic evidence of invasion of soft tissues. Microscopic evidence alone does not constitute extraparenchymal extension for classification purposes.
I T1, N0, M0 T1 = Tumor ≤2 cm in greatest dimension without extraparenchymal extension.c
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 3. Definitions of TNM Stage IIa
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Major salivary glands. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 95–101.
bA designation of U or L may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L). Similarly, clinical and pathological extranodal extension (ENE) should be recorded as ENE(–) or ENE(+).
cExtraparenchymal extension is clinical or macroscopic evidence of invasion of soft tissues. Microscopic evidence alone does not constitute extraparenchymal extension for classification purposes.
II T2, N0, M0 T2 = Tumor >2 cm but ≤4 cm in greatest dimension without extraparenchymal extension.c
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 4. Definitions of TNM Stage IIIa
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis; ENE = extranodal extension.
aReprinted with permission from AJCC: Major salivary glands. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 95–101.
bA designation of U or L may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L). Similarly, clinical and pathological extranodal extension (ENE) should be recorded as ENE(–) or ENE(+).
cExtraparenchymal extension is clinical or macroscopic evidence of invasion of soft tissues. Microscopic evidence alone does not constitute extraparenchymal extension for classification purposes.
III T3, N0, M0 T3 = Tumor >4 cm and/or tumor having extraparenchymal extension.c
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
T0, T1, T2, T3, N1, M0 T0 = No evidence of primary tumor.
T1 = Tumor ≤2 cm in greatest dimension without extraparenchymal extension.c
T2 = Tumor >2 cm but ≤4 cm in greatest dimension without extraparenchymal extension.c
T3 = Tumor >4 cm and/or tumor having extraparenchymal extension.c
N1 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(–).
M0 = No distant metastasis.
Table 5. Definitions of TNM Stages IVA, IVB, and IVCa
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis; ENE = extranodal extension.
aReprinted with permission from AJCC: Major salivary glands. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 95–101.
bA designation of U or L may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L). Similarly, clinical and pathological ENE should be recorded as ENE(–) or ENE(+).
cExtraparenchymal extension is clinical or macroscopic evidence of invasion of soft tissues. Microscopic evidence alone does not constitute extraparenchymal extension for classification purposes.
IVA T4a, N0, N1, M0 T4a = Moderately advanced disease. Tumor invades skin, mandible, ear canal, and/or facial nerve.
N0 = No regional lymph node metastasis.
N1 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(–).
M0 = No distant metastasis.
T0, T1, T2, T3, T4a, N2, M0 T0, T1, T2, T3, T4a = See descriptions below in this table, Stage IVB.
N2 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(+); or >3 cm but ≤6 cm in greatest dimension and ENE(–); or metastases in multiple ipsilateral lymph nodes, none >6 cm in greatest dimension and ENE(–); or in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(–).
–N2a = Metastasis in a single ipsilateral or contralateral node ≤3 cm in greatest dimension and ENE(+) or a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(–).
–N2b = Metastases in multiple ipsilateral nodes, none >6 cm in greatest dimension and ENE(–).
–N2c = Metastases in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(–).
M0 = No distant metastasis.
IVB Any T, N3, M0 TX = Primary tumor cannot be assessed.
T0 = No evidence of primary tumor.
Tis = Carcinoma in situ.
T1 = Tumor ≤2 cm in greatest dimension without extraparenchymal extension.c
T2 = Tumor >2 cm but ≤4 cm in greatest dimension without extraparenchymal extension.c
T3 = Tumor >4 cm and/or tumor having extraparenchymal extension.c
T4 = Moderately advanced or very advanced disease.
–T4a = Moderately advanced disease. Tumor invades skin, mandible, ear canal, and/or facial nerve.
–T4b = Very advanced disease. Tumor invades skull base and/or pterygoid plates and/or encases carotid artery.
N3 = Metastasis in a lymph node >6 cm in greatest dimension and ENE(–); or metastasis in any node(s) with clinically overt ENE(+).
–N3a = Metastasis in a lymph node >6 cm in greatest dimension and ENE(–).
–N3b = Metastasis in any node(s) with clinically overt ENE(+).
M0 = No distant metastasis.
T4b, Any N, M0 T4b = Very advanced disease. Tumor invades skull base and/or pterygoid plates and/or encases carotid artery.
NX = Regional lymph nodes cannot be assessed.
N0 = No regional lymph node metastasis.
N1 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(–).
N2 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(+); or >3 cm but ≤6 cm in greatest dimension and ENE(–); or metastases in multiple ipsilateral lymph nodes, none >6 cm in greatest dimension and ENE(–); or in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(–).
–N2a = Metastasis in a single ipsilateral or contralateral node ≤3 cm in greatest dimension and ENE(+) or a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(–).
–N2b = Metastases in multiple ipsilateral nodes, none >6 cm in greatest dimension and ENE(–).
–N2c = Metastases in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(–).
N3 = Metastasis in a lymph node >6 cm in greatest dimension and ENE(–); or metastasis in any node(s) with clinically overt with ENE(+).
–N3a = Metastasis in a lymph node >6 cm in greatest dimension and ENE(–).
–N3b = Metastasis in any node(s) with clinically overt ENE(+).
M0 = No distant metastasis.
IVC Any T, Any N, M1 Any T = See descriptions above in this table, Stage IVB.
Any N = See descriptions above in this table, Stage IVB.
M1 = Distant metastasis.
References
  1. Spiro RH, Huvos AG, Strong EW: Cancer of the parotid gland. A clinicopathologic study of 288 primary cases. Am J Surg 130 (4): 452-9, 1975. [PUBMED Abstract]
  2. Fu KK, Leibel SA, Levine ML, et al.: Carcinoma of the major and minor salivary glands: analysis of treatment results and sites and causes of failures. Cancer 40 (6): 2882-90, 1977. [PUBMED Abstract]
  3. Levitt SH, McHugh RB, Gómez-Marin O, et al.: Clinical staging system for cancer of the salivary gland: a retrospective study. Cancer 47 (11): 2712-24, 1981. [PUBMED Abstract]
  4. Kuhel W, Goepfert H, Luna M, et al.: Adenoid cystic carcinoma of the palate. Arch Otolaryngol Head Neck Surg 118 (3): 243-7, 1992. [PUBMED Abstract]
  5. Major salivary glands. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 95–101.
  6. Spiro RH: Factors affecting survival in salivary gland cancers. In: McGurk M, Renehan AG, eds.: Controversies in the Management of Salivary Gland Disease. Oxford University Press, 2001, pp 143-50.
  7. Shah GV: MR imaging of salivary glands. Magn Reson Imaging Clin N Am 10 (4): 631-62, 2002. [PUBMED Abstract]

Treatment Option Overview for Salivary Gland Cancer

The minimum therapy for patients with low-grade malignancies of the superficial portion of the parotid gland is a superficial parotidectomy. For all other lesions, a total parotidectomy is often indicated. The facial nerve or its branches should be resected if involved by tumor; repair can be done simultaneously. Evidence suggests that postoperative radiation therapy augments surgical resection, particularly for the high-grade neoplasms, when margins are close or involved, when tumors are large, or when histological evidence of lymph node metastases is present.[18] Clinical trials in the United States and England indicated that fast neutron-beam radiation therapy improves disease-free survival and overall survival in patients with unresectable tumors or for patients with recurrent neoplasms.[912] The availability of facilities with fast neutron-beam radiation therapy is limited in the United States. Accelerated hyperfractionated photon-beam radiation therapy has also resulted in high rates of long-term local regional controls.[13,14] The use of chemotherapy for malignant salivary gland tumors remains under evaluation.[1519]

References
  1. Myers EN, Suen JY, eds.: Cancer of the Head and Neck. 3rd ed. Saunders, 1996.
  2. Freund HR: Principles of Head and Neck Surgery. 2nd ed. Appleton-Century-Crofts, 1979.
  3. Lore JM: An Atlas of Head and Neck Surgery. 3rd ed. Saunders, 1988.
  4. Million RR, Cassisi NJ, eds.: Management of Head and Neck Cancer: A Multidisciplinary Approach. Lippincott, 1994.
  5. Wang CC, ed.: Radiation Therapy for Head and Neck Neoplasms. 3rd ed. Wiley-Liss, 1997.
  6. Cummings CW, Fredrickson JM, Harker LA, et al.: Otolaryngology – Head and Neck Surgery. Mosby-Year Book, Inc., 1998.
  7. Garden AS, el-Naggar AK, Morrison WH, et al.: Postoperative radiotherapy for malignant tumors of the parotid gland. Int J Radiat Oncol Biol Phys 37 (1): 79-85, 1997. [PUBMED Abstract]
  8. Chen AM, Granchi PJ, Garcia J, et al.: Local-regional recurrence after surgery without postoperative irradiation for carcinomas of the major salivary glands: implications for adjuvant therapy. Int J Radiat Oncol Biol Phys 67 (4): 982-7, 2007. [PUBMED Abstract]
  9. Buchholz TA, Laramore GE, Griffin BR, et al.: The role of fast neutron radiation therapy in the management of advanced salivary gland malignant neoplasms. Cancer 69 (11): 2779-88, 1992. [PUBMED Abstract]
  10. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]
  11. Douglas JG, Lee S, Laramore GE, et al.: Neutron radiotherapy for the treatment of locally advanced major salivary gland tumors. Head Neck 21 (3): 255-63, 1999. [PUBMED Abstract]
  12. Douglas JG, Laramore GE, Austin-Seymour M, et al.: Treatment of locally advanced adenoid cystic carcinoma of the head and neck with neutron radiotherapy. Int J Radiat Oncol Biol Phys 46 (3): 551-7, 2000. [PUBMED Abstract]
  13. Wang CC, Goodman M: Photon irradiation of unresectable carcinomas of salivary glands. Int J Radiat Oncol Biol Phys 21 (3): 569-76, 1991. [PUBMED Abstract]
  14. Douglas JG, Koh WJ, Austin-Seymour M, et al.: Treatment of salivary gland neoplasms with fast neutron radiotherapy. Arch Otolaryngol Head Neck Surg 129 (9): 944-8, 2003. [PUBMED Abstract]
  15. Kaplan MJ, Johns ME, Cantrell RW: Chemotherapy for salivary gland cancer. Otolaryngol Head Neck Surg 95 (2): 165-70, 1986. [PUBMED Abstract]
  16. Eisenberger MA: Supporting evidence for an active treatment program for advanced salivary gland carcinomas. Cancer Treat Rep 69 (3): 319-21, 1985. [PUBMED Abstract]
  17. Spiro RH: Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head Neck Surg 8 (3): 177-84, 1986 Jan-Feb. [PUBMED Abstract]
  18. Theriault C, Fitzpatrick PJ: Malignant parotid tumors. Prognostic factors and optimum treatment. Am J Clin Oncol 9 (6): 510-6, 1986. [PUBMED Abstract]
  19. Licitra L, Cavina R, Grandi C, et al.: Cisplatin, doxorubicin and cyclophosphamide in advanced salivary gland carcinoma. A phase II trial of 22 patients. Ann Oncol 7 (6): 640-2, 1996. [PUBMED Abstract]

Treatment of Stage I Major Salivary Gland Cancer

Treatment Options for Low-Grade Stage I Major Salivary Gland Tumors

Treatment options for low-grade stage I major salivary gland tumors include:

  1. Surgery alone.
  2. Postoperative radiation therapy should be considered when the resection margins are positive.

Low-grade stage I tumors of the salivary gland are curable with surgery alone.[13] Radiation therapy may be used for tumors for which resection involves a significant cosmetic or functional deficit or as an adjuvant to surgery when positive margins are present.[4] Neutron-beam therapy is effective in the treatment of patients with malignant salivary gland tumors who have a poor prognosis.[57]

Treatment Options for High-Grade Stage I Major Salivary Gland Tumors

Treatment options for high-grade stage I major salivary gland tumors include:

  1. Localized high-grade salivary gland tumors that are confined to the gland in which they arise may be cured by radical surgery alone.
  2. Postoperative radiation therapy may improve local control and increase survival rates for patients with high-grade tumors, positive surgical margins, or perineural invasion.[8][Level of evidence C2]; [911]
  3. Chemotherapy (under clinical evaluation).[12,13]

Clinical trials exploring newer methods of local control are appropriate.

High-grade stage I salivary gland tumors that are confined to the gland in which they arise may be cured by surgery alone. Adjuvant radiation therapy may be used, especially with the presence of positive margins.

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
  1. Byers RM, Jesse RH, Guillamondegui OM, et al.: Malignant tumors of the submaxillary gland. Am J Surg 126 (4): 458-63, 1973. [PUBMED Abstract]
  2. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  3. Woods JE, Chong GC, Beahrs OH: Experience with 1,360 primary parotid tumors. Am J Surg 130 (4): 460-2, 1975. [PUBMED Abstract]
  4. Guillamondegui OM, Byers RM, Luna MA, et al.: Aggressive surgery in treatment for parotid cancer: the role of adjunctive postoperative radiotherapy. Am J Roentgenol Radium Ther Nucl Med 123 (1): 49-54, 1975. [PUBMED Abstract]
  5. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]
  6. Douglas JG, Lee S, Laramore GE, et al.: Neutron radiotherapy for the treatment of locally advanced major salivary gland tumors. Head Neck 21 (3): 255-63, 1999. [PUBMED Abstract]
  7. Douglas JG, Laramore GE, Austin-Seymour M, et al.: Treatment of locally advanced adenoid cystic carcinoma of the head and neck with neutron radiotherapy. Int J Radiat Oncol Biol Phys 46 (3): 551-7, 2000. [PUBMED Abstract]
  8. Hosokawa Y, Shirato H, Kagei K, et al.: Role of radiotherapy for mucoepidermoid carcinoma of salivary gland. Oral Oncol 35 (1): 105-11, 1999. [PUBMED Abstract]
  9. Garden AS, el-Naggar AK, Morrison WH, et al.: Postoperative radiotherapy for malignant tumors of the parotid gland. Int J Radiat Oncol Biol Phys 37 (1): 79-85, 1997. [PUBMED Abstract]
  10. Mendenhall WM, Morris CG, Amdur RJ, et al.: Radiotherapy alone or combined with surgery for salivary gland carcinoma. Cancer 103 (12): 2544-50, 2005. [PUBMED Abstract]
  11. Chen AM, Granchi PJ, Garcia J, et al.: Local-regional recurrence after surgery without postoperative irradiation for carcinomas of the major salivary glands: implications for adjuvant therapy. Int J Radiat Oncol Biol Phys 67 (4): 982-7, 2007. [PUBMED Abstract]
  12. Kaplan MJ, Johns ME, Cantrell RW: Chemotherapy for salivary gland cancer. Otolaryngol Head Neck Surg 95 (2): 165-70, 1986. [PUBMED Abstract]
  13. Eisenberger MA: Supporting evidence for an active treatment program for advanced salivary gland carcinomas. Cancer Treat Rep 69 (3): 319-21, 1985. [PUBMED Abstract]

Treatment of Stage II Major Salivary Gland Cancer

Treatment Options for Low-Grade Stage II Major Salivary Gland Tumors

Treatment options for low-grade stage II major salivary gland tumors include:

  1. Surgery alone or with postoperative radiation therapy, if indicated, is appropriate.[1,2]
  2. Chemotherapy should be considered in special circumstances, such as when radiation therapy or surgery is refused.

Low-grade stage II tumors of the salivary gland may be cured with surgery alone.[35] Radiation therapy may be used as primary treatment for tumors for in which resection involves a significant cosmetic or functional deficit or as an adjuvant to surgery when positive margins are present.[6]

Treatment Options for High-Grade Stage II Major Salivary Gland Tumors

Treatment options for high-grade stage II major salivary gland tumors include:

  1. Localized high-grade salivary gland tumors that are confined to the gland in which they arise may be cured by radical surgery alone.
  2. Postoperative radiation therapy may improve local control and increase survival rates for patients with high-grade tumors, positive surgical margins, or perineural invasion.[7][Level of evidence C2]; [810]
  3. Fast neutron-beam radiation therapy or accelerated hyperfractionated photon-beam schedules reportedly are more effective than conventional x-ray therapy in the treatment of patients with inoperable, unresectable, or recurrent malignant salivary gland tumors.[1114]
  4. Chemotherapy (under clinical evaluation).[15,16]

Clinical trials exploring ways to improve local control with radiation therapy and/or radiosensitizers are appropriate.

High-grade stage II salivary gland tumors that are confined to the gland in which they arise may be cured by surgery alone, although adjuvant radiation therapy may be used, especially if positive margins are present. Primary radiation therapy may be given for tumors that are inoperable, unresectable, or recurrent. Fast neutron-beam radiation therapy has been shown to improve disease-free survival and overall survival in this clinical situation.[11,13,14]

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
  1. Spiro RH: Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head Neck Surg 8 (3): 177-84, 1986 Jan-Feb. [PUBMED Abstract]
  2. Theriault C, Fitzpatrick PJ: Malignant parotid tumors. Prognostic factors and optimum treatment. Am J Clin Oncol 9 (6): 510-6, 1986. [PUBMED Abstract]
  3. Byers RM, Jesse RH, Guillamondegui OM, et al.: Malignant tumors of the submaxillary gland. Am J Surg 126 (4): 458-63, 1973. [PUBMED Abstract]
  4. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  5. Woods JE, Chong GC, Beahrs OH: Experience with 1,360 primary parotid tumors. Am J Surg 130 (4): 460-2, 1975. [PUBMED Abstract]
  6. Guillamondegui OM, Byers RM, Luna MA, et al.: Aggressive surgery in treatment for parotid cancer: the role of adjunctive postoperative radiotherapy. Am J Roentgenol Radium Ther Nucl Med 123 (1): 49-54, 1975. [PUBMED Abstract]
  7. Hosokawa Y, Shirato H, Kagei K, et al.: Role of radiotherapy for mucoepidermoid carcinoma of salivary gland. Oral Oncol 35 (1): 105-11, 1999. [PUBMED Abstract]
  8. Garden AS, el-Naggar AK, Morrison WH, et al.: Postoperative radiotherapy for malignant tumors of the parotid gland. Int J Radiat Oncol Biol Phys 37 (1): 79-85, 1997. [PUBMED Abstract]
  9. Mendenhall WM, Morris CG, Amdur RJ, et al.: Radiotherapy alone or combined with surgery for salivary gland carcinoma. Cancer 103 (12): 2544-50, 2005. [PUBMED Abstract]
  10. Chen AM, Granchi PJ, Garcia J, et al.: Local-regional recurrence after surgery without postoperative irradiation for carcinomas of the major salivary glands: implications for adjuvant therapy. Int J Radiat Oncol Biol Phys 67 (4): 982-7, 2007. [PUBMED Abstract]
  11. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]
  12. Wang CC, Goodman M: Photon irradiation of unresectable carcinomas of salivary glands. Int J Radiat Oncol Biol Phys 21 (3): 569-76, 1991. [PUBMED Abstract]
  13. Douglas JG, Lee S, Laramore GE, et al.: Neutron radiotherapy for the treatment of locally advanced major salivary gland tumors. Head Neck 21 (3): 255-63, 1999. [PUBMED Abstract]
  14. Douglas JG, Laramore GE, Austin-Seymour M, et al.: Treatment of locally advanced adenoid cystic carcinoma of the head and neck with neutron radiotherapy. Int J Radiat Oncol Biol Phys 46 (3): 551-7, 2000. [PUBMED Abstract]
  15. Suen JY, Johns ME: Chemotherapy for salivary gland cancer. Laryngoscope 92 (3): 235-9, 1982. [PUBMED Abstract]
  16. Posner MR, Ervin TJ, Weichselbaum RR, et al.: Chemotherapy of advanced salivary gland neoplasms. Cancer 50 (11): 2261-4, 1982. [PUBMED Abstract]

Treatment of Stage III Major Salivary Gland Cancer

Treatment Options for Low-Grade Stage III Major Salivary Gland Tumors

Treatment options for low-grade stage III major salivary gland tumors include:

  1. Surgery alone or with postoperative radiation therapy, if indicated, is appropriate.
  2. Chemotherapy should be considered in special circumstances, such as when radiation or surgery is refused or when tumors are recurrent or nonresponsive.
  3. Fast neutron-beam radiation therapy (under clinical evaluation). Data on fast neutron-beam radiation therapy have indicated superior results when compared with conventional radiation therapy using x-rays.[1,2]
  4. Chemotherapy (under clinical evaluation).[3,4]

Patients with low-grade stage III tumors of the salivary gland may be cured with surgery alone.[57] Radiation therapy as primary treatment is not often required but may be used for tumors for which resection involves a significant cosmetic or functional deficit, or as an adjuvant to surgery when positive margins are present.[8] Patients with low-grade tumors that have spread to lymph nodes may be cured with resection of the primary tumor and the involved lymph nodes, with or without radiation therapy. Neutron-beam therapy is effective in the treatment of patients with tumors that have spread to local lymph nodes.

Treatment Options for High-Grade Stage III Major Salivary Gland Tumors

Treatment options for high-grade stage III major salivary gland tumors include:

  1. Patients with localized high-grade salivary gland tumors that are confined to the gland in which they arise may be cured by radical surgery alone.[9,10]
  2. Postoperative radiation therapy may improve local control and increase survival rates for patients with high-grade tumors, positive surgical margins, or perineural invasion.[11][Level of evidence C2]; [1214]
  3. Fast neutron-beam radiation therapy or accelerated hyperfractionated photon-beam schedules have been reported to be more effective than conventional x-ray therapy in the treatment of patients with inoperable, unresectable, or recurrent malignant salivary gland tumors.[1,1517]
  4. Radiation therapy, radiosensitizers, and chemotherapy (under clinical evaluation). Clinical trials are exploring ways to improve local control with these modalities.[24,18,19]

Patients with high-grade stage III salivary gland tumors that are confined to the gland in which they arise may be cured by surgery alone, although adjuvant postoperative radiation therapy may be used, especially if positive margins are present. Primary conventional x-ray radiation therapy may provide palliation for patients with unresectable tumors. Fast neutron beams, however, have been reported to improve disease-free survival and overall survival in this clinical situation.[1,16,17] Patients with tumors that have spread to regional lymph nodes should have a regional lymphadenectomy as part of the initial surgical procedure. Adjuvant radiation therapy for these tumors may reduce the local recurrence rate.

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
  1. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]
  2. Catterall M, Errington RD: The implications of improved treatment of malignant salivary gland tumors by fast neutron radiotherapy. Int J Radiat Oncol Biol Phys 13 (9): 1313-8, 1987. [PUBMED Abstract]
  3. Kaplan MJ, Johns ME, Cantrell RW: Chemotherapy for salivary gland cancer. Otolaryngol Head Neck Surg 95 (2): 165-70, 1986. [PUBMED Abstract]
  4. Eisenberger MA: Supporting evidence for an active treatment program for advanced salivary gland carcinomas. Cancer Treat Rep 69 (3): 319-21, 1985. [PUBMED Abstract]
  5. Byers RM, Jesse RH, Guillamondegui OM, et al.: Malignant tumors of the submaxillary gland. Am J Surg 126 (4): 458-63, 1973. [PUBMED Abstract]
  6. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  7. Woods JE, Chong GC, Beahrs OH: Experience with 1,360 primary parotid tumors. Am J Surg 130 (4): 460-2, 1975. [PUBMED Abstract]
  8. Guillamondegui OM, Byers RM, Luna MA, et al.: Aggressive surgery in treatment for parotid cancer: the role of adjunctive postoperative radiotherapy. Am J Roentgenol Radium Ther Nucl Med 123 (1): 49-54, 1975. [PUBMED Abstract]
  9. Spiro RH: Salivary neoplasms: overview of a 35-year experience with 2,807 patients. Head Neck Surg 8 (3): 177-84, 1986 Jan-Feb. [PUBMED Abstract]
  10. Theriault C, Fitzpatrick PJ: Malignant parotid tumors. Prognostic factors and optimum treatment. Am J Clin Oncol 9 (6): 510-6, 1986. [PUBMED Abstract]
  11. Hosokawa Y, Shirato H, Kagei K, et al.: Role of radiotherapy for mucoepidermoid carcinoma of salivary gland. Oral Oncol 35 (1): 105-11, 1999. [PUBMED Abstract]
  12. Garden AS, el-Naggar AK, Morrison WH, et al.: Postoperative radiotherapy for malignant tumors of the parotid gland. Int J Radiat Oncol Biol Phys 37 (1): 79-85, 1997. [PUBMED Abstract]
  13. Mendenhall WM, Morris CG, Amdur RJ, et al.: Radiotherapy alone or combined with surgery for salivary gland carcinoma. Cancer 103 (12): 2544-50, 2005. [PUBMED Abstract]
  14. Chen AM, Granchi PJ, Garcia J, et al.: Local-regional recurrence after surgery without postoperative irradiation for carcinomas of the major salivary glands: implications for adjuvant therapy. Int J Radiat Oncol Biol Phys 67 (4): 982-7, 2007. [PUBMED Abstract]
  15. Wang CC, Goodman M: Photon irradiation of unresectable carcinomas of salivary glands. Int J Radiat Oncol Biol Phys 21 (3): 569-76, 1991. [PUBMED Abstract]
  16. Douglas JG, Lee S, Laramore GE, et al.: Neutron radiotherapy for the treatment of locally advanced major salivary gland tumors. Head Neck 21 (3): 255-63, 1999. [PUBMED Abstract]
  17. Douglas JG, Laramore GE, Austin-Seymour M, et al.: Treatment of locally advanced adenoid cystic carcinoma of the head and neck with neutron radiotherapy. Int J Radiat Oncol Biol Phys 46 (3): 551-7, 2000. [PUBMED Abstract]
  18. Suen JY, Johns ME: Chemotherapy for salivary gland cancer. Laryngoscope 92 (3): 235-9, 1982. [PUBMED Abstract]
  19. Posner MR, Ervin TJ, Weichselbaum RR, et al.: Chemotherapy of advanced salivary gland neoplasms. Cancer 50 (11): 2261-4, 1982. [PUBMED Abstract]

Treatment of Stage IV Major Salivary Gland Cancer

Treatment Options for Stage IV Major Salivary Gland Tumors

Standard therapy for patients with tumors that have spread to distant sites is not curative.

Treatment options for high-grade stage IV major salivary gland tumors include:

  1. Fast neutron-beam radiation therapy or accelerated hyperfractionated photon-beam schedules have been reported to be more effective than conventional x-ray therapy in the treatment of patients with inoperable, unresectable, or recurrent malignant salivary gland tumors.[15]
  2. Aggressive combinations of chemotherapy and radiation (under clinical evaluation).

Patients with stage IV salivary gland cancer and patients with any metastatic lesions should consider enrollment in clinical trials. Their cancer may be responsive to aggressive combinations of chemotherapy and radiation. Patients with any metastatic lesions could consider clinical trials. Chemotherapy using doxorubicin, cisplatin, cyclophosphamide, and fluorouracil as single agents or in various combinations is associated with modest response rates.[614]

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.[15,16] 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.[1517] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[1820] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[21] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[22]

References
  1. Wang CC, Goodman M: Photon irradiation of unresectable carcinomas of salivary glands. Int J Radiat Oncol Biol Phys 21 (3): 569-76, 1991. [PUBMED Abstract]
  2. Laramore GE, Krall JM, Griffin TW, et al.: Neutron versus photon irradiation for unresectable salivary gland tumors: final report of an RTOG-MRC randomized clinical trial. Radiation Therapy Oncology Group. Medical Research Council. Int J Radiat Oncol Biol Phys 27 (2): 235-40, 1993. [PUBMED Abstract]
  3. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]
  4. Douglas JG, Lee S, Laramore GE, et al.: Neutron radiotherapy for the treatment of locally advanced major salivary gland tumors. Head Neck 21 (3): 255-63, 1999. [PUBMED Abstract]
  5. Douglas JG, Laramore GE, Austin-Seymour M, et al.: Treatment of locally advanced adenoid cystic carcinoma of the head and neck with neutron radiotherapy. Int J Radiat Oncol Biol Phys 46 (3): 551-7, 2000. [PUBMED Abstract]
  6. Eisenberger MA: Supporting evidence for an active treatment program for advanced salivary gland carcinomas. Cancer Treat Rep 69 (3): 319-21, 1985. [PUBMED Abstract]
  7. Venook AP, Tseng A, Meyers FJ, et al.: Cisplatin, doxorubicin, and 5-fluorouracil chemotherapy for salivary gland malignancies: a pilot study of the Northern California Oncology Group. J Clin Oncol 5 (6): 951-5, 1987. [PUBMED Abstract]
  8. Rentschler R, Burgess MA, Byers R: Chemotherapy of malignant major salivary gland neoplasms: a 25-year review of M. D. Anderson Hospital experience. Cancer 40 (2): 619-24, 1977. [PUBMED Abstract]
  9. Posner MR, Ervin TJ, Weichselbaum RR, et al.: Chemotherapy of advanced salivary gland neoplasms. Cancer 50 (11): 2261-4, 1982. [PUBMED Abstract]
  10. Suen JY, Johns ME: Chemotherapy for salivary gland cancer. Laryngoscope 92 (3): 235-9, 1982. [PUBMED Abstract]
  11. Catterall M, Errington RD: The implications of improved treatment of malignant salivary gland tumors by fast neutron radiotherapy. Int J Radiat Oncol Biol Phys 13 (9): 1313-8, 1987. [PUBMED Abstract]
  12. Ono M, Watanabe A, Matsumoto Y, et al.: Methamphetamine modifies the photic entraining responses in the rodent suprachiasmatic nucleus via serotonin release. Neuroscience 72 (1): 213-24, 1996. [PUBMED Abstract]
  13. Saroja KR, Mansell J, Hendrickson FR, et al.: An update on malignant salivary gland tumors treated with neutrons at Fermilab. Int J Radiat Oncol Biol Phys 13 (9): 1319-25, 1987. [PUBMED Abstract]
  14. Licitra L, Cavina R, Grandi C, et al.: Cisplatin, doxorubicin and cyclophosphamide in advanced salivary gland carcinoma. A phase II trial of 22 patients. Ann Oncol 7 (6): 640-2, 1996. [PUBMED Abstract]
  15. 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]
  16. Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
  17. 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]
  18. 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]
  19. 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]
  20. 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]
  21. 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]
  22. 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 Recurrent Major Salivary Gland Cancer

The prognosis for any patient with progressing or relapsing salivary gland cancer is poor, regardless of cell type or stage. Selecting further treatment depends on many factors, including the specific cancer, prior treatment, site of recurrence, and individual patient considerations. Fast neutron-beam radiation therapy is superior to conventional radiation therapy using x-rays and may be curative in selected patients with recurrent disease.[1]

Patients with inoperable, unresectable, or recurrent malignant salivary gland tumors treated with fast neutron-beam radiation therapy have better disease-free survival and overall survival than patients treated with conventional x-ray radiation therapy.[25] Clinical trials are appropriate and should be considered when possible.

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
  1. Laramore GE, Krall JM, Griffin TW, et al.: Neutron versus photon irradiation for unresectable salivary gland tumors: final report of an RTOG-MRC randomized clinical trial. Radiation Therapy Oncology Group. Medical Research Council. Int J Radiat Oncol Biol Phys 27 (2): 235-40, 1993. [PUBMED Abstract]
  2. Laramore GE: Fast neutron radiotherapy for inoperable salivary gland tumors: is it the treatment of choice? Int J Radiat Oncol Biol Phys 13 (9): 1421-3, 1987. [PUBMED Abstract]
  3. Saroja KR, Mansell J, Hendrickson FR, et al.: An update on malignant salivary gland tumors treated with neutrons at Fermilab. Int J Radiat Oncol Biol Phys 13 (9): 1319-25, 1987. [PUBMED Abstract]
  4. Buchholz TA, Laramore GE, Griffin BR, et al.: The role of fast neutron radiation therapy in the management of advanced salivary gland malignant neoplasms. Cancer 69 (11): 2779-88, 1992. [PUBMED Abstract]
  5. Krüll A, Schwarz R, Engenhart R, et al.: European results in neutron therapy of malignant salivary gland tumors. Bull Cancer Radiother 83 (Suppl): 125-9s, 1996. [PUBMED Abstract]

Latest Updates to This Summary (05/13/2025)

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.

General Information About Salivary Gland Cancer

Revised text about the incidence and mortality rates of salivary gland cancer in the United States (cited National Cancer Institute 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 adult salivary gland 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 Salivary Gland Cancer Treatment are:

  • Andrea Bonetti, MD (Pederzoli Hospital)
  • 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 Salivary Gland Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/hp/adult/salivary-gland-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389389]

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.

Oral Cavity and Nasopharyngeal Cancers Screening (PDQ®)–Health Professional Version

Oral Cavity and Nasopharyngeal Cancers Screening (PDQ®)–Health Professional Version

Overview

Oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers may be referred to as head and neck squamous cell cancers. Oral cavity squamous cell cancers most commonly arise from the mucosal surfaces lining the oral cavity. Pharyngeal squamous cell cancers can be categorized into nasopharyngeal, oropharyngeal, and hypopharyngeal cancers on the basis of anatomical landmarks. Figure 1 shows the anatomy of the pharynx.

EnlargeAnatomy of the pharynx; drawing shows the nasopharynx, oropharynx, and hypopharynx. Also shown are the nasal cavity, oral cavity, hyoid bone, larynx, esophagus, and trachea.
Figure 1. Anatomy of the pharynx.

Note: The Overview section summarizes the published evidence on this topic. The rest of the summary describes the evidence in more detail.

Other PDQ summaries on Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers Prevention and Lip and Oral Cavity Cancer Treatment are also available.

Benefits

There is inadequate evidence to establish whether screening would result in a decrease in mortality from oral cavity and nasopharyngeal cancers.

Magnitude of Effect: No evidence of benefit, and harms have not been quantified.

  • Study Design: Evidence obtained from one randomized controlled trial and observational studies.
  • Internal Validity: Poor.
  • Consistency: Not applicable (N/A).
  • External Validity: Poor.

Harms

Harms, although unavoidable, have not been quantified on the basis of the literature. However, there are some unavoidable harms that would be associated with routine screening, including:

  • Unnecessary treatment associated with overdiagnosis.
  • Psychological consequences of false-positive tests.
  • Misdiagnosis because of variability in assessment of biopsies.

Magnitude of Effect: Unknown.

  • Study Design: Observational studies.
  • Internal Validity: Poor.
  • Consistency: N/A.
  • External Validity: Poor.

Incidence and Mortality

An estimated 59,660 new cases of oral cavity and oropharynx cancers will be diagnosed in the United States in 2025, and an estimated 12,770 people will die of these diseases.[1] The overall annual incidence in the United States is about 11 cases per 100,000 men and women; the incidence rate is highest in individuals aged 75 to 84 years.[2]

Between 2012 and 2021, incidence rates increased by 0.7% per year.[1] The incidence has been increasing for oral cavity and oropharyngeal cancers related to human papillomavirus (HPV) infection. About 60% of oral/pharyngeal cancers are moderately advanced (regional stage) or metastatic at the time of diagnosis.[2] The 5-year relative survival rate is 69%.[1]

The estimated annual worldwide number of cases of oral cavity and oropharyngeal cancers is about 275,000, with an approximate 20-fold variation geographically.[3] South and Southeast Asia (India, Sri Lanka, Pakistan, and Bangladesh), France, and Brazil have particularly high rates. In most countries, men have higher rates of oral cavity cancer than women (caused by tobacco use) and higher rates of lip cancer (caused by sunlight exposure from outdoor occupations).[3]

Nasopharyngeal cancers are rare in the United States, with an annual incidence rate of 0.7 cases per 100,000 persons.[4] However, there are marked geographic differences, with an overall incidence in China that is 40- to 380-fold higher than that in the United States.[4] There are elevated rates of nasopharyngeal cancers in the Cantonese population of southern China (including Hong Kong), and intermediate rates are observed in several indigenous populations in Southeast Asia and in natives of the Arctic region, North Africa, and the Middle East. First-generation Chinese immigrants to the United States maintain a high incidence rate, while their descendants born in the United States show a decreased incidence. The 5-year survival rate for keratinizing squamous cell carcinoma, the most common subtype of nasopharyngeal cancer in the United States, is 46%.[5]

References
  1. American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
  2. 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.
  3. Warnakulasuriya S: Global epidemiology of oral and oropharyngeal cancer. Oral Oncol 45 (4-5): 309-16, 2009 Apr-May. [PUBMED Abstract]
  4. Richey LM, Olshan AF, George J, et al.: Incidence and survival rates for young blacks with nasopharyngeal carcinoma in the United States. Arch Otolaryngol Head Neck Surg 132 (10): 1035-40, 2006. [PUBMED Abstract]
  5. Ou SH, Zell JA, Ziogas A, et al.: Epidemiology of nasopharyngeal carcinoma in the United States: improved survival of Chinese patients within the keratinizing squamous cell carcinoma histology. Ann Oncol 18 (1): 29-35, 2007. [PUBMED Abstract]

Risk Factors

The primary risk factors for oral cavity cancers in American men and women are tobacco (including smokeless tobacco) use, alcohol use, betel-quid chewing, and human papillomavirus infection (HPV).

Risk factors for nasopharyngeal cancer include Epstein-Barr virus (EBV) persistent infection.[1]

For more information about factors associated with an increased or decreased risk of oral cavity squamous cell cancers, see Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers Prevention and Lip and Oral Cavity Cancer Treatment.

Epstein-Barr Virus (EBV) Infection

Based on solid evidence, EBV infection causes nasopharyngeal cancer in high-incidence areas.[1] Collective evidence includes numerous case-control studies and cohort studies that show a higher proportion of patients with nasopharyngeal cancer who have anti-EBV antibodies than controls and that seropositive status precedes tumor diagnosis.[2,3] Recent studies have also found circulating cell-free EBV DNA in patients with nasopharyngeal cancer but not in controls.[4] EBV alone is not a sufficient cause because 90% of adults worldwide are infected with the virus, but only a small proportion develop nasopharyngeal cancer.[5] EBV infection is subclinical and occurs early in childhood. The pathogenesis is thought to involve the virus establishing latency in epithelial cells that have already undergone premalignant genetic changes.

One of the first studies to show an association was a cohort study that found higher anti-EBV titers in 84% of the 235 East African and Chinese patients with nasopharyngeal carcinoma.[2,5] The same study found higher anti-EBV titers with higher-stage tumor, and a case-control component of the study revealed that high anti-EBV titers were six times more likely in patients with nasopharyngeal carcinoma than in patients with head and neck cancers at other sites.

Other studies show elevation in both IgG and IgA antibody titers to EBV viral capsid antigen and other latent viral antigens, which precede tumor development by several years and are correlated with tumor burden, remission, and recurrence.[2,3] A large cohort study with 9,699 men measured both IgA antibodies against EBV capsid antigen and neutralizing antibodies against EBV-specific DNase and followed them for a later diagnosis of nasopharyngeal cancer.[3] The relative risk of nasopharyngeal carcinoma was 32.8 for individuals with both antibody markers (95% confidence interval [CI], 7.3–147.2; P < .001), and 4.0 for individuals with one marker (95% CI, 1.6–10.2; P = .003), compared with individuals with neither marker. There was a temporal relationship in that the difference in cumulative incidence between seropositive and seronegative patients increased with a longer duration of follow-up. Another study found circulating cell-free EBV DNA in 95% of patients with advanced nasopharyngeal cancer but not in controls or cured patients.[4]

References
  1. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Biological agents. Volume 100 B. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum 100 (Pt B): 1-441, 2012. [PUBMED Abstract]
  2. Henle W, Henle G, Ho HC, et al.: Antibodies to Epstein-Barr virus in nasopharyngeal carcinoma, other head and neck neoplasms, and control groups. J Natl Cancer Inst 44 (1): 225-31, 1970. [PUBMED Abstract]
  3. Chien YC, Chen JY, Liu MY, et al.: Serologic markers of Epstein-Barr virus infection and nasopharyngeal carcinoma in Taiwanese men. N Engl J Med 345 (26): 1877-82, 2001. [PUBMED Abstract]
  4. Lin JC, Wang WY, Chen KY, et al.: Quantification of plasma Epstein-Barr virus DNA in patients with advanced nasopharyngeal carcinoma. N Engl J Med 350 (24): 2461-70, 2004. [PUBMED Abstract]
  5. Chang ET, Adami HO: The enigmatic epidemiology of nasopharyngeal carcinoma. Cancer Epidemiol Biomarkers Prev 15 (10): 1765-77, 2006. [PUBMED Abstract]

Evidence of Benefit Associated With Screening

No population-based screening programs for oral cavity squamous cell cancers have been implemented in developed countries, although opportunistic screening or screening as part of a periodic health examination has been advocated for the oral cavity, which is the only site accessible without endoscopy.[1,2]

Screening for Oral Cavity Cancers

There are different methods of screening for oral cavity cancers. Oral cavity cancers occur in a region of the body that is generally accessible to physical examination by the patient, the dentist, and the physician. Visual examination is the most common method used to detect visible lesions. Other methods have been used to augment clinical detection of oral lesions and include toluidine blue, brush biopsy, and fluorescence staining.

An inspection of the oral cavity is often part of a physical examination in a dentist’s or physician’s office. Of note, high-risk individuals visit their medical doctors more frequently than they visit their dentists. Although physicians are more likely to provide risk-factor counseling (such as tobacco cessation), they are less likely than dentists to perform an oral cancer examination.[3] Overall, only a fraction (~20%) of Americans receive an oral cancer examination. Black patients, Hispanic patients, and those who have a lower level of education are less likely to have such an examination, perhaps because they lack access to medical care.[3] An oral examination often includes looking for leukoplakia and erythroplakia lesions, which can progress to cancer.[4,5] One study has shown that direct fluorescence visualization (using a simple hand-held device in the operating room) could identify subclinical high-risk fields with cancerous or precancerous changes extending up to 25 mm beyond the primary tumor in 19 of 20 patients undergoing oral surgery for invasive or in situ squamous cell tumors.[6] However, this finding has not yet been tested in a screening setting. Data suggest that molecular markers may be useful in the prognosis of these premalignant oral lesions.[7]

The routine examination of asymptomatic and symptomatic patients can lead to detection of earlier-stage cancers and premalignant lesions. There is no definitive evidence, however, to show that this screening can reduce oral cancer mortality, and there are no randomized controlled trials (RCTs) in any Western or other low-risk populations.[5,811]

In a single RCT of screening versus usual care, 13 geographic clusters in the Trivandrum district of Kerala, India, were randomly assigned to receive systematic oral visual screening by trained health workers (seven screened clusters, six control clusters) every 3 years for four screening rounds between 1996 and 2008. During a 15-year follow-up period, there were 138 deaths from oral cancer in the screening group, with a cause-specific mortality rate of 15.4 per 100,000 person-years, and 154 deaths in the control group, with a mortality rate of 17.1 per 100,000 person-years (relative risk [RR], 0.88; 95% confidence interval [CI], 0.69–1.12). In a subset analysis restricted to tobacco or alcohol users, the mortality rates were 30 and 39 per 100,000 person-years, respectively (RR, 0.76; 95% CI, 0.60–0.97). There was no apparent adjustment of the CIs for the cluster design. In another subgroup analysis, mortality hazard ratios were calculated for groups defined by number of times screened, but the inappropriate comparison in each case was to the control group of the whole study. No data on treatment of oral cancers were presented.[1215]

Aside from the issues of generalizability to other populations and lack of an overall statistically significant result in cause-specific mortality, interpretation of the results is made difficult by serious lacks in methodological detail about the randomization process, allocation concealment, adjustment for clustering effect, and information about treatment. The total number of clusters randomized was small, and there were different distributions of income and household possessions between the two study arms. Withdrawals and dropouts were not clearly described. In summary, the sole randomized trial does not provide solid evidence of a cause-specific mortality benefit associated with systematic oral cavity visual examination.

Techniques such as toluidine blue staining, brush biopsy/cytology, or fluorescence imaging as the primary screening tool or as an adjunct for screening have not been shown to have superior sensitivity and specificity for visual examination alone or to yield better health outcomes.[5,16] In an RCT conducted in Keelung County, Taiwan, 7,975 individuals at high risk of oral cancer due to cigarette smoking or betel-quid chewing were randomly assigned to receive a one-time oral cancer examination after gargling with toluidine blue or a blue placebo dye.[17] The positive test rates were 9.5% versus 8.3%, respectively (P = .047). The detection of premalignant lesions was not statistically different (rate ratio, 1.05; 95% CI, 0.74–1.41). The number of overall oral cancers diagnosed within the short follow-up period of 5 years was too small for valid comparison (six in each group).

The operating characteristics of the various techniques used as an adjunct to oral visual examination are not well established. A systematic literature review of toluidine blue, a variety of other visualization adjuncts, and cytopathology in the screening setting revealed a very broad range of reported sensitivities, specificities, and positive predictive values when biopsy confirmation was used as the gold standard outcome.[18] In part, this range of findings can be attributed to varying study populations, sample size and settings, and criteria for positive-clinical examinations and for scoring a biopsy as positive.

Screening for Nasopharyngeal Cancer

Serum Epstein-Barr virus (EBV)–associated antibodies and circulating cell-free EBV DNA testing have been used for nasopharyngeal cancer diagnosis and screening. In an observational study of 20,349 men aged 40 to 62 years, circulating cell-free EBV DNA testing was used to screen for nasopharyngeal cancer.[19,20] A total of 1.5% of participants tested positive twice for EBV DNA and had further workup, leading to a diagnosis of nasopharyngeal cancer for 34 patients. The EBV DNA test had a sensitivity of 97.1% (95% CI, 95.5%–98.7%) and specificity of 98.6% (95% CI, 98.6%–98.7%). Without a control group, the study compared stage of disease at diagnosis with a historical cohort and found a higher proportion of stage I and stage II disease (71% vs. 20%; P < .001) and superior 3-year progression-free survival in the screen-detected population. However, the survival benefit in the study may also be caused by lead-time bias.

Other screening programs in southern China use EBV-associated antibodies, but their effects are difficult to determine because of lack of controls for comparison of survival outcomes.[2022] In summary, current screening studies for nasopharyngeal cancer do not provide solid evidence of a benefit associated with screening for nasopharyngeal cancer, especially in nonendemic regions such as the United States.

References
  1. Opportunistic oral cancer screening: a management strategy for dental practice. BDA Occasional Paper 6: 1-36, 2000. Also available online. Last accessed April 14, 2025.
  2. Smith RA, Cokkinides V, Brooks D, et al.: Cancer screening in the United States, 2011: A review of current American Cancer Society guidelines and issues in cancer screening. CA Cancer J Clin 61 (1): 8-30, 2011 Jan-Feb. [PUBMED Abstract]
  3. Kerr AR, Changrani JG, Gany FM, et al.: An academic dental center grapples with oral cancer disparities: current collaboration and future opportunities. J Dent Educ 68 (5): 531-41, 2004. [PUBMED Abstract]
  4. Warnakulasuriya S, Johnson NW, van der Waal I: Nomenclature and classification of potentially malignant disorders of the oral mucosa. J Oral Pathol Med 36 (10): 575-80, 2007. [PUBMED Abstract]
  5. Brocklehurst P, Kujan O, Glenny AM, et al.: Screening programmes for the early detection and prevention of oral cancer. Cochrane Database Syst Rev (11): CD004150, 2010. [PUBMED Abstract]
  6. Poh CF, Zhang L, Anderson DW, et al.: Fluorescence visualization detection of field alterations in tumor margins of oral cancer patients. Clin Cancer Res 12 (22): 6716-22, 2006. [PUBMED Abstract]
  7. Poh CF, Zhang L, Lam WL, et al.: A high frequency of allelic loss in oral verrucous lesions may explain malignant risk. Lab Invest 81 (4): 629-34, 2001. [PUBMED Abstract]
  8. Screening for oral cancer. In: Fisher M, Eckhart C, eds.: Guide to Clinical Preventive Services: an Assessment of the Effectiveness of 169 Interventions. Report of the U.S. Preventive Services Task Force. Williams & Wilkins, 1989, pp 91-94.
  9. Antunes JL, Biazevic MG, de Araujo ME, et al.: Trends and spatial distribution of oral cancer mortality in São Paulo, Brazil, 1980-1998. Oral Oncol 37 (4): 345-50, 2001. [PUBMED Abstract]
  10. U.S. Preventive Services Task Force: Screening for Oral Cancer: Recommendation Statement. Rockville, Md: U.S. Preventive Services Task Force, 2013. Available online. Last accessed April 14, 2025.
  11. Scattoloni J: Screening for Oral Cancer: Brief Evidence Update. Rockville, Md: U.S. Preventive Services Task Force, 2004. Available online. Last accessed April 14, 2025.
  12. Sankaranarayanan R, Mathew B, Jacob BJ, et al.: Early findings from a community-based, cluster-randomized, controlled oral cancer screening trial in Kerala, India. The Trivandrum Oral Cancer Screening Study Group. Cancer 88 (3): 664-73, 2000. [PUBMED Abstract]
  13. Ramadas K, Sankaranarayanan R, Jacob BJ, et al.: Interim results from a cluster randomized controlled oral cancer screening trial in Kerala, India. Oral Oncol 39 (6): 580-8, 2003. [PUBMED Abstract]
  14. Sankaranarayanan R, Ramadas K, Thomas G, et al.: Effect of screening on oral cancer mortality in Kerala, India: a cluster-randomised controlled trial. Lancet 365 (9475): 1927-33, 2005 Jun 4-10. [PUBMED Abstract]
  15. Sankaranarayanan R, Ramadas K, Thara S, et al.: Long term effect of visual screening on oral cancer incidence and mortality in a randomized trial in Kerala, India. Oral Oncol 49 (4): 314-21, 2013. [PUBMED Abstract]
  16. Lingen MW, Kalmar JR, Karrison T, et al.: Critical evaluation of diagnostic aids for the detection of oral cancer. Oral Oncol 44 (1): 10-22, 2008. [PUBMED Abstract]
  17. Su WW, Yen AM, Chiu SY, et al.: A community-based RCT for oral cancer screening with toluidine blue. J Dent Res 89 (9): 933-7, 2010. [PUBMED Abstract]
  18. Patton LL, Epstein JB, Kerr AR: Adjunctive techniques for oral cancer examination and lesion diagnosis: a systematic review of the literature. J Am Dent Assoc 139 (7): 896-905; quiz 993-4, 2008. [PUBMED Abstract]
  19. Chan KCA, Woo JKS, King A, et al.: Analysis of Plasma Epstein-Barr Virus DNA to Screen for Nasopharyngeal Cancer. N Engl J Med 377 (6): 513-522, 2017. [PUBMED Abstract]
  20. Cao SM, Simons MJ, Qian CN: The prevalence and prevention of nasopharyngeal carcinoma in China. Chin J Cancer 30 (2): 114-9, 2011. [PUBMED Abstract]
  21. Zeng Y, Zhang LG, Li HY, et al.: Serological mass survey for early detection of nasopharyngeal carcinoma in Wuzhou City, China. Int J Cancer 29 (2): 139-41, 1982. [PUBMED Abstract]
  22. Zeng Y, Zhong JM, Li LY, et al.: Follow-up studies on Epstein-Barr virus IgA/VCA antibody-positive persons in Zangwu County, China. Intervirology 20 (4): 190-4, 1983. [PUBMED Abstract]

Evidence of Harm Associated With Screening

Harms associated with screening for oral cavity squamous cell cancers are poorly studied in any quantifiable way.[1] However, there are some unavoidable harms that would be associated with routine screening, including:

  • Unnecessary treatment of lesions that would not have progressed (overdiagnosis).
  • Psychological consequences of false-positive tests.[2]

An additional potential harm is misdiagnosis and resulting under- or overtreatment, given the subjective pathology judgments in the reading of biopsies of oral lesions. When 87 biopsy diagnoses of oral lesions were compared between 21 local pathologists and double-reading by two of three central pathologists in a multicenter study of patients with prior upper aerodigestive tract cancers, agreement was only fair to good (kappa-weighted statistic, 0.59; 95% confidence interval [CI], 0.45–0.72).[3] In a bivariate categorization of carcinoma in situ plus carcinoma versus less serious lesions, the agreement was poor, but with very wide CIs (kappa statistic, 0.39; 95% CI, -0.12 to -0.97). The investigators in the same study analyzed an agreement between the local and central pathologists on clinically normal tissue adjacent to 67 biopsied, clinically suspicious lesions. The agreement on clinically normal tissue was better than for visibly abnormal lesions, but still not in the excellent range (kappa-weighted statistic, 0.75; 95% CI, 0.64–0.86).[4]

References
  1. Scattoloni J: Screening for Oral Cancer: Brief Evidence Update. Rockville, Md: U.S. Preventive Services Task Force, 2004. Available online. Last accessed April 14, 2025.
  2. Speight PM, Zakrzewska J, Downer MC: Screening for oral cancer and precancer. Eur J Cancer B Oral Oncol 28B (1): 45-8, 1992. [PUBMED Abstract]
  3. Fischer DJ, Epstein JB, Morton TH, et al.: Interobserver reliability in the histopathologic diagnosis of oral pre-malignant and malignant lesions. J Oral Pathol Med 33 (2): 65-70, 2004. [PUBMED Abstract]
  4. Fischer DJ, Epstein JB, Morton TH, et al.: Reliability of histologic diagnosis of clinically normal intraoral tissue adjacent to clinically suspicious lesions in former upper aerodigestive tract cancer patients. Oral Oncol 41 (5): 489-96, 2005. [PUBMED Abstract]

Latest Updates to This Summary (04/14/2025)

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.

Incidence and Mortality

Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 1).

Revised text to state that between 2012 and 2021, incidence rates increased by 0.7% per year.

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 oral cavity and nasopharyngeal cancers screening. 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 Oral Cavity and Nasopharyngeal Cancers Screening. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/hp/oral-screening-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389219]

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.

Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers Prevention (PDQ®)–Health Professional Version

Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers Prevention (PDQ®)–Health Professional Version

Overview

Note: The Overview section summarizes the published evidence on this topic. The rest of the summary describes the evidence in more detail.

Oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers may be referred to as head and neck squamous cell cancers. Head and neck squamous cell cancers most commonly arise from the mucosal surfaces lining the oral cavity, oropharynx, hypopharynx, and larynx. Anatomically, the pharynx includes the nasopharynx, oropharynx, and hypopharynx, but cancers in these sites have distinct clinical and epidemiologic characteristics. Thus, it is inappropriate to group them together.[1] For more information, see the following PDQ summaries:

Figure 1 shows the anatomy of the pharynx.

EnlargeAnatomy of the pharynx; drawing shows the nasopharynx, oropharynx, and hypopharynx. Also shown are the nasal cavity, oral cavity, hyoid bone, larynx, esophagus, and trachea.
Figure 1. Anatomy of the pharynx.

Who Is at Risk?

Head and neck squamous cell cancers have common risk factors. People who use tobacco in any of the commonly available forms (cigarettes, cigars, pipes, and smokeless tobacco) or have a high alcohol intake are at elevated risk of oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers; they are at particularly high risk if they use both tobacco and alcohol.[2] Individuals who chew betel quid (whether mixed with tobacco or not) are also at high risk of cancer of the oral cavity and oropharynx.[3,4] Individuals who have a personal history of cancer of the head and neck region also are at elevated risk of a future primary cancer of the head and neck.[5] Human papillomavirus type 16 (HPV-16) is a sufficient, but not necessary, cause of oral, tongue, and oropharyngeal cancers.[2,6]

Note: Separate PDQ summaries on Oral Cavity and Nasopharyngeal Cancers Screening and Cigarette Smoking: Health Risks and How to Quit are also available.

Factors With Adequate Evidence of an Increased Risk of Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers

Tobacco use

Based on solid evidence from numerous observational studies, tobacco use increases the risk of cancers of the oral cavity, oropharynx, hypopharynx, and larynx.[79]

Magnitude of Effect: Large. Risk for current smokers ranges from fourfold to fivefold for oral cavity, oropharyngeal, and hypopharyngeal cancers to tenfold for laryngeal cancer compared with never-smokers, and is dose related. Most cancers of the oral cavity, oropharynx, hypopharynx, and larynx are attributable to the use of tobacco products.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Alcohol use

Based on solid evidence, alcohol use is a risk factor for the development of head and neck cancers. Its effects are independent of those of tobacco use.[912]

Magnitude of Effect: Lower than the risk associated with tobacco use, but the risk is approximately twofold to sixfold for people who drink two or more alcoholic beverages per day compared with nondrinkers, and is dose related.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Tobacco and alcohol use

The risk of oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers is highest in people who consume large amounts of both alcohol and tobacco. When both risk factors are present, the risk of cancer is greater than a simple multiplicative effect of the two individual risks.[1315]

Magnitude of Effect: About two to three times greater for oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers than the simple multiplicative effect, with risks for persons who both smoke and drink heavily approximately 5- to 14-fold that of persons who both never smoke and never drink.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Betel-quid chewing

Based on solid evidence, chewing betel quid alone or with added tobacco increases the risk of both oral cavity and oropharyngeal cancers.[3,4] Of the three primary components of betel quid (betel leaf, areca nut, and lime), the areca nut is the only one considered to be carcinogenic when chewed.

Magnitude of Effect: Relative risks for oral cavity cancer are high and typically stronger for betel quid with tobacco than for betel quid alone. Both products appear to confer a statistically significant increase in risk of oropharyngeal cancer.[4]

  • Study Design: Case-control, and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Human papillomavirus (HPV) infection

Based on solid evidence, HPV type 16 (HPV-16) infection causes oropharyngeal cancer.[6,16] Other high-risk HPV subtypes, including HPV type 18 (HPV-18), have been found in a small percentage of oropharyngeal cancers.[17,18]

Magnitude of Effect: Large. Oral infection with HPV-16 confers about a 15-fold increase in risk of oropharyngeal cancer relative to individuals without oral HPV-16 infection.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Interventions With Adequate Evidence of a Decreased Risk of Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers

Tobacco cessation

Based on solid evidence, cessation of exposure to tobacco (e.g., cigarettes, pipes, cigars, and smokeless tobacco) leads to a decrease in the risk of oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers.[19]

Magnitude of Effect: Decreased risk, moderate to large magnitude.

  • Study Design: Case-control and cohort studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Interventions With Inadequate Evidence of a Reduced Risk of Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers

Cessation of alcohol consumption

Based on fair evidence, cessation of alcohol consumption leads to a decrease in oral cavity and laryngeal cancer risk 20 years or more after cessation.[19]

Magnitude of Effect: Decreased risk, small to moderate magnitude.

  • Study Design: Case-control studies.
  • Internal Validity: Fair.
  • Consistency: Fair.
  • External Validity: Fair.

Vaccination against HPV-16 and the other high-risk subtypes

Vaccination against HPV-16 and HPV-18 has been shown to prevent approximately 90% of oral HPV-16/HPV-18 infections within 4 years of vaccination.[20] However, no data are available to assess whether vaccination at any age will lead to reduced risk of oropharyngeal cancer at current typical ages of diagnosis.[21]

  • Study Design: No studies available.
  • Internal Validity: Not applicable (N/A).
  • Consistency: N/A.
  • External Validity: N/A.
References
  1. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Personal habits and indoor combustions. Volume 100 E. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum 100 (Pt E): 1-538, 2012. [PUBMED Abstract]
  2. Castellsagué X, Alemany L, Quer M, et al.: HPV Involvement in Head and Neck Cancers: Comprehensive Assessment of Biomarkers in 3680 Patients. J Natl Cancer Inst 108 (6): djv403, 2016. [PUBMED Abstract]
  3. Song H, Wan Y, Xu YY: Betel quid chewing without tobacco: a meta-analysis of carcinogenic and precarcinogenic effects. Asia Pac J Public Health 27 (2): NP47-57, 2015. [PUBMED Abstract]
  4. Guha N, Warnakulasuriya S, Vlaanderen J, et al.: Betel quid chewing and the risk of oral and oropharyngeal cancers: a meta-analysis with implications for cancer control. Int J Cancer 135 (6): 1433-43, 2014. [PUBMED Abstract]
  5. Atienza JA, Dasanu CA: Incidence of second primary malignancies in patients with treated head and neck cancer: a comprehensive review of literature. Curr Med Res Opin 28 (12): 1899-909, 2012. [PUBMED Abstract]
  6. Kreimer AR, Johansson M, Waterboer T, et al.: Evaluation of human papillomavirus antibodies and risk of subsequent head and neck cancer. J Clin Oncol 31 (21): 2708-15, 2013. [PUBMED Abstract]
  7. U.S. Department of Health and Human Services: The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General. U.S. Department of Health and Human Services, CDC, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2014. Also available online. Last accessed December 30, 2024.
  8. Vineis P, Alavanja M, Buffler P, et al.: Tobacco and cancer: recent epidemiological evidence. J Natl Cancer Inst 96 (2): 99-106, 2004. [PUBMED Abstract]
  9. Hashibe M, Brennan P, Benhamou S, et al.: Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. J Natl Cancer Inst 99 (10): 777-89, 2007. [PUBMED Abstract]
  10. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Smokeless tobacco and some tobacco-specific N-nitrosamines. IARC Monogr Eval Carcinog Risks Hum 89: 1-592, 2007. [PUBMED Abstract]
  11. Purdue MP, Hashibe M, Berthiller J, et al.: Type of alcoholic beverage and risk of head and neck cancer–a pooled analysis within the INHANCE Consortium. Am J Epidemiol 169 (2): 132-42, 2009. [PUBMED Abstract]
  12. Islami F, Tramacere I, Rota M, et al.: Alcohol drinking and laryngeal cancer: overall and dose-risk relation–a systematic review and meta-analysis. Oral Oncol 46 (11): 802-10, 2010. [PUBMED Abstract]
  13. Hashibe M, Brennan P, Chuang SC, et al.: Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev 18 (2): 541-50, 2009. [PUBMED Abstract]
  14. Lubin JH, Purdue M, Kelsey K, et al.: Total exposure and exposure rate effects for alcohol and smoking and risk of head and neck cancer: a pooled analysis of case-control studies. Am J Epidemiol 170 (8): 937-47, 2009. [PUBMED Abstract]
  15. Mello FW, Melo G, Pasetto JJ, et al.: The synergistic effect of tobacco and alcohol consumption on oral squamous cell carcinoma: a systematic review and meta-analysis. Clin Oral Investig 23 (7): 2849-2859, 2019. [PUBMED Abstract]
  16. Hobbs CG, Sterne JA, Bailey M, et al.: Human papillomavirus and head and neck cancer: a systematic review and meta-analysis. Clin Otolaryngol 31 (4): 259-66, 2006. [PUBMED Abstract]
  17. D’Souza G, Kreimer AR, Viscidi R, et al.: Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356 (19): 1944-56, 2007. [PUBMED Abstract]
  18. Steinau M, Saraiya M, Goodman MT, et al.: Human papillomavirus prevalence in oropharyngeal cancer before vaccine introduction, United States. Emerg Infect Dis 20 (5): 822-8, 2014. [PUBMED Abstract]
  19. Marron M, Boffetta P, Zhang ZF, et al.: Cessation of alcohol drinking, tobacco smoking and the reversal of head and neck cancer risk. Int J Epidemiol 39 (1): 182-96, 2010. [PUBMED Abstract]
  20. Herrero R, Quint W, Hildesheim A, et al.: Reduced prevalence of oral human papillomavirus (HPV) 4 years after bivalent HPV vaccination in a randomized clinical trial in Costa Rica. PLoS One 8 (7): e68329, 2013. [PUBMED Abstract]
  21. Chaturvedi AK, Graubard BI, Broutian T, et al.: Effect of Prophylactic Human Papillomavirus (HPV) Vaccination on Oral HPV Infections Among Young Adults in the United States. J Clin Oncol 36 (3): 262-267, 2018. [PUBMED Abstract]

Incidence and Mortality

Oral cavity and oropharynx cancers

From 2017 to 2021, the estimated age-adjusted incidence rate for cancers of the oral cavity and oropharynx in the United States was 11.5 cases per 100,000 persons per year.[1] Cancers of the tongue, oropharynx, and tonsil comprise the majority of cases (30% tongue and 24% oropharynx and tonsil). The estimated age-adjusted mortality rate for cancers of the oral cavity and oropharynx from 2018 to 2022 was 2.6 per 100,000 persons per year. U.S. incidence rates are about 1.7 times higher in men than in women. However, mortality rates are about 1.9 times higher in men than in women.[1] It is estimated that there will be 59,660 new cases of oral cavity and oropharynx cancer diagnosed in the United States in 2025 and 12,770 deaths due to these diseases.[2] Rates of oral cavity cancer vary greatly across the world, primarily because of differences in alcohol use, tobacco use, and betel-quid chewing and the products chewed.[3]

Although localized cancers of the oral cavity and oropharynx have an excellent anticipated 5-year survival rate of about 87.5%, only 26.4% of these cancer cases are diagnosed at this stage.[1] The overall 5-year relative survival rate for patients with cancers of the oral cavity and oropharynx combined is only 69%.[2] However, the 5-year relative survival rate is much lower in Black patients (57%) than in White patients (71%).[2] Additionally, the 5-year survival rate varies greatly by cancer site, with 52.1% survival among patients with cancer on the floor of the mouth and 73.8% survival among patients with cancer in the oropharynx and tonsil.[4]

Hypopharyngeal cancer

Hypopharyngeal cancers are rare, with approximately 2,500 new cases diagnosed in the United States each year and an annual incidence of 0.6 cases per 100,000 persons.[5] The 5-year survival rate for hypopharyngeal cancer is 35%.[5] New cases have been falling an average of 2% per year over the last 20 years.[5] This has been attributed to a reduction in cigarette smoking.

Laryngeal cancer

Laryngeal cancers are less common, with an annual incidence of 2.6 cases per 100,000 persons.[6] It is estimated that 13,020 new cases of laryngeal cancer will be diagnosed in the United States in 2025, and an estimated 3,910 individuals will die of this disease.[2] The 5-year relative survival rate for laryngeal cancer is 62%.[2] Age-adjusted death rates have been falling on average 1.6% each year from 2013 to 2022.[6] This has been attributed to a reduction in cigarette smoking.

References
  1. National Cancer Institute: SEER Cancer Stat Facts: Oral Cavity and Pharynx Cancer. National Cancer Institute. Available online. Last accessed December 30, 2024.
  2. American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
  3. Shield KD, Ferlay J, Jemal A, et al.: The global incidence of lip, oral cavity, and pharyngeal cancers by subsite in 2012. CA Cancer J Clin 67 (1): 51-64, 2017. [PUBMED Abstract]
  4. 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.
  5. Kuo P, Chen MM, Decker RH, et al.: Hypopharyngeal cancer incidence, treatment, and survival: temporal trends in the United States. Laryngoscope 124 (9): 2064-9, 2014. [PUBMED Abstract]
  6. National Cancer Institute: SEER Cancer Stat Facts: Laryngeal Cancer. National Cancer Institute. Available online. Last accessed April 14, 2025.

Factors With Adequate Evidence of an Increased Risk of Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers

Tobacco Use

Tobacco use is implicated in most cases of oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers.[1] All forms of tobacco use (cigarettes, pipes, cigars, snuff, chewing tobacco, gutka [betel quid with tobacco added], and other smoked and smokeless products) increase the risk of these cancers.[1,2] Epidemiological studies consistently demonstrate that cigarette smokers have a higher incidence of mortality from head and neck squamous cell cancers compared with lifetime nonsmokers, and there is general consensus that the relationship is causal. Risk for current smokers ranges from fourfold to fivefold for oral cavity, oropharyngeal, and hypopharyngeal cancers to tenfold for laryngeal cancer when compared with never-smokers.[3] However, other epidemiological studies have observed smaller and larger increases in risk, with some variation by anatomic location. Betel-quid chewing is prevalent in many countries in south and southeast Asia, including China and India, and is an important risk factor for both oral cavity and oropharyngeal cancers.[4]

Alcohol Use

Alcohol use is a major independent risk factor for the development of head and neck squamous cell cancer.[5] Most epidemiological studies demonstrate an increase in risk with increasing drinks per day, with a more than twofold to sixfold increase in risk for individuals who consume two or more drinks a day relative to nonconsumers.[6] Associations are observed in studies that adjust for confounding by smoking, as well as in studies of nonsmokers.[7] There is a suggestion that consumption of beer and hard liquor confers a greater risk than does wine consumption.[6]

Tobacco and Alcohol Use

Head and neck squamous cell cancer risk is highest in people who consume large amounts of both alcohol and tobacco.[810] When both risk factors are present, the risk of oral cavity and oropharyngeal cancer is typically about two to three times greater than a simple multiplicative or additive effect.[11] In a study that pooled data from 17 case-control studies, individuals who consumed more than a pack of cigarettes and three or more alcoholic drinks per day had a 15-fold increased risk of oral cavity cancer, 14-fold increased risk of oropharyngeal cancer, and 36-fold increased risk of laryngeal cancer relative to individuals who neither smoked nor drank.[8,11]

Betel-Quid Chewing

Betel quid is composed of betel leaf, areca nut, and lime; gutka is betel quid with added tobacco. Both betel-quid and gutka chewing increase the risk of cancer of the oral cavity and oropharynx.[4,12] The carcinogenic component of chewed betel quid arises from the areca nut.[4]

Relative risks are typically stronger for betel quid with tobacco than for betel quid alone.[12] A meta-analysis of oral cavity cancer studies conducted on the Indian subcontinent calculated a statistically significant eightfold increase in risk for betel quid with tobacco and a statistically significant twofold increase in risk for betel quid alone.[12] A statistically significant tenfold increase in oral cavity cancer risk for betel-quid chewing was demonstrated by studies conducted in China or Taiwan. A meta-analysis of oropharyngeal cancer studies conducted on the Indian subcontinent calculated a statistically significant fourfold increase in risk for betel quid with tobacco and a statistically significant twofold increase in risk for betel quid alone.[12] Studies of head and neck cancer (without specification of subsite) suggest that increases in risk are positively correlated with chewing frequency and duration.[4]

Human Papillomavirus (HPV) Infection

HPV type 16 (HPV-16) infection is a sufficient, but not necessary, cause of head and neck cancers, with a greater causal relationship with oropharyngeal cancer.[13,14] A meta-analysis of five case-control studies of HPV-16 positivity in either serum or tissue calculated an odds ratio of 15.1 (95% confidence interval [CI], 6.8–33.7) for cancer of the tonsils, 4.3 (95% CI, 2.1–8.9) for other oropharyngeal sites, and 2.0 for both oral cavity (95% CI, 1.2–3.4) and larynx (95% CI, 1.0–4.2).[14] In a case-control study, the observed strong association of HPV-16 serologic status and oropharyngeal cancer did not vary at different levels of tobacco or alcohol use.[15] HPV-16 E6 seroconversion was shown to occur over a range of 6 to 28 years before oropharyngeal cancer diagnosis, at a median age of 52 years.[16] Thus, the HPV-16 infection that increases the risk of oropharyngeal cancer may occur in individuals aged 20 to 40 years. A recent national survey observed that men have a higher prevalence of oral HPV than women (11.5% vs. 3.2%), including high-risk HPV subtypes (7.3% of men; 1.4% of women).[17]

Other high-risk HPV subtypes, including HPV type 18, have been found in a smaller proportion of oropharyngeal cancers.[15,18]

References
  1. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Personal habits and indoor combustions. Volume 100 E. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum 100 (Pt E): 1-538, 2012. [PUBMED Abstract]
  2. U.S. Department of Health and Human Services: The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General. U.S. Department of Health and Human Services, CDC, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2014. Also available online. Last accessed December 30, 2024.
  3. Vineis P, Alavanja M, Buffler P, et al.: Tobacco and cancer: recent epidemiological evidence. J Natl Cancer Inst 96 (2): 99-106, 2004. [PUBMED Abstract]
  4. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Betel-quid and areca-nut chewing and some areca-nut derived nitrosamines. IARC Monogr Eval Carcinog Risks Hum 85: 1-334, 2004. [PUBMED Abstract]
  5. Hashibe M, Brennan P, Benhamou S, et al.: Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. J Natl Cancer Inst 99 (10): 777-89, 2007. [PUBMED Abstract]
  6. Purdue MP, Hashibe M, Berthiller J, et al.: Type of alcoholic beverage and risk of head and neck cancer–a pooled analysis within the INHANCE Consortium. Am J Epidemiol 169 (2): 132-42, 2009. [PUBMED Abstract]
  7. Goldstein BY, Chang SC, Hashibe M, et al.: Alcohol consumption and cancers of the oral cavity and pharynx from 1988 to 2009: an update. Eur J Cancer Prev 19 (6): 431-65, 2010. [PUBMED Abstract]
  8. Hashibe M, Brennan P, Chuang SC, et al.: Interaction between tobacco and alcohol use and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. Cancer Epidemiol Biomarkers Prev 18 (2): 541-50, 2009. [PUBMED Abstract]
  9. Lubin JH, Purdue M, Kelsey K, et al.: Total exposure and exposure rate effects for alcohol and smoking and risk of head and neck cancer: a pooled analysis of case-control studies. Am J Epidemiol 170 (8): 937-47, 2009. [PUBMED Abstract]
  10. Mello FW, Melo G, Pasetto JJ, et al.: The synergistic effect of tobacco and alcohol consumption on oral squamous cell carcinoma: a systematic review and meta-analysis. Clin Oral Investig 23 (7): 2849-2859, 2019. [PUBMED Abstract]
  11. Blot WJ, McLaughlin JK, Winn DM, et al.: Smoking and drinking in relation to oral and pharyngeal cancer. Cancer Res 48 (11): 3282-7, 1988. [PUBMED Abstract]
  12. Guha N, Warnakulasuriya S, Vlaanderen J, et al.: Betel quid chewing and the risk of oral and oropharyngeal cancers: a meta-analysis with implications for cancer control. Int J Cancer 135 (6): 1433-43, 2014. [PUBMED Abstract]
  13. Kreimer AR, Johansson M, Waterboer T, et al.: Evaluation of human papillomavirus antibodies and risk of subsequent head and neck cancer. J Clin Oncol 31 (21): 2708-15, 2013. [PUBMED Abstract]
  14. Hobbs CG, Sterne JA, Bailey M, et al.: Human papillomavirus and head and neck cancer: a systematic review and meta-analysis. Clin Otolaryngol 31 (4): 259-66, 2006. [PUBMED Abstract]
  15. D’Souza G, Kreimer AR, Viscidi R, et al.: Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356 (19): 1944-56, 2007. [PUBMED Abstract]
  16. Kreimer AR, Ferreiro-Iglesias A, Nygard M, et al.: Timing of HPV16-E6 antibody seroconversion before OPSCC: findings from the HPVC3 consortium. Ann Oncol 30 (8): 1335-1343, 2019. [PUBMED Abstract]
  17. Sonawane K, Suk R, Chiao EY, et al.: Oral Human Papillomavirus Infection: Differences in Prevalence Between Sexes and Concordance With Genital Human Papillomavirus Infection, NHANES 2011 to 2014. Ann Intern Med 167 (10): 714-724, 2017. [PUBMED Abstract]
  18. Steinau M, Saraiya M, Goodman MT, et al.: Human papillomavirus prevalence in oropharyngeal cancer before vaccine introduction, United States. Emerg Infect Dis 20 (5): 822-8, 2014. [PUBMED Abstract]

Interventions With Adequate Evidence of a Decreased Risk of Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers

Tobacco Cessation

The cessation of cigarette smoking is associated with an approximately 50% reduction in risk of developing oral cavity, oropharyngeal, and hypopharyngeal cancers within 5 to 9 years,[1] and a return to a cancer risk comparable to that of never-smokers within 20 years.[1] For laryngeal cancer, the risk reduction is approximately 40% within 5 to 9 years, and a similar return to cancer risk comparable to that of never-smokers within 20 years.[1]

References
  1. Marron M, Boffetta P, Zhang ZF, et al.: Cessation of alcohol drinking, tobacco smoking and the reversal of head and neck cancer risk. Int J Epidemiol 39 (1): 182-96, 2010. [PUBMED Abstract]

Interventions With Inadequate Evidence of a Reduced Risk of Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers

Cessation of Alcohol Consumption

Because alcohol is associated with oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers in a dose-dependent fashion, it is believed that cessation or avoidance of alcohol use would result in reduced incidence. Most studies suggest that the risk of oral cavity and laryngeal cancer decreases as time from cessation increases; one pooled analysis of 13 studies observed a statistically significant reduction (odds ratio [OR], 0.45; 95% confidence interval [CI], 0.26–0.78) for oral cavity and laryngeal cancers (OR, 0.69; 95% CI, 0.52–0.91), relative to current drinkers, for those who ceased consumption 20 years or more ago. Among never-smokers, cessation of alcohol consumption leads to a decrease in oral cavity and laryngeal cancer risk more than 1 to 4 years after cessation. Data for oropharyngeal and hypopharyngeal cancers did not support risk reduction with alcohol cessation.[1]

Vaccination Against Human Papillomavirus (HPV) Type 16 (HPV-16) and Other High-Risk Subtypes

Vaccination against HPV-16 and type 18 (HPV-18) has been shown to prevent approximately 90% of oral HPV-16/HPV-18 infections within 4 years of vaccination.[2,3] Given the relatively recent adoption of vaccination and the age at which individuals are vaccinated, there is not yet evidence that vaccination at a young age will lead to a substantially reduced risk of HPV-associated oropharyngeal cancer later in life. In addition, no data are available to examine whether incidence or mortality would be reduced if vaccination occurred at an age closer to that at which oropharyngeal cancers tend to present.

References
  1. Marron M, Boffetta P, Zhang ZF, et al.: Cessation of alcohol drinking, tobacco smoking and the reversal of head and neck cancer risk. Int J Epidemiol 39 (1): 182-96, 2010. [PUBMED Abstract]
  2. Herrero R, Quint W, Hildesheim A, et al.: Reduced prevalence of oral human papillomavirus (HPV) 4 years after bivalent HPV vaccination in a randomized clinical trial in Costa Rica. PLoS One 8 (7): e68329, 2013. [PUBMED Abstract]
  3. Chaturvedi AK, Graubard BI, Broutian T, et al.: Effect of Prophylactic Human Papillomavirus (HPV) Vaccination on Oral HPV Infections Among Young Adults in the United States. J Clin Oncol 36 (3): 262-267, 2018. [PUBMED Abstract]

Latest Updates to This Summary (04/14/2025)

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.

Incidence and Mortality

Updated statistics with estimated new cases and deaths for oral cavity and pharynx cancer for 2025 (cited American Cancer Society as reference 2).

Updated statistics with estimated new cases and deaths for laryngeal cancer for 2025.

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 oral cavity, oropharyngeal, hypopharyngeal, and laryngeal cancers 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 Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers Prevention. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/hp/oral-prevention-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389416]

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.

Oropharyngeal Cancer Treatment (PDQ®)–Patient Version

Oropharyngeal Cancer Treatment (PDQ®)–Patient Version

General Information About Oropharyngeal Cancer

Key Points

  • Oropharyngeal cancer is a type of head and neck cancer that starts in the tissues of the oropharynx.
  • Smoking or being infected with human papillomavirus (HPV) can increase the risk of oropharyngeal cancer.
  • Signs and symptoms of oropharyngeal cancer include a lump in the neck and a sore throat.
  • Tests that examine the mouth and throat are used to diagnose and stage oropharyngeal cancer.
  • Some people may decide to get a second opinion.
  • Certain factors affect prognosis (chance of recovery) and treatment options.

Oropharyngeal cancer is a type of head and neck cancer that starts in the tissues of the oropharynx.

The pharynx is a hollow tube in the neck about 5 inches long that is made up of three parts:

  • The nasopharynx is the upper part of the pharynx, located behind the nose. The nostrils are connected to the nasopharynx. Openings on each side of the nasopharynx lead to the ears.
  • The oropharynx is the middle part, located beneath the nasopharynx.
  • The hypopharynx is the lowermost part of the pharynx, opening up to both the trachea (windpipe) and esophagus.

When we breathe or swallow, the pharynx acts as a passageway for air to reach the lungs and food to reach the stomach. Oropharyngeal cancer commonly begins in the squamous cells that line the oropharynx.

EnlargeAnatomy of the pharynx; drawing shows the nasopharynx, oropharynx, and hypopharynx. Also shown are the nasal cavity, oral cavity, hyoid bone, larynx, esophagus, and trachea.
Anatomy of the pharynx. The pharynx is a hollow, muscular tube inside the neck that starts behind the nose and opens into the larynx and esophagus. The three parts of the pharynx are the nasopharynx, oropharynx, and hypopharynx.

The oropharynx includes the:

EnlargeParts of the oropharynx; drawing shows the soft palate, side and back wall of the throat, tonsil, and the back one-third of the tongue.
Parts of the oropharynx. The oropharynx includes the soft palate, the side and back walls of the throat, the tonsils, and the back one-third of the tongue.

Sometimes, a person can have more than one cancer at the same time in the oropharynx and mouth, nose, throat, voice box (larynx), windpipe (trachea), or esophagus.

Smoking or being infected with human papillomavirus (HPV) can increase the risk of oropharyngeal cancer.

Oropharyngeal cancer is caused by certain changes in how oropharyngeal cells function, especially how they grow and divide into new cells. There are many risk factors for oropharyngeal cancer, but many do not directly cause cancer. Instead, they increase the chance of DNA damage in cells that may lead to oropharyngeal cancer. Learn more about how cancer develops at What Is Cancer?

A risk factor is anything that increases a person’s chance of getting a disease. Some risk factors for oropharyngeal cancer, like tobacco use, can be changed. Risk factors also include things you cannot change, like your family history. Learning about risk factors for oropharyngeal cancer can help you make choices that might prevent or lower your risk of getting it.

The most common risk factors for oropharyngeal cancer include:

The number of cases of oropharyngeal cancers linked to HPV infection is increasing. Learn more about HPV and Cancer.

Signs and symptoms of oropharyngeal cancer include a lump in the neck and a sore throat.

Sometimes oropharyngeal cancer does not cause early signs and symptoms. When signs and symptoms occur, they may include:

  • a sore throat that does not go away
  • trouble swallowing
  • trouble opening the mouth fully
  • trouble moving the tongue
  • weight loss for no known reason
  • ear pain
  • a lump in the back of the mouth, throat, or neck
  • a white patch on the tongue or lining of the mouth that does not go away
  • coughing up blood

These problems may be caused by conditions other than oropharyngeal cancer. Check with your doctor if you have any of these problems to find out the cause and begin treatment, if needed.

Tests that examine the mouth and throat are used to diagnose and stage oropharyngeal cancer.

If you have symptoms that suggest oropharyngeal cancer, your doctor will need to find out if these are due to cancer or another problem. They will ask when the symptoms started and how often you have been having them. They will also ask about your personal and family health history and do a physical exam. Based on these results, the doctor may recommend other tests. If you are diagnosed with oropharyngeal cancer, the results of these tests will help you and your doctor plan treatment.

The following tests and procedures are used to diagnose and stage oropharyngeal cancer:

  • During a physical exam of the mouth and neck, the doctor or dentist looks at the mouth and neck, under the tongue, and down the throat with a small, long-handled mirror to check for abnormal areas.
  • A neurological exam uses a series of questions and tests to check the brain, spinal cord, and nerve function. The exam checks your 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.
  • PET-CT scan combines the pictures from a positron emission tomography (PET) scan and a computed tomography (CT) scan. The PET and CT scans are done at the same time with the same machine. The combined scans give more detailed pictures of areas inside the body than either scan gives by itself.
    • For the PET scan, a small amount of radioactive glucose (sugar) is injected into a vein. The scanner rotates around the body and makes a picture of where glucose is being used in the body. Because cancer cells often take up more glucose than normal cells, the pictures can be used to find cancer cells in the body.
    • For the CT scan, a series of detailed pictures of areas inside the body, such as the head, neck, chest, and lymph nodes, is taken from different angles. A dye is injected into a vein or swallowed to help the organs or tissues show up more clearly.
  • 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).
  • 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. A fine-needle biopsy is usually done to remove a sample of tissue using a thin needle.

    The following procedures may be used to remove samples of cells or tissue:

    • Endoscopy is a procedure to look at organs and tissues inside the body to check for abnormal areas. An endoscope is inserted through an incision (cut) in the skin or opening in the body, such as the mouth or nose. An endoscope is a thin, tube-like instrument with a light and a lens for viewing. It may also have a tool to remove abnormal tissue or lymph node samples, which are checked under a microscope for signs of disease. The nose, throat, back of the tongue, esophagus, stomach, larynx, windpipe, and large airways will be checked. The type of endoscopy is named for the part of the body that is being examined. For example, pharyngoscopy is an exam to check the pharynx.
    • Laryngoscopy is a procedure in which the doctor checks the larynx (voice box) with a mirror or a laryngoscope to check for abnormal areas. A laryngoscope is a thin, tube-like instrument with a light and a lens for viewing the inside of the throat and voice box. It may also have a tool to remove tissue samples, which are checked under a microscope for signs of cancer.

    If cancer is found, the following test may be done to study the cancer cells:

    • HPV test (human papillomavirus test) is a laboratory test used to check the sample of tissue for certain types of HPV infection, such as HPV type 16. This test is done because oropharyngeal cancer can be caused by HPV infection. This is important because HPV-positive oropharyngeal cancer has a better prognosis and is treated differently than HPV-negative oropharyngeal cancer.

    Learn about the type of information that can be found in a pathologist’s report about the cells or tissue removed during a biopsy at Pathology Reports.

Some people may decide to get a second opinion.

You may want to get a second opinion to confirm your oropharyngeal 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.

Learn more about choosing a doctor and getting a second opinion at 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, hospital, or getting a second opinion. For questions you might want to ask at your appointments, visit Questions to Ask Your Doctor About Cancer.

Certain factors affect prognosis (chance of recovery) and treatment options.

The prognosis depends on:

  • whether the person has HPV infection of the oropharynx
  • whether the person has a history of smoking cigarettes for 10 or more pack years
  • the stage of the cancer
  • the number and size of lymph nodes with cancer

Oropharyngeal tumors related to HPV infection have a better prognosis and are less likely to recur than tumors not linked to HPV infection.

Treatment options depend on:

  • the stage of the cancer
  • keeping the person’s ability to speak and swallow as normal as possible
  • the person’s general health

People with oropharyngeal cancer have an increased risk of another cancer in the head or neck. This risk is increased if a person continues to smoke or drink alcohol after treatment.

For more information, visit Cigarette Smoking: Health Risks and How to Quit.

Stages of Oropharyngeal Cancer

Key Points

  • The following stages are used for HPV-positive oropharyngeal cancer:
    • Stage I (also called stage 1) oropharyngeal cancer
    • Stage II (also called stage 2) oropharyngeal cancer
    • Stage III (also called stage 3) oropharyngeal cancer
    • Stage IV (also called stage 4) oropharyngeal cancer
  • The following stages are used for HPV-negative oropharyngeal cancer:
    • Stage 0 (also called carcinoma in situ of the oropharynx)
    • Stage I (also called stage 1) oropharyngeal cancer
    • Stage II (also called stage 2) oropharyngeal cancer
    • Stage III (also called stage 3) oropharyngeal cancer
    • Stage IV (also called stage 4) oropharyngeal cancer
  • Oropharyngeal cancer can recur (come back) after it has been treated.

Cancer stage describes the extent of cancer in the body.

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. Knowing the cancer stage helps plan treatment. 

There are several staging systems for cancer that describe the extent of the cancer. Oropharyngeal 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 the cancer. When talking to you about your diagnosis, your doctor may describe the cancer as one of these stages.

Learn more about Cancer Staging. 

The following stages are used for HPV-positive oropharyngeal cancer:

Stage I (also called stage 1) oropharyngeal cancer

In stage I, one of the following is true:

  • one or more lymph nodes with cancer that is HPV 16–positive are found but the place where the cancer began is not known. The lymph nodes with cancer are 6 centimeters or smaller, on one side of the neck; or
  • cancer is found in the oropharynx (throat) and the tumor is 4 centimeters or smaller. Cancer may have spread to one or more lymph nodes that are 6 centimeters or smaller, on the same side of the neck as the primary tumor.
EnlargeDrawing shows different sizes of a tumor in centimeters (cm) compared to the size of a pea (1 cm), a peanut (2 cm), a grape (3 cm), a walnut (4 cm), a lime (5 cm), an egg (6 cm), a peach (7 cm), and a grapefruit (10 cm). Also shown is a 10-cm ruler and a 4-inch ruler.
Tumor 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 II (also called stage 2) oropharyngeal cancer

In stage II, one of the following is true:

  • one or more lymph nodes with cancer that is HPV 16–positive are found but the place where the cancer began is not known. The lymph nodes with cancer are 6 centimeters or smaller, on one or both sides of the neck; or
  • the tumor is 4 centimeters or smaller. Cancer has spread to lymph nodes that are 6 centimeters or smaller, on the opposite side of the neck as the primary tumor or on both sides of the neck; or
  • the tumor is larger than 4 centimeters or cancer has spread to the top of the epiglottis (the flap that covers the trachea during swallowing). Cancer may have spread to one or more lymph nodes that are 6 centimeters or smaller, anywhere in the neck.

Stage III (also called stage 3) oropharyngeal cancer

In stage III, one of the following is true:

  • cancer has spread to the larynx (voice box), front part of the roof of the mouth, lower jaw, muscles that move the tongue, or to other parts of the head or neck. Cancer may have spread to lymph nodes in the neck; or
  • the tumor is any size and cancer may have spread to the larynx, front part of the roof of the mouth, lower jaw, muscles that move the tongue, or to other parts of the head or neck. Cancer has spread to one or more lymph nodes that are larger than 6 centimeters, anywhere in the neck.

Stage IV (also called stage 4) oropharyngeal cancer

In stage IV, cancer has spread to other parts of the body, such as the lung or bone.

Stage IV oropharyngeal cancer is also called metastatic oropharyngeal 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 oropharyngeal cancer spreads to the lung, the cancer cells in the lung are actually oropharyngeal cancer cells. The disease is called metastatic oropharyngeal cancer, not lung cancer. Learn more in Metastatic Cancer: When Cancer Spreads.

The following stages are used for HPV-negative oropharyngeal cancer:

Stage 0 (also called carcinoma in situ of the oropharynx)

In stage 0, abnormal cells are found in the lining of the oropharynx (throat). These abnormal cells may become cancer and spread into nearby normal tissue.

Stage I (also called stage 1) oropharyngeal cancer

In stage I, cancer has formed. The tumor is 2 centimeters or smaller.

EnlargeDrawing shows different sizes of a tumor in centimeters (cm) compared to the size of a pea (1 cm), a peanut (2 cm), a grape (3 cm), a walnut (4 cm), a lime (5 cm), an egg (6 cm), a peach (7 cm), and a grapefruit (10 cm). Also shown is a 10-cm ruler and a 4-inch ruler.
Tumor 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 II (also called stage 2) oropharyngeal cancer

In stage II, the tumor is larger than 2 centimeters but not larger than 4 centimeters.

Stage III (also called stage 3) oropharyngeal cancer

In stage III, the cancer:

  • is either larger than 4 centimeters or has spread to the top of the epiglottis (the flap that covers the trachea during swallowing); or
  • is any size. Cancer has spread to one lymph node that is 3 centimeters or smaller, on the same side of the neck as the primary tumor.

Stage IV (also called stage 4) oropharyngeal cancer

Stage IV is divided into stages IVA, IVB, and IVC.

  • In stage IVA, cancer:
    • has spread to the larynx (voice box), front part of the roof of the mouth, lower jaw, or muscles that move the tongue. Cancer may have spread to one lymph node that is 3 centimeters or smaller, on the same side of the neck as the primary tumor; or
    • is any size and may have spread to the top of the epiglottis, larynx, front part of the roof of the mouth, lower jaw, or muscles that move the tongue. Cancer has spread to one of the following:
      • one lymph node that is larger than 3 centimeters but not larger than 6 centimeters, on the same side of the neck as the primary tumor; or
      • more than one lymph node that is 6 centimeters or smaller, anywhere in the neck.
  • In stage IVB, cancer:
    • has spread to the muscle that moves the lower jaw, the bone attached to the muscle that moves the lower jaw, the base of the skull, or to the area behind the nose or around the carotid artery. Cancer may have spread to lymph nodes in the neck; or
    • may be any size and may have spread to other parts of the head or neck. Cancer has spread to a lymph node that is larger than 6 centimeters or has spread through the outside covering of a lymph node into nearby connective tissue.
  • In stage IVC, cancer has spread to other parts of the body, such as the lung, liver, or bone.

Stage IV oropharyngeal cancer is also called metastatic oropharyngeal 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 oropharyngeal cancer spreads to the lung, the cancer cells in the lung are actually oropharyngeal cancer cells. The disease is called metastatic oropharyngeal cancer, not lung cancer. Learn more in Metastatic Cancer: When Cancer Spreads.

Oropharyngeal cancer can recur (come back) after it has been treated.

Recurrent oropharyngeal cancer is cancer that has come back after it has been treated. If oropharyngeal cancer comes back, it may come back in the oropharynx, lymph nodes, or other parts of the body, such as the lungs, bone, or liver. Tests will help determine where in the body the cancer has returned. The type of treatment that you have for recurrent oropharyngeal cancer will depend on where it has come back.

Learn more in Recurrent Cancer: When Cancer Comes Back.

Treatment Option Overview

Key Points

  • There are different types of treatment for people with oropharyngeal cancer.
  • People with oropharyngeal cancer should have their treatment planned by a team of doctors with expertise in treating head and neck cancer.
  • The following types of treatment are used:
    • Surgery
    • Radiation therapy
    • Chemotherapy
    • Targeted therapy
    • Immunotherapy
  • New types of treatment are being tested in clinical trials.
  • Treatment for oropharyngeal cancer may cause side effects.
  • Follow-up care may be needed.

There are different types of treatment for people with oropharyngeal cancer.

Different types of treatments are available for oropharyngeal 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. Learn more at Questions to Ask Your Doctor About Your Treatment.   

People with oropharyngeal cancer should have their treatment planned by a team of doctors with expertise in treating head and neck cancer.

An oncologist, a doctor who specializes in treating people with cancer, oversees treatment for oropharyngeal cancer. Because the oropharynx helps in breathing, eating, and talking, you may need help adjusting to the side effects of the cancer and its treatment. The oncologist may refer you to other health care providers who are experts in treating head and neck cancer and also specialize in other areas of medicine. Other specialists may include:

The following types of treatment are used:

Surgery

Surgery to remove the tumor is a common treatment for all stages of oropharyngeal cancer. A surgeon may remove the cancer and some of the healthy tissue around the cancer. After the surgeon removes all the cancer that can be seen at the time of the surgery, some people 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.

New types of surgery, including transoral robotic surgery, are being studied for the treatment of oropharyngeal cancer. Transoral robotic surgery may be used to remove cancer from hard-to-reach areas of the mouth and throat. Cameras attached to a robot give a 3-dimensional (3D) image that a surgeon can see. Using a computer, the surgeon guides very small tools at the ends of the robot arms to remove the cancer. This procedure may also be done using an endoscope.

Learn more about Surgery to Treat Cancer.

Radiation therapy

Radiation therapy uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing by damaging their DNA. 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 head and neck; drawing shows a patient lying on a table under a machine that is used to aim high-energy radiation at the cancer. An inset shows a mesh mask that helps keep the patient's head and neck from moving during treatment. The mask has pieces of white tape with small ink marks on it. The ink marks are used to line up the radiation machine in the same position before each treatment.
External-beam radiation therapy of the head and neck. A machine is used to aim high-energy radiation at the cancer. The machine can rotate around the patient, delivering radiation from many different angles to provide highly conformal treatment. A mesh mask helps keep the patient’s head and neck 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 radiation therapy can help keep radiation from damaging nearby healthy tissue. These types of radiation therapy include:

  • 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 body radiation therapy: Stereotactic body radiation therapy is a type of external radiation therapy. Special equipment is used to place the person in the same position for each radiation treatment. Once a day for several days, a radiation machine aims a larger than usual dose of radiation directly at the tumor. By having the person in the same position for each treatment, there is less damage to nearby healthy tissue. This procedure is also called stereotactic external-beam radiation therapy and stereotaxic radiation therapy.

In advanced oropharyngeal cancer, dividing the daily dose of radiation into smaller-dose treatments improves the way the tumor responds to treatment. This is called hyperfractionated radiation therapy.

Radiation therapy may work better in people who have stopped smoking before beginning treatment.

If the thyroid or pituitary gland are part of the radiation treatment area, the person has an increased risk of hypothyroidism (too little thyroid hormone). A blood test to check the thyroid hormone level in the body should be done before and after treatment.

Learn more about External Beam Radiation Therapy for Cancer and Radiation Therapy Side Effects.

Chemotherapy

Chemotherapy uses drugs to stop the growth of cancer cells, either by killing the cells or by stopping them from dividing. Chemotherapy for oropharyngeal cancer is usually systemic, meaning it is taken by mouth or injected into a vein or muscle. When given this way, the drugs enter the bloodstream and can reach cancer cells throughout the body.

Chemotherapy drugs used to treat oropharyngeal cancer include:

Combinations of these drugs may be used. Other chemotherapy drugs not listed here may also be used.

Chemotherapy may be combined with other types of treatment, such as radiation therapy.

To learn more about how chemotherapy works, how it is given, common side effects, and more, visit Chemotherapy to Treat Cancer and Chemotherapy and You: Support for People With Cancer.

Targeted therapy

Targeted therapy uses drugs or other substances to identify and attack specific cancer cells. Cetuximab is a type of targeted therapy used to treat recurrent and metastatic oropharyngeal cancer.

Learn more about Targeted Therapy to Treat Cancer. 

Immunotherapy

Immunotherapy is a treatment that uses a person’s immune system to fight cancer. Your doctor may suggest biomarker tests to help predict your response to certain immunotherapy drugs. Learn more about Biomarker Testing for Cancer Treatment.   

Pembrolizumab and nivolumab are types of immunotherapy used to treat metastatic or recurrent oropharyngeal cancer.

Learn more about Immunotherapy to Treat Cancer.

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.

Learn more about clinical trials, including how to find and join one, at Clinical Trials Information for Patients and Caregivers.

Treatment for oropharyngeal cancer may cause side effects.

For information about side effects caused by treatment for cancer, visit our Side Effects page.

Follow-up care 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).

After treatment is complete, it is important to have head and neck exams to look for signs that the cancer has come back. Check-ups will be done every 6 to 12 weeks in the first year, every 3 months in the second year, every 3 to 4 months in the third year, and every 6 months thereafter.

Treatment of Stage I and Stage II Oropharyngeal Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of newly diagnosed stage I and stage II oropharyngeal cancer may include:

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 Stage III and Nonmetastatic Stage IV Oropharyngeal Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of newly diagnosed stage III oropharyngeal cancer and stage IV oropharyngeal cancer may include:

  • radiation therapy with or without chemotherapy given at the same time, for people with locally advanced cancer
  • radiation therapy alone for people who cannot have chemotherapy
  • chemotherapy given at the same time as radiation therapy
  • chemotherapy followed by radiation therapy given at the same time as more chemotherapy
  • a clinical trial of immunotherapy (nivolumab) with chemotherapy given at the same time as radiation therapy in people with cancer that has a higher risk of coming back
  • a clinical trial of radiation therapy with or without chemotherapy
  • a clinical trial of transoral surgery followed by standard- or low-dose radiation therapy with or without chemotherapy in people with HPV-positive oropharyngeal cancer

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 Metastatic Stage IV and Recurrent Oropharyngeal Cancer

For information about the treatments listed below, see the Treatment Option Overview section.

Treatment of oropharyngeal cancer that has metastasized or recurred in the oropharynx may include:

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 Oropharyngeal Cancer

About This PDQ Summary

About PDQ

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 oropharyngeal 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 Oropharyngeal Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/patient/adult/oropharyngeal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389310]

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.

Oropharyngeal Cancer Treatment (PDQ®)–Health Professional Version

Oropharyngeal Cancer Treatment (PDQ®)–Health Professional Version

General Information About Oropharyngeal Cancer

Incidence and Mortality

Estimated new cases and deaths from cancer of the oral cavity and pharynx in the United States in 2025:[1]

  • New cases: 59,660.
  • Deaths: 12,770.

The increasing incidence of oropharyngeal cancer is attributed to the rise in human papillomavirus (HPV)-associated cases. Men are almost three times as likely as women to have oropharyngeal cancer.[13]

Anatomy

Anatomically, the oropharynx is located between the soft palate superiorly and the hyoid bone inferiorly. It is continuous with the oral cavity anteriorly and communicates with the nasopharynx superiorly and the supraglottic larynx and hypopharynx inferiorly.

The oropharynx is divided into the following parts:[4]

  • Base of the tongue, which includes the pharyngoepiglottic folds and the glossoepiglottic folds.
  • Vallecula.
  • Tonsillar region, which includes the fossa and the anterior and posterior pillars.
  • Soft palate, which includes the uvula.
  • Posterior and lateral pharyngeal walls.

Regional lymph node anatomy of the head and neck

The regional lymph nodes of the head and neck include the lymph nodes that run parallel to the jugular veins, spinal accessory nerve, and facial artery and into the submandibular triangle. An understanding of regional anatomy and the status of regional lymph nodes is critical to the care of patients with head and neck cancer.[3,5,6] To facilitate communication regarding lymph node anatomy, the regions of the neck are described as levels I to V and retropharyngeal:

  • Level I contains the submental and submandibular lymph nodes.
  • Level II contains the upper jugular lymph nodes, which are above the digastric muscle.
  • Level III contains the midjugular lymph nodes, which are between the omohyoid muscle and the digastric muscle.
  • Level IV contains the lower jugular lymph nodes.
  • Level V contains the lymph nodes of the posterior triangle.
  • Retropharyngeal lymph nodes.
EnlargeLymph node groups of the neck; drawing shows six groups of lymph nodes in the neck: group IA and IB, group IIA and IIB, group III, group IV, group VA and VB, and group VI.

The retropharyngeal lymph nodes are a possible site for nodal spread in oropharyngeal cancer. A large retrospective cohort study from the MD Anderson Cancer Center described the clinical features of 981 patients with oropharyngeal cancer who underwent primary radiation therapy.[7][Level of evidence C1][Level of evidence C2]

  • The base of the tongue (47%) and the tonsil (46%) were the most common primary sites.
  • Most patients had stage T1 to T2 primary tumors (64%) and stage III to IVB disease (94%).
  • The incidence of radiographic retropharyngeal node involvement was 10% and was highest for the pharyngeal wall (23%) and lowest for the base of the tongue (6%).
  • Retropharyngeal lymph node involvement was associated with inferior 5-year local control and inferior recurrence-free survival, distant metastases−free survival, and overall survival on multivariate analysis.

Risk Factors

Risk factors for oropharyngeal squamous cell carcinoma (SCC) include:[8]

For more information, see Oral Cavity, Oropharyngeal, Hypopharyngeal, and Laryngeal Cancers Prevention.

HPV infection

Because of the decreased incidence of smoking in the United States, HPV-negative, smoking-related oropharyngeal cancer is decreasing; however, HPV-positive oropharyngeal cancer is increasing. According to the Surveillance, Epidemiology, and End Results (SEER) Program’s tissue repository data from 1988 to 2004, the prevalence of HPV-negative oropharyngeal cancer declined by 50%, while HPV-positive oropharyngeal cancers increased by 225%.[14][Level of evidence C1]

HPV-positive oropharyngeal cancers may represent a distinct disease entity that is caused by HPV infection and associated with an improved prognosis. Several studies indicate that individuals with HPV-positive tumors have significantly improved survival.[12,1517] Due to the prognostic impact of the HPV status in oropharyngeal cancer, the American Joint Committee on Cancer 8th edition staging separates oropharyngeal staging by HPV status.[5,6] In a prospective study of 253 patients with newly diagnosed or recurrent head and neck SCC, HPV was detected in 25% of the patients. Poor tumor grade and an oropharyngeal site independently increased the probability of the presence of HPV.[12] Oropharyngeal tumors are more likely to be HPV positive (57%) than oral cavity (12%) tumor sites and non-oropharyngeal (14%) sites. HPV-positive oropharyngeal cancers predominantly arise in the palatine or lingual tonsils. For tonsil or base-of-tongue sites, 62% of tumors were HPV positive, compared with 25% for other oropharyngeal sites.

Personal history of head and neck cancer

The risk of developing a second primary tumor in patients with tumors of the upper aerodigestive tract has been estimated to be 3% to 7% per year.[18,19] Because of this risk, patients require lifelong surveillance. Smoking and alcohol consumption after treatment are associated with the development of second primary tumors of the aerodigestive tract.[2022] Patients may need counseling to discontinue smoking and alcohol consumption.

The process of field cancerization may be partly responsible for the multiple, synchronous, primary SCCs that occur in oropharyngeal cancer and that are associated with a smoking history. Originally described in 1953, the concept of field cancerization holds that tumors develop in a multifocal fashion within a field of tissue chronically exposed to carcinogens.[23] Molecular studies that detect genetic alterations in histologically normal tissue from high-risk individuals have provided strong support for this concept.[2428]

A comparison of patients (N = 2,230) with index SCC of the oropharynx site and index SCC of non-oropharyngeal sites (i.e., oral cavity, larynx, and hypopharynx) was performed to determine the likelihood of developing second primary malignancies. The second primary malignancy rate was lower for patients with index oropharyngeal SCC than for patients with index non-oropharyngeal cancer (P < .001). Among patients with oropharyngeal SCC, former smokers had a 50% higher risk of second primary malignancy than never-smokers, and current smokers had a 100% higher risk than never-smokers (P trend = .008). These data suggest that patients who fit the typical HPV phenotype have a very low risk of second primary malignancy.[29]

Betel quid

The chewing of betel quid, a stimulant preparation commonly used in parts of Asia, increases the risk of oropharyngeal cancer.[30]

Other risk factors

Other risk factors may include:[8]

  • Defective elimination of acetaldehyde, a carcinogen generated by alcohol metabolism. In individuals, primarily those of East Asian race, who carry an inactive mutant allele of alcohol dehydrogenase-2, alcohol consumption is associated with a susceptibility to multiple metachronous oropharyngeal cancers that are caused by the decreased elimination of acetaldehyde.[31]

SCC of the oropharynx has not been associated with any specific chromosomal or genetic abnormalities. Genetic and chromosomal aberrations in these cancers are complex.[32,33] Despite the lack of specific genetic abnormalities, testing for genetic alterations or ploidy in early oropharyngeal lesions may identify patients who are at the greatest risk of disease progression and may lead to more-definitive therapy.[34]

Clinical Presentation

The clinical presentation of oropharyngeal cancer depends on the tumor’s location in the oropharynx. Oropharyngeal cancer may present in the following locations:

Tonsil

The anterior tonsillar pillar and tonsil are the most common location for a primary tumor of the oropharynx.[4] Lesions involving the anterior tonsillar pillar may appear as areas of dysplasia, inflammation, or a superficial spreading lesion. These cancers can spread across a broad region, including the lateral soft palate, retromolar trigone and buccal mucosa, and tonsillar fossa.[3,4] The lymphatic drainage is primarily to level II nodes.

Tumors of the posterior tonsillar pillar can extend inferiorly to involve the pharyngoepiglottic fold and the posterior aspect of the thyroid cartilage. These lesions more frequently involve level V nodes.

Lesions of the tonsillar fossa may be either exophytic or ulcerative and have a pattern of extension similar to those of the anterior tonsillar pillar. These tumors present as advanced-stage disease more often than do cancers of the tonsillar pillar. Approximately 75% of patients will present with stage III or stage IV disease.[3,4] The lymphatic drainage is primarily to level V nodes. Tumors of the posterior tonsillar pillar can extend inferiorly to involve the pharyngoepiglottic fold and the posterior aspect of the thyroid cartilage. These lesions more frequently involve level V nodes.

Signs and symptoms of tonsillar lesions may include:[3,4]

  • Pain.
  • Dysphagia.
  • Weight loss.
  • Ipsilateral referred otalgia.
  • A mass in the neck.

Base of the tongue

Clinically, cancers of the base of the tongue are insidious. These cancers can grow in either an infiltrative or exophytic pattern. Because the base of the tongue is devoid of pain fibers, these tumors are often asymptomatic until there is significant tumor progression.[4]

Signs and symptoms of advanced base-of-the-tongue cancers may include:[3,4]

  • Pain.
  • Dysphagia.
  • Weight loss.
  • Referred otalgia secondary to cranial nerve involvement.
  • Trismus secondary to pterygoid muscle involvement.
  • Fixation of the tongue that is caused by infiltration of the deep muscle.
  • A mass in the neck.

Lymph node metastasis is common because of the rich lymphatic drainage of the base of the tongue. Approximately 70% or more of patients with advanced base-of-the-tongue cancers have ipsilateral cervical nodal metastases; 30% or fewer of such patients have bilateral, cervical lymph–node metastases.[4,35] The cervical lymph nodes involved commonly include levels II, III, IV, and V and retropharyngeal lymph nodes.

Soft palate

Soft palate tumors are primarily found on the anterior surface.[4] Lesions in this area may remain superficial and in early stages.[3] The lymphatic drainage is primarily to level II nodes.

Pharyngeal wall

Pharyngeal wall lesions can spread superiorly to involve the nasopharynx, posteriorly to infiltrate the prevertebral fascia, and inferiorly to involve the pyriform sinuses and hypopharyngeal walls. Primary lymphatic drainage is to the retropharyngeal nodes and level II and III nodes. Because most pharyngeal tumors extend past the midline, bilateral cervical metastases are common.

Early-stage tumors are often asymptomatic. Tumors of the pharyngeal wall are typically diagnosed in an advanced stage.[3,4]

Signs and symptoms of advanced pharyngeal wall tumors may include:

  • Pain.
  • Bleeding.
  • Dysphagia.
  • Weight loss.
  • A mass in the neck.

Leukoplakia

Leukoplakia is used only as a clinically descriptive term meaning that the observer sees a white patch that does not rub off, the significance of which depends on the histological findings.[8] Leukoplakia can range from hyperkeratosis to an actual early invasive carcinoma or may represent a fungal infection, lichen planus, or other benign oral disease.

Diagnostic Evaluation

The assessment of the primary tumor is based on inspection and palpation, when possible, and by indirect mirror examination. The appropriate nodal drainage areas are examined by careful palpation. The presence of tumor must be confirmed histologically. Any other pathological data obtained from a biopsy and additional radiographical studies are also considered.

The following procedures may be done to evaluate the primary tumor:

  • Positron emission tomography–computed tomography (PET-CT) scan.
  • Magnetic resonance imaging.
  • Endoscopy.
  • Laryngoscopy.
  • Biopsy and p16 testing to assess for HPV status.

A PET-CT scan yields morphological and metabolic data to assess the detection of primary tumor, nodal disease, and distant metastatic disease. It may also be used to guide radiation therapy planning. Retrospective data demonstrate that morphological and PET-glycolytic parameters (as measured by fluorodeoxyglucose PET-CT) are significantly larger (as measured by Response Evaluation Criteria In Solid Tumors [RECIST] longest diameter) and more heterogenous in HPV-negative disease than in HPV-positive disease in the primary tumor for oropharyngeal carcinoma. These PET-CT parameters also show higher standardized uptake value (SUV) max, SUV mean, and metabolic tumor volume in HPV-negative disease. However, the same PET parameters are frequently larger in the regional nodal disease in patients with HPV-positive disease.[36][Level of evidence C3]

Prognostic Factors and Survival

Prognostic factors for oropharyngeal carcinoma include:

  • HPV status.
  • Smoking history (10 or more pack-years).
  • Tumor stage and nodal status.

The criteria described in Table 1 are used to determine whether patients have low-, intermediate-, or high-risk oropharyngeal carcinoma. These criteria have been defined by using recursive partitioning analysis in a retrospective analysis of a randomized trial of patients with stage III and IV oropharyngeal SCC treated with chemoradiation.[17]

Table 1. Characteristics Associated With the Risk of Oropharyngeal Cancera
Degree of Risk Characteristics 3-y OS Rate
CI = confidence interval; HPV = human papillomavirus; OS = overall survival; + = positive; – = negative. For more information, see the AJCC Staging Groupings and TNM Definitions section.
aAng KK, Harris J, Wheeler R, et al.: Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363 (1): 24–35, 2010.
Low HPV+, smoking history of ≤10 pack-years, and N0–N2a nodal history 93% (95% CI, 88.3%–97.7%)
Intermediate HPV+, smoking history of >10 pack-years, and N2b–N3 nodal disease; or 70.8% (95% CI, 60.7%–80.8%)
HPV-, smoking history of ≤10 pack-years, and N2b–N3 nodal disease or T2–T3 tumors
High HPV- and smoking history >10 pack-years; or 46.2% (95% CI, 34.7%–57.7%)
HPV-, smoking history ≤10 pack-years, and T4 disease

Follow-Up After Treatment

A careful examination of the patient’s head and neck allows the physician to look for recurrence every 6 to 12 weeks for the first posttreatment year, every 3 months for the second year, every 3 to 4 months for the third year, and every 6 months thereafter.

References
  1. American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
  2. 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]
  3. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  4. Choi WH, Hu KS, Culliney B, et al.: Cancer of the oropharynx. In: Harrison LB, Sessions RB, Hong WK, eds.: Head and Neck Cancer: A Multidisciplinary Approach. 3rd ed. Lippincott, William & Wilkins, 2009, pp 285-335.
  5. HPV-Mediated (p16+) Oropharyngeal Cancer. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 113-21.
  6. Oropharynx (p16-) and Hypopharynx. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 123-35.
  7. Gunn GB, Debnam JM, Fuller CD, et al.: The impact of radiographic retropharyngeal adenopathy in oropharyngeal cancer. Cancer 119 (17): 3162-9, 2013. [PUBMED Abstract]
  8. Neville BW, Day TA: Oral cancer and precancerous lesions. CA Cancer J Clin 52 (4): 195-215, 2002 Jul-Aug. [PUBMED Abstract]
  9. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  10. Licitra L, Bernier J, Grandi C, et al.: Cancer of the oropharynx. Crit Rev Oncol Hematol 41 (1): 107-22, 2002. [PUBMED Abstract]
  11. Mork J, Lie AK, Glattre E, et al.: Human papillomavirus infection as a risk factor for squamous-cell carcinoma of the head and neck. N Engl J Med 344 (15): 1125-31, 2001. [PUBMED Abstract]
  12. Gillison ML, Koch WM, Capone RB, et al.: Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst 92 (9): 709-20, 2000. [PUBMED Abstract]
  13. D’Souza G, Kreimer AR, Viscidi R, et al.: Case-control study of human papillomavirus and oropharyngeal cancer. N Engl J Med 356 (19): 1944-56, 2007. [PUBMED Abstract]
  14. Chaturvedi AK, Engels EA, Pfeiffer RM, et al.: Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol 29 (32): 4294-301, 2011. [PUBMED Abstract]
  15. Ringström E, Peters E, Hasegawa M, et al.: Human papillomavirus type 16 and squamous cell carcinoma of the head and neck. Clin Cancer Res 8 (10): 3187-92, 2002. [PUBMED Abstract]
  16. Schwartz SR, Yueh B, McDougall JK, et al.: Human papillomavirus infection and survival in oral squamous cell cancer: a population-based study. Otolaryngol Head Neck Surg 125 (1): 1-9, 2001. [PUBMED Abstract]
  17. Ang KK, Harris J, Wheeler R, et al.: Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med 363 (1): 24-35, 2010. [PUBMED Abstract]
  18. Khuri FR, Lippman SM, Spitz MR, et al.: Molecular epidemiology and retinoid chemoprevention of head and neck cancer. J Natl Cancer Inst 89 (3): 199-211, 1997. [PUBMED Abstract]
  19. León X, Quer M, Diez S, et al.: Second neoplasm in patients with head and neck cancer. Head Neck 21 (3): 204-10, 1999. [PUBMED Abstract]
  20. Do KA, Johnson MM, Doherty DA, et al.: Second primary tumors in patients with upper aerodigestive tract cancers: joint effects of smoking and alcohol (United States). Cancer Causes Control 14 (2): 131-8, 2003. [PUBMED Abstract]
  21. Khuri FR, Kim ES, Lee JJ, et al.: The impact of smoking status, disease stage, and index tumor site on second primary tumor incidence and tumor recurrence in the head and neck retinoid chemoprevention trial. Cancer Epidemiol Biomarkers Prev 10 (8): 823-9, 2001. [PUBMED Abstract]
  22. Day GL, Blot WJ, Shore RE, et al.: Second cancers following oral and pharyngeal cancers: role of tobacco and alcohol. J Natl Cancer Inst 86 (2): 131-7, 1994. [PUBMED Abstract]
  23. Slaughter DP, Southwick HW, Smejkal W: Field cancerization in oral stratified squamous epithelium: clinical implications of multicentric origin. Cancer 6 (5): 963-8, 1953. [PUBMED Abstract]
  24. Braakhuis BJ, Tabor MP, Leemans CR, et al.: Second primary tumors and field cancerization in oral and oropharyngeal cancer: molecular techniques provide new insights and definitions. Head Neck 24 (2): 198-206, 2002. [PUBMED Abstract]
  25. Braakhuis BJ, Tabor MP, Kummer JA, et al.: A genetic explanation of Slaughter’s concept of field cancerization: evidence and clinical implications. Cancer Res 63 (8): 1727-30, 2003. [PUBMED Abstract]
  26. Tabor MP, Brakenhoff RH, van Houten VM, et al.: Persistence of genetically altered fields in head and neck cancer patients: biological and clinical implications. Clin Cancer Res 7 (6): 1523-32, 2001. [PUBMED Abstract]
  27. Tabor MP, Brakenhoff RH, Ruijter-Schippers HJ, et al.: Multiple head and neck tumors frequently originate from a single preneoplastic lesion. Am J Pathol 161 (3): 1051-60, 2002. [PUBMED Abstract]
  28. Ha PK, Califano JA: The molecular biology of mucosal field cancerization of the head and neck. Crit Rev Oral Biol Med 14 (5): 363-9, 2003. [PUBMED Abstract]
  29. Gan SJ, Dahlstrom KR, Peck BW, et al.: Incidence and pattern of second primary malignancies in patients with index oropharyngeal cancers versus index nonoropharyngeal head and neck cancers. Cancer 119 (14): 2593-601, 2013. [PUBMED Abstract]
  30. Ho PS, Ko YC, Yang YH, et al.: The incidence of oropharyngeal cancer in Taiwan: an endemic betel quid chewing area. J Oral Pathol Med 31 (4): 213-9, 2002. [PUBMED Abstract]
  31. Yokoyama A, Watanabe H, Fukuda H, et al.: Multiple cancers associated with esophageal and oropharyngolaryngeal squamous cell carcinoma and the aldehyde dehydrogenase-2 genotype in male Japanese drinkers. Cancer Epidemiol Biomarkers Prev 11 (9): 895-900, 2002. [PUBMED Abstract]
  32. Tremmel SC, Götte K, Popp S, et al.: Intratumoral genomic heterogeneity in advanced head and neck cancer detected by comparative genomic hybridization. Cancer Genet Cytogenet 144 (2): 165-74, 2003. [PUBMED Abstract]
  33. Brieger J, Jacob R, Riazimand HS, et al.: Chromosomal aberrations in premalignant and malignant squamous epithelium. Cancer Genet Cytogenet 144 (2): 148-55, 2003. [PUBMED Abstract]
  34. Forastiere A, Koch W, Trotti A, et al.: Head and neck cancer. N Engl J Med 345 (26): 1890-900, 2001. [PUBMED Abstract]
  35. Lindberg R: Distribution of cervical lymph node metastases from squamous cell carcinoma of the upper respiratory and digestive tracts. Cancer 29 (6): 1446-9, 1972. [PUBMED Abstract]
  36. Tahari AK, Alluri KC, Quon H, et al.: FDG PET/CT imaging of oropharyngeal squamous cell carcinoma: characteristics of human papillomavirus-positive and -negative tumors. Clin Nucl Med 39 (3): 225-31, 2014. [PUBMED Abstract]

Cellular Classification of Oropharyngeal Cancer

Most oropharyngeal cancers are squamous cell carcinomas (SCCs).[13] SCCs may be noninvasive or invasive. For noninvasive SCC, the term carcinoma in situ is used. Histologically, invasive carcinomas are classified as well differentiated, moderately differentiated, poorly differentiated, or undifferentiated. SCCs are usually moderately or poorly differentiated.[4] Grading the deep invasive margins (i.e., invasive front) of SCC may provide better prognostic information than grading the entire tumor.[5] Human papillomavirus (HPV)-positive oropharyngeal cancers arising from the lingual and palatine tonsils are a distinct molecular-pathological entity that is linked to infection with HPV, especially HPV-16. Compared with HPV-negative tumors, HPV-positive tumors are more frequently poorly differentiated and nonkeratinizing. They are strongly associated with basaloid morphology and less likely to have TP53 variants.[6]

Immunohistochemical examination of tissues for the expression of the biomarker Ki-67, a proliferation antigen, may complement histological grading. As a molecular indicator of epithelial dysplasia of the oropharynx, Ki-67 expression appears to correlate well with loss of heterozygosity (LOH) in tumor cells. In a retrospective study involving 43 tissue samples from 25 patients, the assessment of proliferation with Ki-67 was a better surrogate for LOH than was histological grading.[7]

Other types of oropharyngeal cancer include:

  • Minor salivary gland tumors.
  • Lymphomas.
  • Lymphoepitheliomas (e.g., tonsillar fossa).

For more information, see Salivary Gland Cancer Treatment, Hodgkin Lymphoma Treatment, Indolent B-Cell Non-Hodgkin Lymphoma Treatment, Aggressive B-Cell Non-Hodgkin Lymphoma Treatment, and Peripheral T-Cell Non-Hodgkin Lymphoma Treatment.

References
  1. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  2. HPV-Mediated (p16+) Oropharyngeal Cancer. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 113-21.
  3. Oropharynx (p16-) and Hypopharynx. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 123-35.
  4. Oral cavity and oropharynx. In: Rosai J, ed.: Rosai and Ackerman’s Surgical Pathology. Vol. 1. 10th ed. Mosby Elsevier, 2011, pp. 237-264.
  5. Bryne M, Boysen M, Alfsen CG, et al.: The invasive front of carcinomas. The most important area for tumour prognosis? Anticancer Res 18 (6B): 4757-64, 1998 Nov-Dec. [PUBMED Abstract]
  6. Gillison ML, Koch WM, Capone RB, et al.: Evidence for a causal association between human papillomavirus and a subset of head and neck cancers. J Natl Cancer Inst 92 (9): 709-20, 2000. [PUBMED Abstract]
  7. Tabor MP, Braakhuis BJ, van der Wal JE, et al.: Comparative molecular and histological grading of epithelial dysplasia of the oral cavity and the oropharynx. J Pathol 199 (3): 354-60, 2003. [PUBMED Abstract]

Stage Information for Oropharyngeal Cancer

The staging system for oropharyngeal cancer is clinical rather than pathological. It is based on the best estimate of the extent of disease before treatment.

Clinical anatomical staging of oropharyngeal cancer involves the following clinical assessment and imaging techniques:

  • Inspection and palpation of sites (when feasible) and neck nodes.
  • Neurological examination of all cranial nerves.
  • A head and neck computed tomography (CT) scan with contrast to evaluate the mandible and maxilla.[1]
  • Magnetic resonance imaging to evaluate the extent of disease in the soft tissues.
  • Positron emission tomography (PET)–CT scan to assess primary, regional, and distant metastatic spread.
  • Complete endoscopy after completion of other staging studies to assess the surface extent of the tumor accurately and to facilitate biopsy. This procedure is typically performed under general anesthesia, which also allows palpation for deep muscle invasion. Because of the incidence of multiple primary tumors occurring simultaneously, a careful search for other primary tumors of the upper aerodigestive tract is indicated.[2]

PET has been investigated as an imaging modality for recurrent oropharyngeal cancer.[3]

American Joint Committee on Cancer (AJCC) Staging Groupings and TNM Definitions

The AJCC has designated staging by TNM (tumor, node, metastasis) classification to define oropharyngeal cancer.[2,4] Nonepithelial tumors such as those of lymphoid tissue, soft tissue, bone, and cartilage are not included.

The AJCC uses separate staging systems for human papillomavirus (HPV)-related squamous cell carcinoma of the oropharynx [4] and p16-negative squamous cancers of the oropharynx.[2]

AJCC prognostic stage groups for HPV-mediated (p16-positive) oropharyngeal cancer

Table 2. Definitions of TNM Stage Ia
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: HPV-Mediated (p16+) Oropharyngeal Cancer. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 113–21.
I T0, T1, or T2; N0 or N1; M0 T0, T1, or T2 = See Stage IV in Table 5 below.
N0 or N1 = See Stage IV in Table 5 below.
M0 = No distant metastasis.
Table 3. Definitions of TNM Stage IIa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: HPV-Mediated (p16+) Oropharyngeal Cancer. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 113–21.
II T0, T1, or T2; N2; M0 T0, T1, or T2 = See Stage IV in Table 5 below.
N2 = Contralateral or bilateral lymph nodes, none >6 cm.
M0 = No distant metastasis.
T3; N0, N1, or N2; M0 T3 = Tumor >4 cm in greatest dimension or extension to lingual surface of the epiglottis.
N0, N1, or N2 = See Stage IV in Table 5 below.
M0 = No distant metastasis.
Table 4. Definitions of TNM Stage IIIa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: HPV-Mediated (p16+) Oropharyngeal Cancer. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 113–21.
bMucosal extension to the lingual surface of the epiglottis from primary tumors of the base of the tongue and vallecula does not constitute invasion of the larynx.
III T0, T1, T2, T3, or T4; N3; M0 T0, T1, T2, T3, or T4 = See Stage IV in Table 5 below.
N3 = Lymph node(s) >6 cm.
M0 = No distant metastasis.
T4; N0, N1, N2, or N3; M0 T4 = Moderately advanced local disease. Tumor invades the larynx, extrinsic muscle of the tongue, medial pterygoid, hard palate, or mandible or beyond.b
N0, N1, N2, or N3 = See Stage IV in Table 5 below.
M0 = No distant metastasis.
Table 5. Definitions of TNM Stage IVa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: HPV-Mediated (p16+) Oropharyngeal Cancer. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 113–21.
bMucosal extension to the lingual surface of the epiglottis from primary tumors of the base of the tongue and vallecula does not constitute invasion of the larynx.
IV Any T, Any N, M1 T0 = No primary identified.
T1 = Tumor ≤2 cm in greatest dimension.
T2 = Tumor >2 cm but ≤4 cm in greatest dimension.
T3 = Tumor >4 cm in greatest dimension or extension to lingual surface of the epiglottis.
T4 = Moderately advanced local disease. Tumor invades the larynx, extrinsic muscle of the tongue, medial pterygoid, hard palate, or mandible or beyond.b
NX = Regional lymph nodes cannot be assessed.
N0 = No regional lymph node metastasis.
N1 = One or more ipsilateral lymph nodes, none >6 cm.
N2 = Contralateral or bilateral lymph nodes, none >6 cm.
N3 = Lymph node(s) >6 cm.
M1 = Distant metastasis.

AJCC prognostic stage groups for p16-negative squamous cancers of the oropharynx

Table 6. Definitions of TNM Stage 0a
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Oropharynx (p16−) and Hypopharynx. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 123–35.
The explanation for superscript b is at the end of Table 10.
0 Tis, N0, M0 Tis = Carcinoma in situ.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 7. Definitions of TNM Stage Ia
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Oropharynx (p16−) and Hypopharynx. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 123–35.
The explanation for superscript b is at the end of Table 10.
I T1, N0, M0 T1 = Tumor ≤2 cm in greatest dimension.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 8. Definitions of TNM Stage IIa
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis.
aReprinted with permission from AJCC: Oropharynx (p16−) and Hypopharynx. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 123–35.
The explanation for superscript b is at the end of Table 10.
II T2, N0, M0 T2 = Tumor >2 cm but ≤4 cm in greatest dimension.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
Table 9. Definitions of TNM Stage IIIa
Stage TNbM Description
T = primary tumor; N = regional lymph node; M = distant metastasis; ENE = extranodal extension.
aReprinted with permission from AJCC: Oropharynx (p16−) and Hypopharynx. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 123–35.
The explanation for superscript b is at the end of Table 10.
III T3, N0, M0 T3 = Tumor >4 cm in greatest dimension or extension to lingual surface of epiglottis.
N0 = No regional lymph node metastasis.
M0 = No distant metastasis.
T1, T2, T3; N1; M0 T1, T2, T3 = See Stage IVC in Table 10 below.
N1 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(–).
M0 = No distant metastasis.
Table 10. Definitions of TNM Stage IVA, IVB, and IVCa
Stage TNM Description
T = primary tumor; N = regional lymph node; M = distant metastasis; ENE = extranodal extension.
aReprinted with permission from AJCC: Oropharynx (p16−) and Hypopharynx. In: Edge SB, Byrd DR, Compton CC, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp 123–35.
bA designation of U or L may be used for any N category to indicate metastasis above the lower border of the cricoid (U) or below the lower border of the cricoid (L). Similarly, clinical and pathological ENE should be recorded as ENE(−) or ENE(+).
cMucosal extension to the lingual surface of the epiglottis from primary tumors of the base of the tongue and vallecula does not constitute invasion of the larynx.
IVA T4a; N0, N1; M0 T4a = Moderately advanced local disease. Tumor invades the larynx, extrinsic muscle of the tongue, medial pterygoid, hard palate, or mandible.c
N0, N1 = See Stage IVC below in this table.
M0 = No distant metastasis.
T1, T2, T3, T4a; N2; M0 T1, T2, T3, T4a = See Stage IVC below in this table.
N2 = Metastasis in a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(−); or metastases in multiple ipsilateral lymph nodes, none >6 cm in greatest dimension and ENE(−); or in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(−).
M0 = No distant metastasis.
IVB Any T, N3, M0 Any T = See Stage IVC below in this table.
N3 = Metastasis in a lymph node >6 cm in greatest dimension and ENE(−); or metastasis in any node(s) and clinically overt ENE(+).
M0 = No distant metastases.
T4b, Any N, M0 T4b = Very advanced local disease. Tumor invades lateral pterygoid muscle, pterygoid plates, lateral nasopharynx, or skull base or encases carotid artery.
Any N = See Stage IVC below in this table.
M0 = No distant metastasis.
IVC Any T, Any N, M1 TX = Primary tumor cannot be assessed.
Tis = Carcinoma in situ.
T1 = Tumor ≤2 cm in greatest dimension.
T2 = Tumor >2 cm but ≤4 cm in greatest dimension.
T3 = Tumor >4 cm in greatest dimension or extension to lingual surface of epiglottis.
T4 = Moderately advanced or very advanced local disease.
−T4a = Moderately advanced local disease. Tumor invades the larynx, extrinsic muscle of the tongue, medial pterygoid, hard palate, or mandible.c
−T4b = Very advanced local disease. Tumor invades lateral pterygoid muscle, pterygoid plates, lateral nasopharynx, or skull base or encases carotid artery.
NX = Regional lymph nodes cannot be assessed.
N0 = No regional lymph node metastasis.
N1 = Metastasis in a single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE(−).
N2 = Metastasis in a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(−); or metastases in multiple ipsilateral lymph nodes, none >6 cm in greatest dimension and ENE(−); or in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(−).
−N2a = Metastasis in a single ipsilateral node >3 cm but ≤6 cm in greatest dimension and ENE(−).
−N2b = Metastases in multiple ipsilateral nodes, none >6 cm in greatest dimension and ENE(−).
−N2c = Metastases in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE(−).
N3 = Metastasis in a lymph node >6 cm in greatest dimension and ENE(−); or metastasis in any node(s) and clinically overt ENE(+).
−N3a = Metastasis in a lymph node >6 cm in greatest dimension and ENE(−).
−N3b = Metastasis in any node(s) and clinically overt ENE(+).
M1 = Distant metastasis.
References
  1. Weber AL, Romo L, Hashmi S: Malignant tumors of the oral cavity and oropharynx: clinical, pathologic, and radiologic evaluation. Neuroimaging Clin N Am 13 (3): 443-64, 2003. [PUBMED Abstract]
  2. Oropharynx (p16-) and Hypopharynx. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 123-35.
  3. Wong RJ, Lin DT, Schöder H, et al.: Diagnostic and prognostic value of [(18)F]fluorodeoxyglucose positron emission tomography for recurrent head and neck squamous cell carcinoma. J Clin Oncol 20 (20): 4199-208, 2002. [PUBMED Abstract]
  4. HPV-Mediated (p16+) Oropharyngeal Cancer. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 113-21.

Treatment Option Overview for Oropharyngeal Cancer

An optimal approach for the treatment of oropharyngeal cancer is not easily defined because no single regimen offers a distinct superior-survival advantage. The literature reports various therapeutic options but does not present any valid comparative studies of these options. Treatment considerations should account for functional and performance status, including speech and swallowing outcomes.

Table 11. Treatment Options for Oropharyngeal Cancer
Stage (TNM Definitions) Treatment Options
Stage I and stage II oropharyngeal cancer Radiation therapy using standard fractionation
Surgery
Stage III and stage IV oropharyngeal cancer Surgery followed by postoperative radiation therapy (PORT) with or without concurrent chemotherapy for patients with locally advanced disease
Radiation therapy using altered fractionation
Concurrent chemoradiation therapy
Neoadjuvant chemotherapy followed by concurrent chemoradiation therapy
Chemoradiation therapy with immunotherapy (under clinical evaluation)
Treatment de-intensification using radiation dose de-escalation (under clinical evaluation)
Treatment de-intensification using transoral surgery followed by radiation dose de-escalation (under clinical evaluation)
Metastatic and recurrent oropharyngeal cancer Surgical resection, if technically feasible and the tumor does not respond to radiation therapy
Radiation therapy, if the tumor is not completely removed by surgery and curative doses of radiation have not been given previously
A second surgery, if the tumor was not completely removed initially and if technically feasible
Chemotherapy, for unresectable locoregionally recurrent disease
Additional radiation therapy using conventionally fractionated radiation therapy or hyperfractionated radiation therapy with concurrent chemotherapy
Stereotactic body radiation therapy with concurrent cetuximab
Immunotherapy
Clinical trials evaluating additional radiation therapy using hyperfractionated radiation therapy with concurrent chemotherapy, targeted therapy, stereotactic body radiation therapy, or immunotherapy

Surgery and/or Radiation Therapy

Surgery and radiation therapy have been the standard treatment for oropharyngeal cancer; however, outcome data from randomized trials are limited. Studies have evaluated whether to use surgery or radiation but have been underpowered.[1]

Evidence (surgery and/or radiation therapy):

  1. In a prospective randomized trial, 564 patients with head and neck cancer and N2 or N3 disease were assigned to either planned neck dissection or surveillance with positron emission tomography–computed tomography (PET-CT) scan.[2]
    • With a median follow up of 36 months, PET-CT surveillance resulted in fewer neck dissections compared with the surgical group (54 vs. 221), with a 2-year survival rate of 84.9% for the neck dissection group and 81.5% for the surgical group. The hazard ratio (HR) for death (HRdeath) slightly favored PET-CT−guided surveillance and indicated noninferiority (upper boundary 95% confidence interval [CI] for HR <1.50; P = .004).
  2. A pooled analysis of 6,400 patients with base-of-the-tongue oropharyngeal carcinoma from 51 reported series between 1970 and 2000 demonstrated the following:[3]
    • Local control rates of 79% (surgery with or without radiation therapy) and 76% (radiation therapy alone), (P = .087); locoregional control was 60% for surgery with or without radiation therapy versus 69% for radiation therapy alone (P = .009).
    • The 5-year survival rate was 49% for surgery with or without radiation therapy versus 52% (P = .2) for radiation therapy with or without neck dissection.
    • The rate of severe complications was 32% for the surgery group versus 3.8% for the radiation therapy group (P < .001).
    • The rate of fatal complications was 3.5% for the surgery group versus 0.4% for the radiation therapy group (P < .001).

    Historically, the posttreatment performance and functional status of patients with base-of-the-tongue primary tumors was better after radiation therapy than after surgery. Local control and survival are similar in both treatment options.[4,5]

  3. In the same study, the results for patients with squamous cell carcinoma (SCC) in the tonsillar region who underwent surgery with or without radiation therapy versus radiation therapy with or without neck dissection were as follows:[3]
    • Local control rates of 70% (surgery with or without radiation therapy) and 68% (radiation therapy), (P = .2); locoregional control was 65% for surgery with or without radiation therapy versus 69% for radiation therapy alone (P = .1).
    • The 5-year survival rate was 47% for surgery with or without radiation therapy versus 43% (P = .2) for radiation therapy with or without neck dissection.
    • The rate of severe complications was 23% for the surgery group versus 6% for the radiation therapy group (P < .001)
    • The rate of fatal complications was 3.2% for the surgery group versus 0.8% for the radiation therapy group (P < .001).

For patients with early-stage disease, single-modality treatment is preferred. Historically, radiation alone has been preferred, although use of new surgical techniques, including transoral surgery and transoral robotic surgery, is increasing. Nonrandomized comparisons of transoral surgery versus primary radiation therapy suggest superior quality of life (QOL) with minimally invasive surgical techniques.[6] Historically, more–invasive surgical techniques were associated with inferior QOL and greater morbidity.

A prospective multicenter trial (ECOG-3311 [NCT01898494]) evaluating transoral surgery approaches in human papillomavirus−positive oropharyngeal carcinoma with postoperative radiation dose de-escalation is currently under way.

Surgery Followed by Postoperative Radiation Therapy (PORT) With or Without Concurrent Chemotherapy for Patients With Locally Advanced Disease

New surgical techniques for resection and reconstruction that provide access and functional preservation have extended the surgical options for patients with stage III or stage IV oropharyngeal cancer. Specific surgical procedures and their modifications are not described here because of the wide variety of surgical approaches, the variety of opinions about the role of modified neck dissections, and the multiple reconstructive techniques that may give the same results. This group of patients are managed by head and neck surgeons who are skilled in the multiple procedures available and are actively involved in the care of these patients.

Depending on pathological findings after primary surgery, PORT with or without chemotherapy is used in the adjuvant setting for patients with the following histological findings:

  • T4 disease.
  • Perineural invasion.
  • Lymphovascular invasion.
  • Positive margins or margins less than 5 mm.
  • Extracapsular extension of a lymph node.
  • Two or more involved lymph nodes.

The addition of chemotherapy to PORT for oropharyngeal SCC demonstrates a locoregional control and overall survival (OS) benefit compared with radiation therapy alone in patients who have high-risk pathological risk factors, extracapsular extension (ECE) of a lymph node, or positive margins, based on a pooled analysis of the EORTC-22931 (NCT00002555) and RTOG-9501 (NCT00002670) studies.[710][Level of evidence A1]

For patients with intermediate pathological risk factors, the addition of cisplatin chemotherapy given concurrently with PORT is unclear. Intermediate pathological risk factors include:

  • T3 and T4 disease (or stage III and stage IV disease).
  • Perineural infiltration.
  • Vascular embolisms.
  • Clinically enlarged level IV–V lymph nodes secondary to tumors arising in the oral cavity or oropharynx.
  • Two or more histopathologically involved lymph nodes without ECE.
  • Close margins less than 5 mm.

The addition of cetuximab with radiation therapy in the postoperative setting for these intermediate pathological risk factors is being tested in a randomized trial (RTOG-0920 [NCT00956007]).

Radiation Therapy

A review of published clinical results of radiation therapy for head and neck cancer suggested a significant loss of local control when the administration of radiation therapy was prolonged. Therefore, extending standard treatment schedules is detrimental.[11,12]

Patients who are smokers appear to have lower response rates and shorter survival times than those who do not smoke while receiving radiation therapy.[13] Counseling patients to stop smoking before beginning radiation therapy may be beneficial.

Intensity-modulated radiation therapy (IMRT) has become a standard technique for head and neck radiation therapy. IMRT allows a dose-painting technique, also known as a simultaneous-integrated-boost (SIB) technique, with a dose per fraction slightly higher than 2 Gy, which allows slight shortening of overall treatment time and increases the biologically equivalent dose to the tumor.

Evidence (definitive radiation therapy):

  1. IMRT was studied in a phase II trial (RTOG-0022 [NCT00006360]) of 69 patients with stages T1 to T2, N0 to N1, M0 oropharyngeal carcinoma who were treated with primary radiation therapy without chemotherapy.[14] The median follow-up was 2.8 years. The prescribed planning target volume (PTV) dose to the primary tumor and involved nodes was 66 Gy at 2.2 Gy per fraction over 6 weeks. Subclinical PTVs received simultaneously 54 to 60 Gy at 1.8 to 2.0 Gy per fraction using an SIB technique. The following results were observed:
    • The 2-year estimated locoregional failure rate was 9%. Two of four patients (50%), who had major underdose deviations, had locoregional failure compared with 3 of 49 patients (6%) without such deviations (P = .04).
    • Maximal late toxicities with a grade of 2 or higher were skin (12%), mucosa (24%), salivary (67%), esophagus (19%), and osteoradionecrosis (6%).
    • Longer follow-up revealed reduced late toxicity in all categories. Xerostomia grade 2 or higher was observed in 55% of patients at 6 months but was reduced to 25% of patients at 12 months and 16% of patients at 24 months.

    The RTOG-0022 study showed high control rates and the feasibility of IMRT at a multi-institutional level; the study also showed high tumor control rates and reduced salivary toxicity compared with previous Radiation Therapy Oncology Group (RTOG) studies. However, major target underdose deviations were associated with a higher locoregional failure rate.

  2. Similar nonrandomized multicenter studies used fractionally escalated doses, ranging from 2.3 to 2.5 Gy with IMRT. These doses have been safe when given without concurrent chemotherapy for pharyngolaryngeal T2N0, T2N1, or laryngeal T3N0 SCC.[1519]
    • No toxicity difference was observed between the different dose-escalated groups.
  3. A randomized trial (PARSPORT [NCT00081029]) conducted in the United Kingdom compared conventional 3-dimensional conformal radiation therapy with IMRT. The following results were observed:[20][Level of evidence A1]
    • The rate of xerostomia was significantly lower in the IMRT group than in the conventional group.
    • Fatigue was more prevalent in the IMRT group.
    • No significant differences were seen in nonxerostomia late toxicities, locoregional control, or OS at 24 months.

Altered fractionation versus standard fractionation radiation therapy

Radiation therapy alone with altered fractionation may be used for patients with locally advanced oropharyngeal cancer who are not candidates for chemotherapy. Altered fractionated radiation therapy yields a higher locoregional control rate than standard fractionated radiation therapy for patients with stage III or stage IV oropharyngeal cancer.

Evidence (altered fractionation vs. standard fractionation):

  1. The randomized RTOG-9003 trial (NCT00771641) included four radiation therapy treatment arms:[21,22][Level of evidence A1]
    • Standard fractionation (SFX) to 70 Gy in 35 daily fractions for 7 weeks.
    • Hyperfractionation (HFX) to 81.6 Gy in 68 twice-daily fractions for 7 weeks.
    • Accelerated fractionation split course (AFX-S) to 67.2 Gy in 42 fractions for 6 weeks with a 2-week rest after 38.4 Gy.
    • Accelerated concurrent boost fractionation (AFX-C) to 72 Gy in 42 fractions for 6 weeks.

    In a long-term analysis, the three investigational arms were compared with SFX.

    • Only the HFX arm showed superior locoregional control and survival at 5 years compared with the SFX arm (HR, 0.79; 95% CI, 0.62–1.00; P = .05).
    • AFX-C was associated with increased late toxicity compared with SFX.
  2. The following results were shown in a meta-analysis of 15 randomized trials with a total of 6,515 patients and a median follow-up of 6 years involving the assessment of HFX or AFX-S for patients with stage III and stage IV oropharyngeal cancer:[23][Level of evidence A1]
    • There was a significant survival benefit with altered-fractionated radiation therapy and a 3.4% absolute benefit at 5 years (HR, 0.92; 95% CI, 0.86–0.97; P = .003).
    • Altered-fractionation radiation therapy improves locoregional control, with greater benefit shown in younger patients.
    • HFX demonstrated a greater survival benefit (8% at 5 years) than did AFX-S (2% with accelerated fractionation without total dose-reduction and 1.7% with total dose-reduction at 5 years, P = .02).

An additional late effect from radiation therapy is hypothyroidism, which occurs in 30% to 40% of patients who have received external-beam radiation therapy to the entire thyroid gland. Thyroid function testing of patients is considered before therapy and as part of posttreatment follow-up.[24,25]

Prospective data of two randomized controlled trials reported the incidence of hypothyroidism.[26]

  • At a median follow-up of 41 months, 55.1% of the patients developed hypothyroidism (39.3% subclinical, 15.7% biochemical).
  • Patients who underwent IMRT had higher subclinical hypothyroidism (51.1% vs. 27.3%; P = .021), peaking around 1 year after radiation therapy.
  • Younger age, hypopharynx/larynx primary, node positivity, higher dose/fraction (IMRT arm), and D100 were statistically significant factors for developing hypothyroidism.[26][Level of evidence A3]

For patients with well-lateralized oropharyngeal cancer, such as a T1 or T2 tonsil primary tumor with limited extension into the palate or tongue base, and limited ipsilateral lymph node involvement without extracapsular extension, elective treatment to the ipsilateral lymph nodes results in only minimal risk of spread to the contralateral neck.[27] For T3 and T4 tumors that are midline or approach the midline, bilateral nodal treatment is a consideration. In addition to the cervical lymph node chain, retropharyngeal lymph nodes can also be encompassed in the elective nodal treatment.

Concurrent Chemoradiation Therapy

Concurrent chemoradiation therapy is a standard treatment option for patients with locally advanced (stage III and stage IV) oropharyngeal carcinoma and is superior to radiation therapy alone.[28] This treatment approach emphasizes organ preservation and functionality.[29,30]

Evidence (concurrent chemoradiation therapy):

  1. A meta-analysis that originally included 93 randomized prospective head and neck cancer trials published between 1965 and 2000 was updated with the addition of 16 new trials (including 2,767 patients) and 11 updated trials. The results confirmed the benefit and superiority of the addition of concurrent chemotherapy for nonmetastatic head and neck cancer.[31,32][Level of evidence A2]
    • The subset of patients who received chemotherapy and radiation therapy had a 6.5% absolute survival advantage, with OS increasing from 27.7% to 33.6% at 5 years and from 17.3% to 20.9% at 10 years.
    • Patients who received concurrent chemotherapy had a greater survival benefit than did those who received neoadjuvant chemotherapy.
  2. Postoperative chemoradiation therapy with cisplatin 100 mg/m2 given once every 3 weeks is standard treatment for patients with disease at high risk for recurrence, mainly those with extracapsular lymph node extension and positive surgical margins. However, this dosage has raised concerns about insufficient cisplatin delivery because of high-dose–related toxicity. Chemoradiation therapy with weekly cisplatin is widely used as an alternative with a better safety profile.

    A multi-institutional, open-label, noninferiority, phase II/III trial compared different cisplatin schedules as part of postoperative treatment for patients with high-risk locally advanced SCC of the head and neck. Patients received either cisplatin (40 mg/m2) once weekly or standard-dose cisplatin (100 mg/m2) once every 3 weeks, both combined with radiation therapy. OS was the primary end point of the phase III portion of the study. An HR of 1.32 was set as the noninferiority margin. A total of 261 patients were enrolled (cisplatin every 3 weeks, 132 patients; cisplatin weekly, 129 patients).[33]

    • At the planned third interim analysis in the phase III part of the trial, after a median follow-up of 2.2 years, chemoradiation therapy with weekly cisplatin was noninferior to cisplatin every 3 weeks in terms of OS (HR, 0.69; 99.1% CI, 0.374–1.273 [<1.32]; one-sided P for noninferiority = .0027).[33][Level of evidence A1]
    • Grade 3 or 4 neutropenia and infection were less frequent in patients who received cisplatin weekly. Grade 3 or 4 neutropenia occurred in 49% of patients who received cisplatin every 3 weeks and 35% of patients who received cisplatin weekly. Grade 3 or 4 infection occurred in 12% of patients who received cisplatin every 3 weeks and 7% of patients who received cisplatin weekly. Grade 3 or 4 renal impairment and hearing impairment were also less frequent in patients who received cisplatin weekly. No treatment-related deaths were reported among patients who received cisplatin every 3 weeks, and two deaths were reported among patients who received weekly cisplatin (1.6%).

    Regimens with cisplatin given weekly or every 3 weeks are both considered standard care. A large, randomized, prospective trial is evaluating the equivalent efficacy of these regimens.

  3. A phase III randomized trial in India included patients with locally advanced SCC of the head and neck unsuitable for cisplatin-based chemoradiation. This study evaluated using docetaxel as a radiosensitizer. Patients with an Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0 to 2 were randomly assigned (1:1) to receive either radiation therapy alone or with concurrent docetaxel (15 mg/m2) once weekly for a maximum of seven cycles. The primary end point was 2-year disease-free survival (DFS). Most patients (about 60%) received the treatment as definitive. Cisplatin ineligibility was defined by multiple parameters, including an ECOG PS of 2, calculated creatinine clearance <50 mL/min, and organ dysfunction of grade 2 or higher. A total of 356 patients were enrolled (176 in the radiation therapy arm and 180 in the radiation-and-docetaxel arm).[34]
    • The 2-year DFS rate was 30.3% in the radiation-alone arm (95% CI, 23.6%–37.4%) and 42% in the radiation-and-docetaxel arm (95% CI, 34.6%–49.2%) (HR, 0.673; 95% CI, 0.521–0.868; P = .002).
    • The corresponding median OS was 15.3 months in the radiation-alone arm (95% CI, 13.1–22.0) and 25.5 months in the radiation-and-docetaxel arm (95% CI, 17.6–32.5) (log-rank P = .035). The 2-year OS rate was 41.7% in the radiation-alone arm (95% CI, 34.1%–49.1%) and 50.8% in the radiation-and-docetaxel arm (95% CI, 43.1%–58.1%) (HR, 0.747; 95% CI, 0.569–0.980; P = .035).[34][Level of evidence A1]
    • There was a higher incidence of grade 3 or higher mucositis (22.2% vs. 49.7%; P < .001), odynophagia (33.5% vs. 52.5%; P < .001), and dysphagia (33% vs. 49.7%; P = .002) for patients who received radiation and docetaxel versus radiation alone.
    • The increase of toxicity observed in the radiation-and-docetaxel arm did not significantly impair the total radiation dose the patients received. A total of 88.9% of patients in the radiation-and-docetaxel arm and 93.8% of patients in the radiation-alone arm received the total radiation dose. In the radiation-and-docetaxel arm, 85.6% of patients received five or more cycles of chemotherapy.
  4. In a randomized trial of patients with locally advanced head and neck cancer, curative-intent radiation therapy alone (213 patients) was compared with radiation therapy plus weekly cetuximab (211 patients).[35] The initial dose of cetuximab was 400 mg/m2 of body-surface area 1 week before radiation therapy was started, followed by a weekly dose of 250 mg/m2 of body-surface area for the duration of the radiation therapy. This study allowed altered-fractionation regimens to be used in both arms.[35,36][Level of evidence A1]
    • At a median follow-up of 54 months, patients treated with cetuximab and radiation therapy demonstrated significantly higher progression-free survival (PFS) (HRdeath or for disease progression, 0.70; P = .006).
    • Patients in the cetuximab arm experienced higher rates of acneiform rash and infusion reactions, although the incidence of other grade 3 or higher toxicities, including mucositis, did not differ significantly between the two groups.

Cetuximab versus cisplatin in patients with human papillomavirus (HPV)-positive oropharyngeal cancer

Studies evaluating de-intensification using reduced-dose radiation therapy (NRG-HN002 [NCT02254278] and ECOG-3311 [NCT01898494]) are ongoing in patients with low-risk HPV-positive oropharyngeal cancer. Cetuximab, an epidermal growth factor receptor inhibitor, has been evaluated in two randomized trials as a proposed de-intensification strategy to reduce the toxicity of cisplatin-based treatment.

Evidence (cetuximab versus cisplatin in patients with HPV-positive oropharyngeal cancer):

  1. In the randomized RTOG-1016 trial (NCT01302834), patients with HPV-positive (determined by central confirmation of p16 immunohistochemistry) oropharyngeal cancer were randomly assigned (1:1) to receive either radiation therapy with cetuximab or radiation therapy with cisplatin. This trial aimed to determine whether treatment with radiation therapy and cetuximab produced noninferior survival compared with treatment using radiation therapy and cisplatin. Of the 987 patients enrolled, 849 were randomly assigned to receive radiation therapy plus cetuximab (n = 425) or radiation therapy plus cisplatin (n = 424). Subsequently, 399 patients assigned to receive cetuximab and 406 patients assigned to receive cisplatin were determined to be eligible. Patients received 70 Gy of radiation therapy in 6 weeks accelerated (six fractions/week) with either two cycles of cisplatin (100 mg/m2) every 3 weeks or weekly cetuximab.[37][Level of evidence A1]
    • After a median follow-up duration of 4.5 years, radiation therapy plus cetuximab did not meet the noninferiority criteria for OS (HR, 1.45; one-sided 95% upper CI, 1.94; P = .5056 for noninferiority; one-sided log-rank P = .0163).[38][Level of evidence A1]
    • The estimated 5-year OS rate was 77.9% (95% CI, 73.4%–82.5%) in the cetuximab group versus 84.6% (80.6%–88.6%) in the cisplatin group.
    • PFS was significantly lower in the cetuximab group than in the cisplatin group (HR, 1.72; 95% CI, 1.29–2.29; P = .0002; 5-year PFS rate 67.3%; 95% CI, 62.4%–72.2% vs. 78.4%, 73.8%–83.0%), and locoregional failure was significantly higher in the cetuximab group than in the cisplatin group.
    • Acute moderate to severe toxicity (77.4%, 95% CI, 73.0%–81.5% vs. 81.7%, 77.5%–85.3%; P = .1586) and late moderate to severe toxicity (16.5%, 95% CI, 12.9%–20.7% vs. 20.4%, 16.4%–24.8%; P = .1904) were similar between the cetuximab and cisplatin groups.
  2. De-ESCALaTE HPV [NCT01874171] was an open-label, randomized, controlled, phase III trial at 32 head and neck treatment centers in Ireland, the Netherlands, and the United Kingdom. It included patients aged 18 years or older with HPV-positive, low-risk oropharyngeal cancer (nonsmokers or lifetime smokers with a smoking history of <10 pack-years).[39] Patients were randomly assigned (1:1) to receive, in addition to radiation therapy (70 Gy in 35 fractions), either intravenous cisplatin (100 mg/m2 on days 1, 22, and 43 of radiation therapy) or intravenous cetuximab (400 mg/m2 loading dose followed by seven weekly infusions of 250 mg/m2).

    The primary outcome was overall severe (grades 3–5) toxicity events at 24 months from the end of treatment. The primary outcome was assessed by intention-to-treat and per-protocol analyses. Between Nov 12, 2012, and Oct 1, 2016, 334 patients were recruited (166 in the cisplatin group and 168 in the cetuximab group).

    • Overall (acute and late) severe (grades 3–5) toxicity did not differ significantly between treatment groups at 24 months (mean number of events per patient, 4.8 [95% CI, 4.2–5.4] with cisplatin vs. 4.8 [4.2–5.4] with cetuximab; P = .98).[39][Level of evidence A1]
    • At 24 months, overall all-grade toxicity did not differ significantly (mean number of events per patient, 29.2 [95% CI, 27.3–31.0] with cisplatin vs. 30.1 [28.3–31.9] with cetuximab; P = .49).
    • OS was inferior and local recurrence rate was higher in the cetuximab arm. The 2-year OS rate was 97.5% for the cisplatin group versus 89.4% for the cetuximab group (HR, 5.0; 95% CI, 1.7–14.7; P = .001), and the 2-year recurrence rate was 6.0% for the cisplatin group versus 16.1% for the cetuximab group (HR, 3.4; 1.6–7.2; P = .0007).

These findings showed the inferiority of cetuximab compared with cisplatin for OS and local recurrence rates for patients with locoregionally advanced HPV-related oropharyngeal cancer and also did not demonstrate reduced toxicity with cetuximab and radiation therapy compared with cisplatin. Treatment with the combination of radiation therapy and cetuximab resulted in inferior OS and PFS compared with treatment using radiation therapy and cisplatin; therefore, treatment with radiation therapy and cisplatin remains the standard of care.

For more information about oral toxicities, see Oral Complications of Cancer Therapies.

Neoadjuvant Chemotherapy Followed by Concurrent Chemoradiation Therapy

In a meta-analysis of five randomized trials, a total of 1,022 patients with locally advanced head and neck SCC were assigned to receive either neoadjuvant chemotherapy with TPF (docetaxel, cisplatin, and fluorouracil) followed by concurrent chemoradiation therapy or concurrent chemoradiation therapy alone. The analysis failed to show an OS (HR, 1.01; 95% confidence limits [CLs], 0.84, 1.21; P = .92) or PFS (HR, 0.91; 95% CLs, 0.75, 1.1; P = .32) advantage for neoadjuvant chemotherapy using the TPF regimen over concurrent chemoradiation therapy alone.[40][Level of evidence A1]

Evidence (neoadjuvant chemotherapy followed by concurrent chemoradiation therapy):

  1. In a phase II study of neoadjuvant chemotherapy using cisplatin, paclitaxel, and cetuximab in patients with HPV-associated oropharyngeal SCC (ECOG 1308 [NCT01084083]), patients who achieved a complete clinical response to three cycles of neoadjuvant chemotherapy received reduced-dose IMRT of 54 Gy with weekly cetuximab. Patients with less than a clinical complete response received 69.3 Gy of radiation to the primary site or nodes and cetuximab.[41]
    • With a median follow-up of 35.4 months, the 2-year PFS rate was 80% and the OS rate was 94% for patients who achieved a complete clinical response and were treated with 54 Gy of radiation.
    • For patients whose primary tumor (T) was less than T4 and regional lymph nodes (N) were less than N2c and had a smoking history of less than 10 pack-years, the 2-year PFS rate was 96% and the OS rate was 96%.
    • Lower-dose radiation using 54 Gy was associated with lower rates of dysphagia with solid foods and less-impaired nutrition.
    • There does not appear to be a benefit from treatment with neoadjuvant chemotherapy followed by full-dose (≥70 Gy) concurrent chemoradiation therapy. There may be a role for radiation dose de-escalation in HPV-positive patients with low-risk disease who achieve a complete clinical response after neoadjuvant chemotherapy.

Overall, the role of neoadjuvant chemotherapy for patients with oropharyngeal cancer remains unclear. However, in HPV-defined subsets, more information is needed because, as this phase II study suggests, in that setting, neoadjuvant chemotherapy may be used with less chemoradiation.[40,4245][Level of evidence A1]

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.[46,47] 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.[4648] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[4951] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[52] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[53]

References
  1. Iyer NG, Tan DS, Tan VK, et al.: Randomized trial comparing surgery and adjuvant radiotherapy versus concurrent chemoradiotherapy in patients with advanced, nonmetastatic squamous cell carcinoma of the head and neck: 10-year update and subset analysis. Cancer 121 (10): 1599-607, 2015. [PUBMED Abstract]
  2. Mehanna H, Wong WL, McConkey CC, et al.: PET-CT Surveillance versus Neck Dissection in Advanced Head and Neck Cancer. N Engl J Med 374 (15): 1444-54, 2016. [PUBMED Abstract]
  3. Parsons JT, Mendenhall WM, Stringer SP, et al.: Squamous cell carcinoma of the oropharynx: surgery, radiation therapy, or both. Cancer 94 (11): 2967-80, 2002. [PUBMED Abstract]
  4. Harrison LB, Zelefsky MJ, Armstrong JG, et al.: Performance status after treatment for squamous cell cancer of the base of tongue–a comparison of primary radiation therapy versus primary surgery. Int J Radiat Oncol Biol Phys 30 (4): 953-7, 1994. [PUBMED Abstract]
  5. Mendenhall WM, Morris CG, Amdur RJ, et al.: Definitive radiotherapy for squamous cell carcinoma of the base of tongue. Am J Clin Oncol 29 (1): 32-9, 2006. [PUBMED Abstract]
  6. Chen AM, Daly ME, Luu Q, et al.: Comparison of functional outcomes and quality of life between transoral surgery and definitive chemoradiotherapy for oropharyngeal cancer. Head Neck 37 (3): 381-5, 2015. [PUBMED Abstract]
  7. Cooper JS, Pajak TF, Forastiere AA, et al.: Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med 350 (19): 1937-44, 2004. [PUBMED Abstract]
  8. Bernier J, Domenge C, Ozsahin M, et al.: Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Engl J Med 350 (19): 1945-52, 2004. [PUBMED Abstract]
  9. Bernier J, Cooper JS, Pajak TF, et al.: Defining risk levels in locally advanced head and neck cancers: a comparative analysis of concurrent postoperative radiation plus chemotherapy trials of the EORTC (#22931) and RTOG (# 9501). Head Neck 27 (10): 843-50, 2005. [PUBMED Abstract]
  10. Cooper JS, Zhang Q, Pajak TF, et al.: Long-term follow-up of the RTOG 9501/intergroup phase III trial: postoperative concurrent radiation therapy and chemotherapy in high-risk squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys 84 (5): 1198-205, 2012. [PUBMED Abstract]
  11. Fowler JF, Lindstrom MJ: Loss of local control with prolongation in radiotherapy. Int J Radiat Oncol Biol Phys 23 (2): 457-67, 1992. [PUBMED Abstract]
  12. Allal AS, de Pree C, Dulguerov P, et al.: Avoidance of treatment interruption: an unrecognized benefit of accelerated radiotherapy in oropharyngeal carcinomas? Int J Radiat Oncol Biol Phys 45 (1): 41-5, 1999. [PUBMED Abstract]
  13. Browman GP, Wong G, Hodson I, et al.: Influence of cigarette smoking on the efficacy of radiation therapy in head and neck cancer. N Engl J Med 328 (3): 159-63, 1993. [PUBMED Abstract]
  14. Eisbruch A, Harris J, Garden AS, et al.: Multi-institutional trial of accelerated hypofractionated intensity-modulated radiation therapy for early-stage oropharyngeal cancer (RTOG 00-22). Int J Radiat Oncol Biol Phys 76 (5): 1333-8, 2010. [PUBMED Abstract]
  15. Leclerc M, Maingon P, Hamoir M, et al.: A dose escalation study with intensity modulated radiation therapy (IMRT) in T2N0, T2N1, T3N0 squamous cell carcinomas (SCC) of the oropharynx, larynx and hypopharynx using a simultaneous integrated boost (SIB) approach. Radiother Oncol 106 (3): 333-40, 2013. [PUBMED Abstract]
  16. Buettner F, Miah AB, Gulliford SL, et al.: Novel approaches to improve the therapeutic index of head and neck radiotherapy: an analysis of data from the PARSPORT randomised phase III trial. Radiother Oncol 103 (1): 82-7, 2012. [PUBMED Abstract]
  17. Gulliford SL, Miah AB, Brennan S, et al.: Dosimetric explanations of fatigue in head and neck radiotherapy: an analysis from the PARSPORT Phase III trial. Radiother Oncol 104 (2): 205-12, 2012. [PUBMED Abstract]
  18. Kohler RE, Sheets NC, Wheeler SB, et al.: Two-year and lifetime cost-effectiveness of intensity modulated radiation therapy versus 3-dimensional conformal radiation therapy for head-and-neck cancer. Int J Radiat Oncol Biol Phys 87 (4): 683-9, 2013. [PUBMED Abstract]
  19. Gupta T, Agarwal J, Jain S, et al.: Three-dimensional conformal radiotherapy (3D-CRT) versus intensity modulated radiation therapy (IMRT) in squamous cell carcinoma of the head and neck: a randomized controlled trial. Radiother Oncol 104 (3): 343-8, 2012. [PUBMED Abstract]
  20. Nutting CM, Morden JP, Harrington KJ, et al.: Parotid-sparing intensity modulated versus conventional radiotherapy in head and neck cancer (PARSPORT): a phase 3 multicentre randomised controlled trial. Lancet Oncol 12 (2): 127-36, 2011. [PUBMED Abstract]
  21. Fu KK, Pajak TF, Trotti A, et al.: A Radiation Therapy Oncology Group (RTOG) phase III randomized study to compare hyperfractionation and two variants of accelerated fractionation to standard fractionation radiotherapy for head and neck squamous cell carcinomas: first report of RTOG 9003. Int J Radiat Oncol Biol Phys 48 (1): 7-16, 2000. [PUBMED Abstract]
  22. Beitler JJ, Zhang Q, Fu KK, et al.: Final results of local-regional control and late toxicity of RTOG 9003: a randomized trial of altered fractionation radiation for locally advanced head and neck cancer. Int J Radiat Oncol Biol Phys 89 (1): 13-20, 2014. [PUBMED Abstract]
  23. Baujat B, Bourhis J, Blanchard P, et al.: Hyperfractionated or accelerated radiotherapy for head and neck cancer. Cochrane Database Syst Rev (12): CD002026, 2010. [PUBMED Abstract]
  24. Turner SL, Tiver KW, Boyages SC: Thyroid dysfunction following radiotherapy for head and neck cancer. Int J Radiat Oncol Biol Phys 31 (2): 279-83, 1995. [PUBMED Abstract]
  25. Constine LS: What else don’t we know about the late effects of radiation in patients treated for head and neck cancer? Int J Radiat Oncol Biol Phys 31 (2): 427-9, 1995. [PUBMED Abstract]
  26. Murthy V, Narang K, Ghosh-Laskar S, et al.: Hypothyroidism after 3-dimensional conformal radiotherapy and intensity-modulated radiotherapy for head and neck cancers: prospective data from 2 randomized controlled trials. Head Neck 36 (11): 1573-80, 2014. [PUBMED Abstract]
  27. O’Sullivan B, Warde P, Grice B, et al.: The benefits and pitfalls of ipsilateral radiotherapy in carcinoma of the tonsillar region. Int J Radiat Oncol Biol Phys 51 (2): 332-43, 2001. [PUBMED Abstract]
  28. Denis F, Garaud P, Bardet E, et al.: Final results of the 94-01 French Head and Neck Oncology and Radiotherapy Group randomized trial comparing radiotherapy alone with concomitant radiochemotherapy in advanced-stage oropharynx carcinoma. J Clin Oncol 22 (1): 69-76, 2004. [PUBMED Abstract]
  29. Mendenhall WM, Werning JW, Pfister DG: Treatment of head and neck cancer. In: DeVita VT Jr, Lawrence TS, Rosenberg SA: Cancer: Principles and Practice of Oncology. 9th ed. Lippincott Williams & Wilkins, 2011, pp 729-80.
  30. Adelstein DJ: Oropharyngeal cancer: the role of chemotherapy. Curr Treat Options Oncol 4 (1): 3-13, 2003. [PUBMED Abstract]
  31. Pignon JP, le Maître A, Maillard E, et al.: Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): an update on 93 randomised trials and 17,346 patients. Radiother Oncol 92 (1): 4-14, 2009. [PUBMED Abstract]
  32. Lacas B, Carmel A, Landais C, et al.: Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): An update on 107 randomized trials and 19,805 patients, on behalf of MACH-NC Group. Radiother Oncol 156: 281-293, 2021. [PUBMED Abstract]
  33. Kiyota N, Tahara M, Mizusawa J, et al.: Weekly Cisplatin Plus Radiation for Postoperative Head and Neck Cancer (JCOG1008): A Multicenter, Noninferiority, Phase II/III Randomized Controlled Trial. J Clin Oncol 40 (18): 1980-1990, 2022. [PUBMED Abstract]
  34. Patil VM, Noronha V, Menon N, et al.: Results of Phase III Randomized Trial for Use of Docetaxel as a Radiosensitizer in Patients With Head and Neck Cancer, Unsuitable for Cisplatin-Based Chemoradiation. J Clin Oncol 41 (13): 2350-2361, 2023. [PUBMED Abstract]
  35. Bonner JA, Harari PM, Giralt J, et al.: Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 354 (6): 567-78, 2006. [PUBMED Abstract]
  36. Curran D, Giralt J, Harari PM, et al.: Quality of life in head and neck cancer patients after treatment with high-dose radiotherapy alone or in combination with cetuximab. J Clin Oncol 25 (16): 2191-7, 2007. [PUBMED Abstract]
  37. Trotti A, Harris J, Gillison M, et al.: NRG-RTOG 1016: phase III trial comparing radiation/cetuximab to radiation/cisplatin in HPV-related cancer of the oropharynx. [Abstract] Int J Radiat Oncol Biol Phys 102 (5): A-LBA4, 1604-5, 2018. Also available online. Last accessed November 18, 2024.
  38. Gillison ML, Trotti AM, Harris J, et al.: Radiotherapy plus cetuximab or cisplatin in human papillomavirus-positive oropharyngeal cancer (NRG Oncology RTOG 1016): a randomised, multicentre, non-inferiority trial. Lancet 393 (10166): 40-50, 2019. [PUBMED Abstract]
  39. Mehanna H, Robinson M, Hartley A, et al.: Radiotherapy plus cisplatin or cetuximab in low-risk human papillomavirus-positive oropharyngeal cancer (De-ESCALaTE HPV): an open-label randomised controlled phase 3 trial. Lancet 393 (10166): 51-60, 2019. [PUBMED Abstract]
  40. Budach W, Bölke E, Kammers K, et al.: Induction chemotherapy followed by concurrent radio-chemotherapy versus concurrent radio-chemotherapy alone as treatment of locally advanced squamous cell carcinoma of the head and neck (HNSCC): A meta-analysis of randomized trials. Radiother Oncol 118 (2): 238-43, 2016. [PUBMED Abstract]
  41. Marur S, Li S, Cmelak AJ, et al.: E1308: Phase II Trial of Induction Chemotherapy Followed by Reduced-Dose Radiation and Weekly Cetuximab in Patients With HPV-Associated Resectable Squamous Cell Carcinoma of the Oropharynx- ECOG-ACRIN Cancer Research Group. J Clin Oncol 35 (5): 490-497, 2017. [PUBMED Abstract]
  42. Haddad R, O’Neill A, Rabinowits G, et al.: Induction chemotherapy followed by concurrent chemoradiotherapy (sequential chemoradiotherapy) versus concurrent chemoradiotherapy alone in locally advanced head and neck cancer (PARADIGM): a randomised phase 3 trial. Lancet Oncol 14 (3): 257-64, 2013. [PUBMED Abstract]
  43. Cohen EE, Karrison TG, Kocherginsky M, et al.: Phase III randomized trial of induction chemotherapy in patients with N2 or N3 locally advanced head and neck cancer. J Clin Oncol 32 (25): 2735-43, 2014. [PUBMED Abstract]
  44. Hitt R, Grau JJ, López-Pousa A, et al.: A randomized phase III trial comparing induction chemotherapy followed by chemoradiotherapy versus chemoradiotherapy alone as treatment of unresectable head and neck cancer. Ann Oncol 25 (1): 216-25, 2014. [PUBMED Abstract]
  45. Driessen CM, de Boer JP, Gelderblom H, et al.: Induction chemotherapy with docetaxel/cisplatin/5-fluorouracil followed by randomization to two cisplatin-based concomitant chemoradiotherapy schedules in patients with locally advanced head and neck cancer (CONDOR study) (Dutch Head and Neck Society 08-01): A randomized phase II study. Eur J Cancer 52: 77-84, 2016. [PUBMED Abstract]
  46. 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]
  47. Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
  48. 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]
  49. 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]
  50. 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]
  51. 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]
  52. 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]
  53. 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 I and Stage II Oropharyngeal Cancer

Treatment Options for Stage I and Stage II Oropharyngeal Cancer

The management of stage I and stage II carcinomas of the oropharynx requires multidisciplinary input to establish the optimal treatment. Radiation therapy or surgery is equally successful in controlling stage I and stage II oropharyngeal cancer. For more information, see the Treatment Option Overview for Oropharyngeal Cancer section.

The choice of treatment is dictated by the anticipated functional speech and swallowing and cosmetic outcome of the treatment options and by the available expertise of the surgeon or radiation oncologist. Treatment is individualized for each patient.

Treatment options for stage I and stage II oropharyngeal cancer include:

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 Stage III and Stage IV Oropharyngeal Cancer

The management of stage III and stage IV carcinomas of the oropharynx is complex and requires multidisciplinary input to establish the optimal treatment. For more information, see the Treatment Option Overview for Oropharyngeal Cancer section.

Treatment Options for Stage III and Stage IV Oropharyngeal Cancer

Treatment options for stage III and stage IV oropharyngeal cancer include:

  1. Surgery and postoperative radiation therapy (PORT) with or without chemotherapy for patients with advanced disease.[14][Level of evidence A1]
  2. Radiation therapy using altered fractionation.[59][Level of evidence A1]
  3. Concurrent chemoradiation therapy.[1015][Level of evidence A1]
  4. Neoadjuvant chemotherapy followed by concurrent chemoradiation therapy.
  5. Chemoradiation therapy with immunotherapy (under clinical evaluation). RTOG 3504 (NCT02764593) evaluated concurrent chemoradiation therapy with nivolumab in patients with intermediate- to high-risk head and neck cancer.
  6. Treatment de-intensification using radiation dose de-escalation is being studied in NRG-HN002 (NCT02254278).
  7. Treatment de-intensification using transoral surgery followed by radiation dose de-escalation is being studied in ECOG-3311 (NCT01898494).

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
  1. Bernier J, Cooper JS, Pajak TF, et al.: Defining risk levels in locally advanced head and neck cancers: a comparative analysis of concurrent postoperative radiation plus chemotherapy trials of the EORTC (#22931) and RTOG (# 9501). Head Neck 27 (10): 843-50, 2005. [PUBMED Abstract]
  2. Cooper JS, Zhang Q, Pajak TF, et al.: Long-term follow-up of the RTOG 9501/intergroup phase III trial: postoperative concurrent radiation therapy and chemotherapy in high-risk squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys 84 (5): 1198-205, 2012. [PUBMED Abstract]
  3. Cooper JS, Pajak TF, Forastiere AA, et al.: Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med 350 (19): 1937-44, 2004. [PUBMED Abstract]
  4. Bernier J, Domenge C, Ozsahin M, et al.: Postoperative irradiation with or without concomitant chemotherapy for locally advanced head and neck cancer. N Engl J Med 350 (19): 1945-52, 2004. [PUBMED Abstract]
  5. Horiot JC, Le Fur R, N’Guyen T, et al.: Hyperfractionation versus conventional fractionation in oropharyngeal carcinoma: final analysis of a randomized trial of the EORTC cooperative group of radiotherapy. Radiother Oncol 25 (4): 231-41, 1992. [PUBMED Abstract]
  6. Bourhis J, Lapeyre M, Tortochaux J, et al.: Phase III randomized trial of very accelerated radiation therapy compared with conventional radiation therapy in squamous cell head and neck cancer: a GORTEC trial. J Clin Oncol 24 (18): 2873-8, 2006. [PUBMED Abstract]
  7. Overgaard J, Hansen HS, Specht L, et al.: Five compared with six fractions per week of conventional radiotherapy of squamous-cell carcinoma of head and neck: DAHANCA 6 and 7 randomised controlled trial. Lancet 362 (9388): 933-40, 2003. [PUBMED Abstract]
  8. Overgaard J, Mohanti BK, Begum N, et al.: Five versus six fractions of radiotherapy per week for squamous-cell carcinoma of the head and neck (IAEA-ACC study): a randomised, multicentre trial. Lancet Oncol 11 (6): 553-60, 2010. [PUBMED Abstract]
  9. Fu KK, Pajak TF, Trotti A, et al.: A Radiation Therapy Oncology Group (RTOG) phase III randomized study to compare hyperfractionation and two variants of accelerated fractionation to standard fractionation radiotherapy for head and neck squamous cell carcinomas: first report of RTOG 9003. Int J Radiat Oncol Biol Phys 48 (1): 7-16, 2000. [PUBMED Abstract]
  10. Bonner JA, Harari PM, Giralt J, et al.: Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 354 (6): 567-78, 2006. [PUBMED Abstract]
  11. Curran D, Giralt J, Harari PM, et al.: Quality of life in head and neck cancer patients after treatment with high-dose radiotherapy alone or in combination with cetuximab. J Clin Oncol 25 (16): 2191-7, 2007. [PUBMED Abstract]
  12. Denis F, Garaud P, Bardet E, et al.: Final results of the 94-01 French Head and Neck Oncology and Radiotherapy Group randomized trial comparing radiotherapy alone with concomitant radiochemotherapy in advanced-stage oropharynx carcinoma. J Clin Oncol 22 (1): 69-76, 2004. [PUBMED Abstract]
  13. Olmi P, Crispino S, Fallai C, et al.: Locoregionally advanced carcinoma of the oropharynx: conventional radiotherapy vs. accelerated hyperfractionated radiotherapy vs. concomitant radiotherapy and chemotherapy–a multicenter randomized trial. Int J Radiat Oncol Biol Phys 55 (1): 78-92, 2003. [PUBMED Abstract]
  14. Semrau R, Mueller RP, Stuetzer H, et al.: Efficacy of intensified hyperfractionated and accelerated radiotherapy and concurrent chemotherapy with carboplatin and 5-fluorouracil: updated results of a randomized multicentric trial in advanced head-and-neck cancer. Int J Radiat Oncol Biol Phys 64 (5): 1308-16, 2006. [PUBMED Abstract]
  15. Pignon JP, le Maître A, Maillard E, et al.: Meta-analysis of chemotherapy in head and neck cancer (MACH-NC): an update on 93 randomised trials and 17,346 patients. Radiother Oncol 92 (1): 4-14, 2009. [PUBMED Abstract]

Treatment of Metastatic and Recurrent Oropharyngeal Cancer

Treatment Options for Metastatic and Recurrent Oropharyngeal Cancer

The management of metastatic and recurrent carcinomas of the oropharynx is complex and requires multidisciplinary input to establish the optimal treatment. For more information, see the Treatment Option Overview for Oropharyngeal Cancer section.

Treatment options for metastatic and recurrent oropharyngeal cancer include:

  1. Surgical resection, if technically feasible and the tumor does not respond to radiation therapy.[1]
  2. Radiation therapy, if the tumor is not completely removed by surgery and curative doses of radiation have not been given previously.[2]
  3. A second surgery, if the tumor was not completely removed initially and if technically feasible.[1]
  4. Chemotherapy, for unresectable locoregionally recurrent disease.
  5. Additional radiation therapy using conventionally fractionated radiation therapy, or hyperfractionated radiation therapy (HFX) with concurrent chemotherapy.[3]
  6. Stereotactic body radiation therapy with concurrent cetuximab.[4]
  7. Immunotherapy (inhibitor of the programmed death-ligand 1 [PD-L1] pathway) can be used after platinum-based chemotherapy failure [5,6] or up front in patients with metastatic or locally recurrent disease.[7,8]
  8. Clinical trials evaluating additional radiation therapy using HFX with concurrent chemotherapy, targeted therapy, stereotactic body radiation therapy, or immunotherapy.[9]

Chemotherapy

Platinum-based chemotherapy is often used as first-line treatment for patients with metastatic or recurrent squamous cell carcinoma (SCC) of the head and neck.

Evidence (chemotherapy):

  1. In a phase III randomized trial of 442 patients with untreated metastatic or recurrent SCC of the head and neck, adding cetuximab to platinum plus fluorouracil (5-FU) improved overall survival (OS), compared with platinum plus 5-FU alone. The median survival was 10.1 months versus 7.4 months (hazard ratio [HR]death, 0.80; 95% confidence interval [CI], 0.64–0.99; P = .04).[10]
    • Quality of life (QOL) was not adversely affected by adding cetuximab to this platinum-based regimen.[11]

    Tumor EGFR gene copy number was not a predictive biomarker for the efficacy of cetuximab plus platinum and 5-FU as first-line therapy for patients with metastatic or recurrent SCC of the head and neck.[12][Level of evidence A1]

  2. An open-label, phase III, randomized trial demonstrated improved progression-free survival (PFS) for patients who received afatinib compared with patients who received methotrexate.[13]
    1. After a median follow-up of 6.7 months, the median PFS was 2.6 months (95% CI, 2.0–2.7) for the afatinib group and 1.7 months (95% CI, 1.5–2.4) for the methotrexate group (HR, 0.80; 95% CI, 0.65–0.98; P = .030).
    2. The most frequent grade 3 or grade 4 drug-related adverse events for patients treated with afatinib or methotrexate included:
      • Rash or acne (10% for afatinib vs. 0% for methotrexate).
      • Diarrhea (9% for afatinib vs. 2% for methotrexate).
      • Stomatitis (6% for afatinib vs. 8% for methotrexate).
      • Fatigue (6% for afatinib vs. 3% for methotrexate).
      • Neutropenia (<1% for afatinib vs. 7% for methotrexate).
    3. Overall, serious adverse events occurred in 14% of patients treated with afatinib and 11% of patients treated with methotrexate.

Immunotherapy

Pembrolizumab

Pembrolizumab is a monoclonal antibody and an inhibitor of the programmed death-1 (PD-1) pathway. Studies have evaluated pembrolizumab in patients with incurable metastatic or recurrent head and neck squamous cell carcinoma (SCC).

Evidence (pembrolizumab as first-line therapy):

  1. KEYNOTE-048 (NCT02358031) was a nonblinded, randomized, phase III study of participants with untreated locally incurable metastatic or recurrent head and neck SCC that was performed at 200 sites in 37 countries.[7] A total of 882 patients were randomly assigned in a 1:1:1 ratio to receive pembrolizumab alone (n = 301), pembrolizumab plus a platinum and fluorouracil (5-FU) (pembrolizumab with chemotherapy) (n = 281), or cetuximab plus a platinum and 5-FU (cetuximab with chemotherapy) (n = 300). Investigators, patients, and representatives of the sponsor were masked to the programmed death-ligand 1 (PD-L1) combined positive score (CPS) results; PD-L1 positivity was not required for study entry. A total of 754 patients (85%) had a CPS of 1 or higher and 381 patients (43%) had a CPS of 20 or higher.

    The primary end points were overall survival (OS) and progression-free survival (PFS). Progression was defined as radiographically confirmed disease progression or death from any cause, whichever came first, in the intention-to-treat population.

    1. At the second interim analysis, pembrolizumab alone showed improved or noninferior OS compared with cetuximab with chemotherapy. The median OS results were reported as follows:[7][Level of evidence A1]
      • Among the population with a CPS of 20 or higher, the median OS was 14.9 months in patients who received pembrolizumab alone and 10.7 months in patients who received cetuximab with chemotherapy (hazard ratio [HR], 0.61; 95% confidence interval [CI], 0.45–0.83; P = .0007).
      • Among the population with a CPS of 1 or higher, the median OS was 12.3 months in patients who received pembrolizumab alone and 10.3 months in patients who received cetuximab with chemotherapy (HR, 0.78; 95% CI, 0.64–0.96; P = .0086).
      • Among the total population, patients who received pembrolizumab alone had noninferior OS (11.6 months) compared with patients who received cetuximab with chemotherapy (10.7 months) (HR, 0.85; 95% CI, 0.71–1.03; P = .0456).
    2. Pembrolizumab with chemotherapy showed improved OS versus cetuximab with chemotherapy. The OS results were reported as follows:
      • At the second interim analysis, among the total population, the median OS was 13.0 months in patients who received pembrolizumab with chemotherapy and 10.7 months in patients who received cetuximab with chemotherapy (HR, 0.77; 95% CI, 0.63–0.93; P = .0034).
      • At the final analysis, among the population with a CPS of 20 or higher, the median OS was 14.7 months in patients who received pembrolizumab with chemotherapy and 11.0 months in patients who received cetuximab with chemotherapy (HR, 0.60; 95% CI, 0.45–0.82; P = .0004).
      • At the final analysis, among the population with a CPS of 1 or higher, the median OS was 13.6 months in patients who received pembrolizumab with chemotherapy and 10.4 months in patients who received cetuximab with chemotherapy (HR, 0.65; 95% CI, 0.53–0.80; P < .0001).
    3. At the second interim analysis, neither pembrolizumab alone nor pembrolizumab with chemotherapy improved PFS.
    4. At the final analysis, grade 3 or higher all-cause adverse events occurred in 164 of 300 patients (55%) in the pembrolizumab-alone group, 235 of 276 patients (85%) who received pembrolizumab with chemotherapy, and 239 of 287 patients (83%) who received cetuximab with chemotherapy.
    5. Adverse events led to death in 25 patients (8%) in the pembrolizumab-alone group, 32 patients (12%) who received pembrolizumab with chemotherapy, and 28 patients (10%) who received cetuximab with chemotherapy.

Pembrolizumab plus a platinum and 5-FU is an appropriate first-line treatment for patients with metastatic or recurrent head and neck SCC. Pembrolizumab monotherapy is an appropriate first-line treatment for patients with PD-L1–positive metastatic or recurrent head and neck SCC. These results were confirmed at a longer median follow-up of 45 months (interquartile range, 41.0–49.2).[8]

Evidence (pembrolizumab after progression on platinum-based treatment):

  1. The phase III KEYNOTE-040 (NCT02252042) trial included patients with incurable metastatic or recurrent head and neck SCC who had received platinum-based treatment within 3 to 6 months.[5] Patients were randomly assigned to the pembrolizumab arm (200 mg every 3 weeks [247 patients]) or to the standard therapy arm of the investigator’s choice (methotrexate, docetaxel, or cetuximab [248 patients]). Patients received treatment until progression or toxicity. The maximum duration of pembrolizumab was 24 months. The primary end point was OS in the intention-to-treat population.
    • The median OS was 8.4 months in the pembrolizumab arm and 6.9 months in the standard therapy arm (HR, 0.80; 95% CI, 0.65–0.98; nominal P = .0161).[5][Level of evidence A1]
    • Pembrolizumab was associated with fewer grade 3 or higher adverse events (pembrolizumab, 13% vs. standard therapy, 36%). The most common treatment-related adverse events were hypothyroidism (13%) in the pembrolizumab arm and fatigue (18%) in the standard therapy arm.
    • In patients who received pembrolizumab, there were four treatment-related deaths resulting from large intestinal perforation, Stevens-Johnson syndrome, and unspecified malignant progression. Two treatment-related deaths in the standard therapy arm resulted from malignant progression and pneumonia.
    • The PD-L1 CPS was 1 or higher in 79% of the patients in the pembrolizumab arm and 77% of the patients in the standard therapy arm.
    • Compared with patients treated with standard therapy, a reduced HRdeath was noted for patients who received pembrolizumab and had PD-1 expression on their tumors or in the tumor microenvironment as noted by a PD-L1 CPS of 1 or higher (HR, 0.74; 95% CI, 0.58–0.93; nominal P = .0049) or a PD-L1 tumor proportion score of 50% or higher (HR, 0.53; 95% CI, 0.35–0.81; nominal P = .0014).
Nivolumab

Nivolumab is a fully human immunoglobulin G4 anti–PD-1 monoclonal antibody.

Evidence (nivolumab combined with ipilimumab in patients who have not previously received systemic therapy):

  1. The CheckMate 651 trial (NCT02741570) evaluated first-line nivolumab plus ipilimumab versus EXTREME (cetuximab, cisplatin/carboplatin, and 5-FU for up to six cycles followed by cetuximab maintenance) in patients with recurrent or metastatic head and neck SCC.[14] The primary end points were OS in all randomly assigned patients and patients with a PD-L1 CPS of 20 or higher. Secondary end points included OS in patients with a PD-L1 CPS of 1 or higher and PFS, objective response rate, and duration of response in all randomly assigned patients and patients with a PD-L1 CPS of 20 or higher.
    • Among all randomly assigned patients, there was no statistically significant difference in OS with nivolumab plus ipilimumab versus EXTREME (median OS, 13.9 vs. 13.5 months; HR, 0.95; 97.9% CI, 0.80–1.13; P = .4951). Among patients with a PD-L1 CPS of 20 or higher, there was also no statistically significant OS difference between the two treatments (median OS, 17.6 vs. 14.6 months; HR, 0.78; 97.51% CI, 0.59–1.03; P = .0469).[14][Level of evidence A1]
    • In patients with a CPS of 1 or higher, the median OS was 15.7 months for patients who received nivolumab plus ipilimumab versus 13.2 months for patients who received EXTREME (HR, 0.82; 95% CI, 0.69–0.97).
    • Among patients with a CPS of 20 or higher, the median PFS was 5.4 months for patients who received nivolumab plus ipilimumab and 7.0 months for patients who received EXTREME. The objective response rate was 34.1% for patients who received nivolumab plus ipilimumab and 36.0% for patients who received EXTREME.
    • Grade 3 or 4 treatment-related adverse events occurred in 28.2% of patients who received nivolumab plus ipilimumab and 70.7% of patients who received EXTREME.
    • CheckMate 651 did not meet its primary end points of OS in the randomly assigned or CPS of 20 or higher populations.

    The absence of a survival benefit for immune checkpoint inhibitors in this trial was an unexpected outcome, given the similarity of nivolumab to pembrolizumab in the studies of patients with cisplatin-refractory disease.[5,6] An editorial accompanying the CheckMate 651 trial analyzed some of the factors that may have contributed to a different result. The editorial suggested that survival in the control group, which was longer than that reported in prior studies, may have been impacted by the greater availability of second-line immunotherapy in the control group (46% in CheckMate 651 compared with 25% in the KEYNOTE-048 trial). The authors also suggested that the coadministration of ipilimumab detracted from the activity of nivolumab, as shown in the CheckMate 714 trial.[15]

  2. CheckMate 714 (NCT02823574), a double-blind phase II trial, evaluated the clinical benefit of first-line nivolumab plus ipilimumab versus nivolumab alone in 425 patients with recurrent or metastatic head and neck SCC.[16] Patients were randomly assigned in a 2:1 ratio to receive either nivolumab (3 mg/kg intravenously [IV] every 2 weeks) plus ipilimumab (1 mg/kg IV every 6 weeks) or nivolumab (3 mg/kg IV every 2 weeks) plus placebo. Treatment continued for up to 2 years or until disease progression, unacceptable toxic effects, or consent withdrawal. The primary end points were objective response rate and duration of response between treatment arms by blinded independent central review in the population with platinum-refractory recurrent or metastatic disease. These were patients who had recurrent disease less than 6 months after completion of platinum-based chemotherapy (adjuvant or neoadjuvant, or as part of multimodal treatment [chemotherapy, surgery, and/or radiation therapy]). Among the 241 patients (56.7%) with platinum-refractory disease, 159 were assigned to receive nivolumab plus ipilimumab and 82 were assigned to receive nivolumab alone. Among the 184 patients (43.3%) with platinum-eligible disease, 123 were assigned to receive nivolumab plus ipilimumab and 61 were assigned to receive nivolumab alone.[16][Level of evidence B3]
    • At primary database lock, the objective response rate in the population with platinum-refractory disease was 13.2% (95% CI, 8.4%–19.5%) with nivolumab plus ipilimumab and 18.3% (95% CI, 10.6%–28.4%) with nivolumab alone (odds ratio, 0.68; 95.5% CI, 0.33–1.43; P = .29).
    • The median duration of response was not reached (NR) in the nivolumab-plus-ipilimumab group (95% CI, 11.0 months–NR) and was 11.1 months (95% CI, 4.1–NR) in the nivolumab-alone group. In the population with platinum-eligible disease, the objective response rate was 20.3% (95% CI, 13.6%–28.5%) with nivolumab plus ipilimumab and 29.5% (95% CI, 18.5%–42.6%) with nivolumab alone.
    • Among the population with platinum-refractory disease, grade 3 or 4 treatment-related adverse events occurred in 25 of 158 patients (15.8%) who received nivolumab plus ipilimumab and in 12 of 82 patients (14.6%) who received nivolumab alone. Among the population with platinum-eligible disease, grade 3 or 4 treatment-related adverse events occurred in 30 of 122 patients (24.6%) who received nivolumab plus ipilimumab and in 8 of 61 patients (13.1%) who received nivolumab alone.
    • This trial did not meet its primary end point of objective response rate benefit with first-line nivolumab plus ipilimumab versus nivolumab alone in patients with platinum-refractory recurrent or metastatic head and neck SCC.

Evidence (nivolumab after progression on platinum-based treatment):

  1. A phase III open-label trial included 361 patients with recurrent SCC of the head and neck and disease progression within 6 months after platinum-based chemotherapy. Patients were randomly assigned in a 2:1 ratio to receive either nivolumab (at a dose of 3 mg/kg of body weight) every 2 weeks or standard single-agent systemic therapy (methotrexate, docetaxel, or cetuximab). The primary end point was OS.[6]
    • The median OS was 7.5 months (95% CI, 5.5–9.1) in the nivolumab group versus 5.1 months (95% CI, 4.0–6.0) in the standard therapy group. OS was statistically significantly longer with nivolumab than with standard therapy (HRdeath, 0.70; 97.73% CI, 0.51–0.96; P = .01). The estimated 1-year survival rate was approximately 19% higher in patients who received nivolumab (36.0%) than in those who received standard therapy (16.6%).[6][Level of evidence A1]
    • There was no statistically significant difference in median PFS between treatment groups. The 6-month PFS rate was 19.7% with nivolumab versus 9.9% with standard therapy.
    • The response rate was 13.3% in the nivolumab group versus 5.8% in the standard therapy group.
    • Grade 3 or 4 treatment-related adverse events occurred in 13.1% of the patients in the nivolumab group compared with 35.1% of the patients in the standard therapy group.
    • Quality-of-life outcomes—including physical, role, and social functioning and pain, sensory, and social contact problems—were stable in the nivolumab group but worse in the standard therapy group. These outcomes were assessed using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire (QLQ) Core Module (QLQ-C30) and the Head and Neck Module (QLQ-H&N35).
    • In the subgroup of patients with a PD-L1 expression level of 1% or higher, the HRdeath among patients treated with nivolumab versus standard therapy was 0.55 (95% CI, 0.36–0.83). In the subgroup of patients with a PD-L1 expression level lower than 1%, the HR was 0.89 (95% CI, 0.54–1.45; P = .17 for interaction).
  2. A randomized, phase III, superiority study in India evaluated the dose of immune checkpoint inhibitors in the setting of palliative care for patients with advanced head and neck cancer. Low-dose IV nivolumab (20 mg every 3 weeks) was added to a triple metronomic chemotherapy regimen of oral methotrexate (9 mg/m2 once weekly), celecoxib (200 mg twice daily), and erlotinib (150 mg once daily). Notably, this nivolumab dose is less than 10% of the dose recommended by the U.S. Food and Drug Administration and the European Medicines Agency. A total of 151 patients were randomly assigned to receive either triple metronomic chemotherapy alone (n = 75) or triple metronomic chemotherapy with nivolumab (n = 76). The primary end point was 1-year OS.[17]
    • The addition of low-dose nivolumab to triple metronomic chemotherapy improved the 1-year OS rate from 16.3% (95% CI, 8.0%–27.4%) to 43.4% (95% CI, 30.8%–55.3%) (HR, 0.545; 95% CI, 0.362–0.820; P = .0036).[17][Level of evidence A1]
    • The median OS was 6.7 months (95% CI, 5.8–8.1) for patients who received triple metronomic chemotherapy alone and 10.1 months (95% CI, 7.4–12.6) for patients who received triple metronomic chemotherapy with nivolumab (P = .0052).
    • The rate of grade 3 or higher adverse events was 50% for patients who received triple metronomic chemotherapy alone and 46.1% for patients who received triple metronomic chemotherapy with nivolumab (P = .744).

    Although the control arm in this study cannot be considered standard care, lower doses of immunotherapy appeared to have some benefit in this setting.[18]

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
  1. Wong LY, Wei WI, Lam LK, et al.: Salvage of recurrent head and neck squamous cell carcinoma after primary curative surgery. Head Neck 25 (11): 953-9, 2003. [PUBMED Abstract]
  2. Vikram B, Strong EW, Shah JP, et al.: Intraoperative radiotherapy in patients with recurrent head and neck cancer. Am J Surg 150 (4): 485-7, 1985. [PUBMED Abstract]
  3. Spencer SA, Harris J, Wheeler RH, et al.: RTOG 96-10: reirradiation with concurrent hydroxyurea and 5-fluorouracil in patients with squamous cell cancer of the head and neck. Int J Radiat Oncol Biol Phys 51 (5): 1299-304, 2001. [PUBMED Abstract]
  4. Vargo JA, Ferris RL, Ohr J, et al.: A prospective phase 2 trial of reirradiation with stereotactic body radiation therapy plus cetuximab in patients with previously irradiated recurrent squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys 91 (3): 480-8, 2015. [PUBMED Abstract]
  5. Cohen EEW, Soulières D, Le Tourneau C, et al.: Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study. Lancet 393 (10167): 156-167, 2019. [PUBMED Abstract]
  6. Ferris RL, Blumenschein G, Fayette J, et al.: Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med 375 (19): 1856-1867, 2016. [PUBMED Abstract]
  7. Burtness B, Harrington KJ, Greil R, et al.: Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet 394 (10212): 1915-1928, 2019. [PUBMED Abstract]
  8. Harrington KJ, Burtness B, Greil R, et al.: Pembrolizumab With or Without Chemotherapy in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: Updated Results of the Phase III KEYNOTE-048 Study. J Clin Oncol 41 (4): 790-802, 2023. [PUBMED Abstract]
  9. Tortochaux J, Tao Y, Tournay E, et al.: Randomized phase III trial (GORTEC 98-03) comparing re-irradiation plus chemotherapy versus methotrexate in patients with recurrent or a second primary head and neck squamous cell carcinoma, treated with a palliative intent. Radiother Oncol 100 (1): 70-5, 2011. [PUBMED Abstract]
  10. Vermorken JB, Mesia R, Rivera F, et al.: Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med 359 (11): 1116-27, 2008. [PUBMED Abstract]
  11. Mesía R, Rivera F, Kawecki A, et al.: Quality of life of patients receiving platinum-based chemotherapy plus cetuximab first line for recurrent and/or metastatic squamous cell carcinoma of the head and neck. Ann Oncol 21 (10): 1967-73, 2010. [PUBMED Abstract]
  12. Licitra L, Mesia R, Rivera F, et al.: Evaluation of EGFR gene copy number as a predictive biomarker for the efficacy of cetuximab in combination with chemotherapy in the first-line treatment of recurrent and/or metastatic squamous cell carcinoma of the head and neck: EXTREME study. Ann Oncol 22 (5): 1078-87, 2011. [PUBMED Abstract]
  13. Machiels JP, Haddad RI, Fayette J, et al.: Afatinib versus methotrexate as second-line treatment in patients with recurrent or metastatic squamous-cell carcinoma of the head and neck progressing on or after platinum-based therapy (LUX-Head & Neck 1): an open-label, randomised phase 3 trial. Lancet Oncol 16 (5): 583-94, 2015. [PUBMED Abstract]
  14. Haddad RI, Harrington K, Tahara M, et al.: Nivolumab Plus Ipilimumab Versus EXTREME Regimen as First-Line Treatment for Recurrent/Metastatic Squamous Cell Carcinoma of the Head and Neck: The Final Results of CheckMate 651. J Clin Oncol 41 (12): 2166-2180, 2023. [PUBMED Abstract]
  15. Burtness B: First-Line Nivolumab Plus Ipilimumab in Recurrent/Metastatic Head and Neck Cancer-What Happened? J Clin Oncol 41 (12): 2134-2137, 2023. [PUBMED Abstract]
  16. Harrington KJ, Ferris RL, Gillison M, et al.: Efficacy and Safety of Nivolumab Plus Ipilimumab vs Nivolumab Alone for Treatment of Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck: The Phase 2 CheckMate 714 Randomized Clinical Trial. JAMA Oncol 9 (6): 779-789, 2023. [PUBMED Abstract]
  17. Patil VM, Noronha V, Menon N, et al.: Low-Dose Immunotherapy in Head and Neck Cancer: A Randomized Study. J Clin Oncol 41 (2): 222-232, 2023. [PUBMED Abstract]
  18. Mitchell AP, Goldstein DA: Cost Savings and Increased Access With Ultra-Low-Dose Immunotherapy. J Clin Oncol 41 (2): 170-172, 2023. [PUBMED Abstract]

Latest Updates to This Summary (05/14/2025)

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.

Editorial changes were made to this summary.

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 oropharyngeal 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 Oropharyngeal Cancer Treatment are:

  • Andrea Bonetti, MD (Pederzoli Hospital)
  • 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 Oropharyngeal Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/head-and-neck/hp/adult/oropharyngeal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389168 ]

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.