Childhood Colorectal Cancer (PDQ®)–Patient Version

Childhood Colorectal Cancer (PDQ®)–Patient Version

What is childhood colorectal cancer?

Childhood colorectal cancer is a rare cancer that forms in the tissues of the colon or the rectum. In the United States, there are fewer than 100 children diagnosed with colorectal cancer each year.

The colon is part of the body’s digestive system. The digestive system removes and processes nutrients (vitamins, minerals, carbohydrates, fats, proteins, and water) from foods and helps pass waste material out of the body. The digestive system is made up of the mouth, throat, esophagus, stomach, and the small and large intestines. The colon (large bowel) is the main part of the large intestine and is about 5 feet long in an adult. Together, the rectum and anal canal make up the last part of the large intestine and are 6 to 8 inches long. The anal canal ends at the anus (the opening of the large intestine to the outside of the body).

EnlargeGastrointestinal (digestive) system anatomy; drawing shows the esophagus, liver, stomach, colon, small intestine, rectum, and anus.
Anatomy of the lower gastrointestinal (digestive) system showing the colon, rectum, and anus. Other organs that make up the digestive system are also shown.

Causes and risk factors for childhood colorectal cancer

Childhood colorectal cancer is caused by certain changes to the way the cells in the colon or rectum 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. Not every child with one or more of these risk factors will develop colorectal cancer. And it will develop in some children who don’t have a known risk factor.

Childhood colorectal cancer may be part of an inherited cancer syndrome. Inherited cancer syndromes are caused by changes in certain genes passed from parents to children. The following inherited cancer syndromes increase a child’s risk of colorectal cancer:

Children with Crohn disease or ulcerative colitis may be at increased risk of colorectal cancer.

Polyps that form in the colon of children who do not have an inherited syndrome are not linked to an increased risk of cancer.

Talk with your child’s doctor if you think your child may be at risk.

Genetic counseling for children with colorectal cancer

It may not be clear from the family medical history whether your child’s colorectal cancer is part of an inherited condition. Genetic counseling can assess the likelihood that your child’s cancer is inherited and whether genetic testing is needed. Genetic counselors and other specially trained health professionals can discuss your child’s diagnosis and your family’s medical history to help you understand the:

  • options for testing for changes in the APC, NF1, MUTYH, NTHL1, and other genes
  • risk of other cancers for your child
  • risk of colorectal cancer or other cancers for your child’s siblings
  • risks and benefits of learning genetic information

Genetic counselors can also help you cope with your child’s genetic testing results, including how to discuss the results with family members. They can also advise about whether other members of your family should receive genetic testing.

Learn more about Genetic Testing for Inherited Cancer Risk.

Symptoms of childhood colorectal cancer

Symptoms of childhood colorectal cancer usually depend on where the tumor forms. It’s important to check with your child’s doctor if your child has:

  • pain in the abdomen
  • constipation
  • diarrhea
  • blood in the stool
  • nausea and vomiting
  • weight loss for no known reason
  • a lump in the abdomen
  • loss of appetite
  • anemia (tiredness, dizziness, fast or irregular heartbeat, shortness of breath, pale skin)

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

Tests to diagnose childhood colorectal cancer

If your child has symptoms that suggest colorectal 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, they may recommend other tests.

Diagnosis of childhood colorectal cancer

The following tests and procedures are used to diagnose colorectal cancer. The results will also help you and your child’s doctor plan treatment.

Colonoscopy

A colonoscopy looks inside the rectum and colon for polyps, abnormal areas, or cancer. A colonoscope is inserted through the rectum into the colon. A colonoscope is a thin, tube-like instrument with a light and a lens for viewing. It also has a tool to remove polyps or tissue samples, which are checked under a microscope for cancer.

Barium enema

A barium enema is a series of x-rays of the lower gastrointestinal tract. A liquid that contains barium (a silver-white metallic compound) is put into the rectum. The barium coats the lower gastrointestinal tract and x-rays are taken. This procedure is also called a lower GI series.

Fecal occult blood test

A fecal occult blood test checks stool for blood that can only be seen with a microscope. Small samples of stool are placed on special cards and returned to the doctor or laboratory for testing.

Complete blood count (CBC)

A CBC checks a sample of blood for:

Carcinoembryonic antigen (CEA) assay

A CEA test measures the level of carcinoembryonic antigen (CEA) in the blood. CEA is released into the bloodstream from both cancer cells and normal cells. When found in higher-than-normal amounts, CEA can be a sign of colorectal cancer or other conditions.

Molecular testing

A molecular test checks for certain genes, proteins, or other molecules in a sample of tissue, blood, or bone marrow. Molecular tests also check for certain changes in a gene or chromosome that may cause or affect the chance of developing colorectal cancer. A molecular test may be used to help plan treatment, find out how well treatment is working, or make a prognosis.

The Molecular Characterization Initiative offers free molecular testing to children, adolescents, and young adults with certain types of newly diagnosed cancer. The program is offered through NCI’s Childhood Cancer Data Initiative. To learn more, visit About the Molecular Characterization Initiative.

Tests to stage childhood colorectal cancer

If your child has been diagnosed with colorectal cancer, your child will be referred to a pediatric oncologist. This is a doctor who specializes in diagnosing and treating childhood cancers. Your child’s doctor will recommend tests to find out if the cancer has spread and if so, how far. The process of learning the extent of cancer in the body is called staging. At the time of diagnosis, colorectal cancer in children has often spread to the lymph nodes, outside the colon or rectum, or to other organs in the abdomen.

The following imaging tests and procedures may be used to determine the stage of colorectal cancer:

PET scan

A 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 a picture 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. When this procedure is done at the same time as a CT scan or an MRI, it is called a PET-CT scan or a PET-MRI.

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 chest, abdomen, and pelvis. 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, such as the chest. The pictures are taken from different angles and are used to create 3-D views of tissues and organs. A dye may be injected into a vein or swallowed to help the organs or tissues show up more clearly. This procedure is also called computed tomography, computerized tomography, or computerized axial tomography. Learn more at 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 genetic test results, pathology report, slides, and scans. This doctor may agree with the first doctor, suggest changes to the treatment plan, or provide more information about your child’s 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 colorectal cancer

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 colorectal cancer to plan the best treatment.

There are several staging systems for cancer that describe the extent of the cancer. Colorectal cancer staging for adults and children usually uses the TNM staging system. You may see your child’s cancer 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, 4) is assigned to your child’s cancer. When talking to you about your child’s cancer, the doctor may describe it as one of these stages.

For information about how doctors stage colorectal cancer, visit the Tests to stage colorectal cancer section. Learn more about the TNM colorectal cancer staging system in the Stages of Colon Cancer section of Colon Cancer Treatment.

Types of treatment for childhood cancer

Who treats children with colorectal cancer?

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

There are different types of treatment for children and adolescents with colorectal 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.

Types of treatment your child might have include:

Surgery

Surgery to remove the cancer is done if the cancer has not spread to other parts of the body at diagnosis. 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. Colorectal cancer may be 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.

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.

For colorectal cancer, chemotherapy is taken by mouth or injected into a vein. When given this way, the drugs enter the bloodstream to reach cancer cells throughout the body. Chemotherapy drugs used alone or in combination to treat colorectal cancer in children include:

Other chemotherapy drugs not listed here may also be used.

To learn more about how chemotherapy works, how it is given, common side effects, and more, visit Chemotherapy to Treat Cancer.

Immunotherapy

Immunotherapy helps a child’s immune system fight cancer. Immunotherapy drugs that may be used to treat colorectal cancer include:

Learn more about how immunotherapy works against cancer, how it is given, possible side effects, and more at 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 colorectal cancer

Treatment of newly diagnosed colorectal cancer in children may include:

  • surgery to remove the tumor if it has not spread
  • radiation therapy and chemotherapy for tumors in the rectum or lower colon
  • combination chemotherapy, for advanced colorectal cancer

Children with certain inherited cancer syndromes may be treated with:

  • surgery to remove the colon before cancer forms
  • medicine to decrease the number of polyps in the colon

Treatment of colorectal cancer that cannot be removed by surgery, has spread to other parts of the body, or has continued to grow and spread after treatment may include immunotherapy with nivolumab or pembrolizumab. Immunotherapy is only given if your child has certain inherited cancer syndromes or if the cancer has specific gene changes.

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.

Prognostic factors for childhood colorectal cancer

If your child has been diagnosed with colorectal 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 depends on:

  • whether the tumor was completely removed by surgery
  • whether the cancer has spread to other parts of the body, such as the lymph nodes, lung, liver, pelvis, ovaries, or bone
  • whether the cancer has just been diagnosed or has recurred (come back)

Childhood colorectal cancer is challenging to treat because it has usually spread to other areas in the body at diagnosis. Your child’s 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:

  • physical problems
  • 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 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).

To learn more about follow-up tests, visit Tests to diagnose childhood colorectal cancer.

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. Learn more at Support for Families: Childhood Cancer and in 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 colorectal 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 Colorectal Cancer. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/colorectal/patient/child-colorectal-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.

Skin Cancer Prevention (PDQ®)–Health Professional Version

Skin Cancer Prevention (PDQ®)–Health Professional Version

Who Is at Risk?

Individuals who have light-hair and -eye color, freckles, and who sunburn easily are particularly susceptible to developing skin cancer.[1] There are two primary types of skin cancer, keratinocyte carcinoma (including basal cell carcinoma and squamous cell carcinoma [SCC]) and melanoma. Observational and analytic epidemiological studies have consistently shown that increased cumulative sun exposure is a risk factor for keratinocyte carcinoma.[1,2] Melanoma risk correlates with common and atypical nevi.[3] Some studies suggest that there may be an interplay between genetic phenotype and sun exposure and that there may be two pathways to melanoma development.[47]

Organ transplant recipients taking immunosuppressive drugs are at an elevated risk of developing skin cancer, particularly SCC.[8,9] Arsenic exposure also increases the risk of keratinocytic cancers [10] and melanoma.[11]

References
  1. Preston DS, Stern RS: Nonmelanoma cancers of the skin. N Engl J Med 327 (23): 1649-62, 1992. [PUBMED Abstract]
  2. English DR, Armstrong BK, Kricker A, et al.: Case-control study of sun exposure and squamous cell carcinoma of the skin. Int J Cancer 77 (3): 347-53, 1998. [PUBMED Abstract]
  3. Gandini S, Sera F, Cattaruzza MS, et al.: Meta-analysis of risk factors for cutaneous melanoma: I. Common and atypical naevi. Eur J Cancer 41 (1): 28-44, 2005. [PUBMED Abstract]
  4. Armstrong BK, Cust AE: Sun exposure and skin cancer, and the puzzle of cutaneous melanoma: A perspective on Fears et al. Mathematical models of age and ultraviolet effects on the incidence of skin cancer among whites in the United States. American Journal of Epidemiology 1977; 105: 420-427. Cancer Epidemiol 48: 147-156, 2017. [PUBMED Abstract]
  5. Olsen CM, Pandeya N, Law MH, et al.: Does polygenic risk influence associations between sun exposure and melanoma? A prospective cohort analysis. Br J Dermatol 183 (2): 303-310, 2020. [PUBMED Abstract]
  6. Davis LE, Shalin SC, Tackett AJ: Current state of melanoma diagnosis and treatment. Cancer Biol Ther 20 (11): 1366-1379, 2019. [PUBMED Abstract]
  7. Gershenwald JE, Guy GP: Stemming the Rising Incidence of Melanoma: Calling Prevention to Action. J Natl Cancer Inst 108 (1): , 2016. [PUBMED Abstract]
  8. Ascha M, Ascha MS, Tanenbaum J, et al.: Risk Factors for Melanoma in Renal Transplant Recipients. JAMA Dermatol 153 (11): 1130-1136, 2017. [PUBMED Abstract]
  9. Rollan MP, Cabrera R, Schwartz RA: Current knowledge of immunosuppression as a risk factor for skin cancer development. Crit Rev Oncol Hematol 177: 103754, 2022. [PUBMED Abstract]
  10. Tseng WP, Chu HM, How SW, et al.: Prevalence of skin cancer in an endemic area of chronic arsenicism in Taiwan. J Natl Cancer Inst 40 (3): 453-63, 1968. [PUBMED Abstract]
  11. Beane Freeman LE, Dennis LK, Lynch CF, et al.: Toenail arsenic content and cutaneous melanoma in Iowa. Am J Epidemiol 160 (7): 679-87, 2004. [PUBMED Abstract]

Overview

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 containing information related to skin cancer prevention include the following:

Factors Associated With an Increased Risk of Keratinocyte Carcinoma (Basal Cell Carcinoma, Squamous Cell Carcinoma)

Fair skin

Based on solid evidence, individuals with fair skin types (light or pale skin, light-hair and -eye color, freckles, or those who burn easily) are associated with an increased risk of squamous cell carcinoma (SCC) and basal cell carcinoma (BCC).

Magnitude of Effect: Substantial, depending on the amount of exposure.

  • Study Design: Observational studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Sun and UV radiation exposure

Based on solid evidence, sun and UV radiation exposure are associated with an increased risk of SCC and BCC.

Magnitude of Effect: Substantial, depending on the amount of exposure.

  • Study Design: Observational studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Immunosuppression

Based on solid evidence, immunosuppression after organ transplant is associated with an increased risk of SCC and BCC.

Magnitude of Effect: Substantial, although not consistently quantitated.

  • Study Design: Observational studies.
  • Internal Validity: Good.
  • Consistency: Good.
  • External Validity: Good.

Arsenic exposure

Based on fair evidence, arsenic exposure is associated with an increased risk of keratinocyte carcinoma.

Magnitude of Effect: Arsenic exposure is associated with keratinocyte carcinoma.

  • Study Design: One case-control study.
  • Internal Validity: Good.
  • Consistency: Fair.
  • External Validity: Fair.

Factors Associated With an Increased Risk of Melanoma

Sun and UV radiation exposure

Based on fair evidence, intermittent acute sun exposure leading to sunburn is associated with an increased risk of melanoma.

Magnitude of Effect: Unknown.

  • Study Design: Observational studies.
  • Internal Validity: Fair.
  • Consistency: Fair.
  • External Validity: Poor.

Arsenic exposure

Based on fair evidence, arsenic exposure is associated with an increased risk of melanoma.

Magnitude of Effect: Arsenic exposure is associated with double the incidence of melanoma.

  • Study Design: One case-control study.
  • Internal Validity: Good.
  • Consistency: Fair.
  • External Validity: Fair.

Interventions for Skin Cancer Prevention With Adequate Evidence

Treatment of sun-damaged skin to prevent skin cancer: Benefits

There is one well designed randomized controlled trial (RCT) that demonstrated the use of topical fluorouracil on sun-damaged skin prevents additional actinic keratoses and SCC requiring surgery.[1]

Magnitude of Effect: Moderate net benefit in preventing SCC requiring surgery.

  • Study Design: RCT.
  • Internal Validity: Good.
  • Consistency: N/A (single study).
  • External Validity: Fair.

Treatment of sun-damaged skin to prevent skin cancer: Harms

The primary side effect is local erythema, irritation, and crusting.

Interventions for Skin Cancer Prevention With Inadequate Evidence

Behavior counseling to change sun-protection practices: Benefits

Evidence from 21 RCTs demonstrated that behavior counseling for children and families and for adults improves sun protective behaviors. These trials showed an inconsistent effect on reducing sunburns and do not provide direct evidence on reduction of SCC, BCC, or melanoma.[2]

Magnitude of Benefit: Moderate net benefit for improving sun protective behaviors, but there is inadequate direct evidence to determine the impact on the development of skin cancer.

  • Study Design: Systematic review including 21 RCTs.
  • Internal Validity: Good.
  • Consistency: Good for behaviors. Poor for sunburns.
  • External Validity: Good.

Behavior counseling to change sun-protection practices: Harms

Avoiding sun exposure can result in harms, such as mood disorders, sleep disturbances, elevated blood pressure, and impaired vitamin D metabolism, which is associated with increased incidence of colon, ovary, and breast cancers, and multiple myeloma.[3]

Topical treatments to prevent skin cancer—sunscreen: Benefits

Sunscreen has been shown to prevent sunburns and actinic keratoses. RCTs showed inconsistent benefit in preventing SCC and showed no benefit in preventing melanoma.

Magnitude of Effect: Inadequate evidence to assess magnitude of effect for sunscreen.

  • Study Design: RCTs and observational cohort studies.
  • Internal Validity: Poor.
  • Consistency: Inconsistent.
  • External Validity: Poor.

Topical treatments to prevent skin cancer—sunscreen: Harms

Harms of sunscreen for the user are mild and mainly include skin allergic reactions. Because sunscreen use prevents sunburns, it may encourage more sun exposure to fair skinned people at risk of developing skin cancer.

Systemic treatments to prevent skin cancer (nonsteroidal anti-inflammatory drugs [NSAIDs], nicotinamide, isotretinoin, selenium, beta carotene, alpha-difluoromethylornithine [DFMO]): Benefits

There is no evidence showing that NSAIDs and nicotinamide prevent SCC. RCTs found no benefit in preventing SCC, BCC, or melanoma for topical or oral retinoids, selenium, and beta carotene. One RCT showed a slight reduction in BCC for DFMO, but no change in SCC or melanoma.

Magnitude of Effect: Inadequate evidence to assess magnitude of effect for topical retinoids, and nicotinamide. Harms likely outweigh potential benefits for NSAIDs, oral retinoids, beta carotene, and DFMO.

  • Study Design: RCTs and observational cohort studies.
  • Internal Validity: Poor.
  • Consistency: Inconsistent.
  • External Validity: Poor.

Systemic treatments to prevent skin cancer (NSAIDs, nicotinamide, isotretinoin, selenium, beta carotene, DFMO): Harms

NSAIDs are associated with adverse cardiovascular effects, gastrointestinal bleeding, and kidney damage. Oral retinoids are hepatotoxic and cause hypertriglyceridemia. In RCTs, beta carotene is associated with an increased risk of lung cancer incidence and mortality in smokers. Isotretinoin has dose-related skin toxicity. Patients discontinue DFMO at high rates because of hearing loss.

References
  1. Weinstock MA, Thwin SS, Siegel JA, et al.: Chemoprevention of Basal and Squamous Cell Carcinoma With a Single Course of Fluorouracil, 5%, Cream: A Randomized Clinical Trial. JAMA Dermatol 154 (2): 167-174, 2018. [PUBMED Abstract]
  2. Henrikson NB, Morrison CC, Blasi PR, et al.: Behavioral Counseling for Skin Cancer Prevention: Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 319 (11): 1143-1157, 2018. [PUBMED Abstract]
  3. Mead MN: Benefits of sunlight: a bright spot for human health. Environ Health Perspect 116 (4): A160-7, 2008. [PUBMED Abstract]

Incidence and Mortality of Skin Cancer

There are two main types of skin cancer:

  • Keratinocyte carcinoma.
    • Basal cell carcinoma (BCC).
    • Squamous cell carcinoma (SCC).
  • Melanoma.

BCC and SCC are the most common forms of skin cancer but have substantially better prognoses than the less common, generally more aggressive, melanoma.

Keratinocyte carcinomas are the most commonly occurring cancer in the United States, but exact incidence figures are unavailable because cases are not required to be reported to cancer registries. Incidence rates appear to have been increasing for a number of years,[1] in part due to increased screening and biopsy of skin lesions. Based on an extrapolation of Medicare fee-for-service data to the U.S. population, about 3 million individuals were estimated to have been diagnosed with keratinocyte carcinomas in 2012,[1,2] exceeding all other cancer cases (approximately 2 million) estimated by the American Cancer Society in 2025.[1]

Melanoma cases are reported to U.S. cancer registries, so data are available. In 2025, an estimated 104,960 individuals in the United States will be diagnosed with melanoma and approximately 8,430 will die of the disease.[1] While only 2% of skin cancers are melanomas, melanoma causes more than 80% of deaths from skin cancer.[3]

References
  1. American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
  2. Rogers HW, Weinstock MA, Feldman SR, et al.: Incidence Estimate of Nonmelanoma Skin Cancer (Keratinocyte Carcinomas) in the U.S. Population, 2012. JAMA Dermatol 151 (10): 1081-6, 2015. [PUBMED Abstract]
  3. Weinstock MA, Bogaars HA, Ashley M, et al.: Nonmelanoma skin cancer mortality. A population-based study. Arch Dermatol 127 (8): 1194-7, 1991. [PUBMED Abstract]

Accuracy of Making a Clinical Diagnosis of Melanoma

Observer variability among physicians has been noted in the evaluation of skin lesions and subsequent biopsy specimens. A systematic review of 32 studies that compared the accuracy of dermatologists and primary care physicians in making a clinical diagnosis of melanoma concluded that there was no statistically significant difference in accuracy. However, the results were inconclusive, owing to small sample sizes and study design weaknesses.[1] Subsequent studies have noted a higher accuracy for dermatologists in the diagnosis of melanocytic lesions,[2,3] yet there is a shortage of dermatologists to meet the demands of population-level screening.

A study of 187 pathologists in the United States found that cases of moderately dysplastic nevi to early-stage invasive melanoma had less than 50% agreement with a reference diagnosis defined by consensus of experienced pathologists.[4] At a U.S. population level, it is estimated that 82.8% (95% confidence interval, 81.0%–84.5%) of melanocytic skin biopsy diagnoses would be verified if they were reviewed by a consensus reference panel of experienced pathologists.[4] In addition, differentiating between benign and malignant melanocytic tumors during histological examinations of biopsy specimens has been shown to be inconsistent, even in the hands of experienced dermatopathologists.[5,6] This variability in the diagnosis of melanocytic lesions undermines the results of studies that examine screening effectiveness and also may undermine the effectiveness of any screening intervention. Furthermore, this finding suggests that requesting a second opinion regarding the pathology of biopsy specimens may be important.[57] A standard approach to the classification of melanocytic skin lesions by pathologists may also reduce confusion and improve communication between clinicians.[4,6,8,9]

References
  1. Chen SC, Bravata DM, Weil E, et al.: A comparison of dermatologists’ and primary care physicians’ accuracy in diagnosing melanoma: a systematic review. Arch Dermatol 137 (12): 1627-34, 2001. [PUBMED Abstract]
  2. Chen SC, Pennie ML, Kolm P, et al.: Diagnosing and managing cutaneous pigmented lesions: primary care physicians versus dermatologists. J Gen Intern Med 21 (7): 678-82, 2006. [PUBMED Abstract]
  3. Corbo MD, Wismer J: Agreement between dermatologists and primary care practitioners in the diagnosis of malignant melanoma: review of the literature. J Cutan Med Surg 16 (5): 306-10, 2012 Sep-Oct. [PUBMED Abstract]
  4. Elmore JG, Barnhill RL, Elder DE, et al.: Pathologists’ diagnosis of invasive melanoma and melanocytic proliferations: observer accuracy and reproducibility study. BMJ 357: j2813, 2017. [PUBMED Abstract]
  5. Farmer ER, Gonin R, Hanna MP: Discordance in the histopathologic diagnosis of melanoma and melanocytic nevi between expert pathologists. Hum Pathol 27 (6): 528-31, 1996. [PUBMED Abstract]
  6. Lott JP, Elmore JG, Zhao GA, et al.: Evaluation of the Melanocytic Pathology Assessment Tool and Hierarchy for Diagnosis (MPATH-Dx) classification scheme for diagnosis of cutaneous melanocytic neoplasms: Results from the International Melanoma Pathology Study Group. J Am Acad Dermatol 75 (2): 356-63, 2016. [PUBMED Abstract]
  7. Piepkorn MW, Longton GM, Reisch LM, et al.: Assessment of Second-Opinion Strategies for Diagnoses of Cutaneous Melanocytic Lesions. JAMA Netw Open 2 (10): e1912597, 2019. [PUBMED Abstract]
  8. Piepkorn MW, Barnhill RL, Elder DE, et al.: The MPATH-Dx reporting schema for melanocytic proliferations and melanoma. J Am Acad Dermatol 70 (1): 131-41, 2014. [PUBMED Abstract]
  9. Radick AC, Reisch LM, Shucard HL, et al.: Terminology for melanocytic skin lesions and the MPATH-Dx classification schema: A survey of dermatopathologists. J Cutan Pathol 48 (6): 733-738, 2021. [PUBMED Abstract]

Risk Factors for Skin Cancer

Epidemiological evidence suggests that exposure to UV radiation and the sensitivity of an individual’s skin to UV radiation are the main risk factors for skin cancer, although the type of exposure (high-intensity and short-duration vs. chronic exposure) and the pattern of exposure (continuous vs. intermittent) may differ among the two main skin cancer types.[13]

The immune system plays a role in the pathogenesis of skin cancer: organ transplant recipients taking immunosuppressive drugs are at an elevated risk of skin cancer, both squamous cell carcinoma (SCC) and melanoma.[4] Arsenic exposure also increases the risk of cutaneous SCC.[4]

The visible evidence of susceptibility to skin cancer (skin type and precancerous lesions), presence of sun-induced skin damage (sunburn and solar keratoses), and increased number of nevi and atypical nevi are associated with an increased risk of melanoma.[5,6]

Factors Associated With Increased Risk of Keratinocyte Carcinoma

UV radiation exposure

Most evidence about UV radiation exposure and the prevention of skin cancer comes from observational and analytic epidemiological studies. Such studies have consistently shown that increased cumulative sun exposure is a risk factor for keratinocyte carcinomas.[2,3] Individuals whose skin tans poorly or burns easily after sun exposure are particularly susceptible.[2]

Actinic keratoses

It is generally felt that one-half or more of SCCs arise from actinic keratoses. However, nearly one-half of SCCs occur in clinically normal skin.[7] A longitudinal study has shown that the progression rate from actinic keratoses to SCC is about 0.075% to 0.096% per year, or less than 1 case in 1,000 per year.[7] Moreover, in a population-based longitudinal study, there was an approximately 26% spontaneous regression rate of actinic keratoses within 1 year of a screening examination.[8]

Factors Associated With an Increased Risk of Melanoma

UV radiation exposure

The relationship between UV radiation exposure and cutaneous melanoma is less clear than the relationship between UV exposure and keratinocyte carcinoma. In the case of melanoma, it seems that intermittent acute sun exposure leading to sunburn is more important than cumulative sun exposure;[9] such exposures during childhood or adolescence may be particularly important.[1]

Multiple case control studies have also documented the association between sun exposure and melanoma. Total sun exposure in childhood is associated with an increased risk of melanoma (odds ratio, 1.81–4.4) as is recreational sun exposure during childhood and adulthood, while occupational sun exposure may be associated with a decreased risk of melanoma.[10,11] Fair skin that sunburns easily has a twofold risk of melanoma compared with skin phenotypes that never burn. Natural red and blond hair and natural blond hair also confers a twofold to fourfold increased risk of melanoma.[12]

References
  1. Koh HK: Cutaneous melanoma. N Engl J Med 325 (3): 171-82, 1991. [PUBMED Abstract]
  2. Preston DS, Stern RS: Nonmelanoma cancers of the skin. N Engl J Med 327 (23): 1649-62, 1992. [PUBMED Abstract]
  3. English DR, Armstrong BK, Kricker A, et al.: Case-control study of sun exposure and squamous cell carcinoma of the skin. Int J Cancer 77 (3): 347-53, 1998. [PUBMED Abstract]
  4. Beane Freeman LE, Dennis LK, Lynch CF, et al.: Toenail arsenic content and cutaneous melanoma in Iowa. Am J Epidemiol 160 (7): 679-87, 2004. [PUBMED Abstract]
  5. Gandini S, Sera F, Cattaruzza MS, et al.: Meta-analysis of risk factors for cutaneous melanoma: I. Common and atypical naevi. Eur J Cancer 41 (1): 28-44, 2005. [PUBMED Abstract]
  6. Cho E, Rosner BA, Colditz GA: Risk factors for melanoma by body site. Cancer Epidemiol Biomarkers Prev 14 (5): 1241-4, 2005. [PUBMED Abstract]
  7. Marks R, Rennie G, Selwood TS: Malignant transformation of solar keratoses to squamous cell carcinoma. Lancet 1 (8589): 795-7, 1988. [PUBMED Abstract]
  8. Marks R, Foley P, Goodman G, et al.: Spontaneous remission of solar keratoses: the case for conservative management. Br J Dermatol 115 (6): 649-55, 1986. [PUBMED Abstract]
  9. Gandini S, Sera F, Cattaruzza MS, et al.: Meta-analysis of risk factors for cutaneous melanoma: II. Sun exposure. Eur J Cancer 41 (1): 45-60, 2005. [PUBMED Abstract]
  10. Lin JS, Eder M, Weinmann S, et al.: Behavioral Counseling to Prevent Skin Cancer: Systematic Evidence Review to Update the 2003 U.S. Preventive Services Task Force Recommendation. Agency for Healthcare Research and Quality, 2011. Report No.: 11-05152-EF-1. Also available online. Last accessed April 8, 2025.
  11. Henrikson NB, Morrison CC, Blasi PR, et al.: Behavioral Counseling for Skin Cancer Prevention: Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 319 (11): 1143-1157, 2018. [PUBMED Abstract]
  12. Gandini S, Sera F, Cattaruzza MS, et al.: Meta-analysis of risk factors for cutaneous melanoma: III. Family history, actinic damage and phenotypic factors. Eur J Cancer 41 (14): 2040-59, 2005. [PUBMED Abstract]

Interventions for Skin Cancer Prevention With Adequate Evidence of Benefit

Treatment of Sun-Damaged Skin to Prevent Skin Cancer

Topical fluorouracil

Daily application of topical fluorouracil for up to 4 weeks onto actinic keratosis has been shown to reduce the development of new actinic keratoses.[1,2] A randomized controlled trial included 932 veterans with sun-damaged skin (two or more keratinocyte carcinomas in the 5 years before enrollment). Participants were randomly assigned to a 2- to 4-week single course of 5% topical fluorouracil or vehicle control cream. The fluorouracil group had fewer actinic keratosis cases when compared with the control group at 6 months (3.0 vs. 8.1; P < .001) and for the overall study duration (P < .001). Topical fluorouracil also reduced the risk of squamous cell carcinoma (SCC) requiring surgery at those sites for 1 year, but no effect was seen on basal cell carcinoma (BCC) in year 1 or on SCC or BCC over 4 years. Erythema, crusting, and irritation were reported by 82% of participants, with 40% reporting symptoms as severe. However, at study completion, 87% reported a willingness to repeat the treatment if needed.[1]

References
  1. Weinstock MA, Thwin SS, Siegel JA, et al.: Chemoprevention of Basal and Squamous Cell Carcinoma With a Single Course of Fluorouracil, 5%, Cream: A Randomized Clinical Trial. JAMA Dermatol 154 (2): 167-174, 2018. [PUBMED Abstract]
  2. Rosenberg AR, Tabacchi M, Ngo KH, et al.: Skin cancer precursor immunotherapy for squamous cell carcinoma prevention. JCI Insight 4 (6): , 2019. [PUBMED Abstract]

Interventions for Skin Cancer Prevention With Inadequate Evidence of Benefit

Behavioral Interventions to Change Sun-Protective Practices

The U.S. Preventive Services Task Force (USPSTF) commissioned a systematic review of primary care behavioral counseling interventions for skin cancer prevention.[1] The review identified 21 trials on promoting protective behaviors in 27 publications with 20,561 participants. Protective behaviors included use of protective clothing to limit UV radiation exposure, sun-avoidance behaviors, and use of sunscreen. Interventions included physician counseling, tailored mailings and texts, educational presentations, and interactive web programs involving patients and families. Five of six trials in children found that interventions reduced parent-reported composite sun protection scores at 3 months to 3 years.[26] Six of twelve trials in adults also showed that interventions resulted in a reduced patient-reported composite sun protection score, with the greatest change being increased use of sunscreen.[713]

The trials did not show a consistent change in sunburns for children or adults. In the three trials of children that assessed changes in sunburn frequency (n = 2,508),[1416] only one trial showed a reduction in nonsevere burns, but no change in severe burns.[15] In the six trials of adults that assessed changes in sunburn frequency (n = 3,959),[1722] only one trial showed a slight reduction in red or painful burns at 3 months.[17] There were no changes reported in any trial showing reductions in skin cancer (keratinocyte carcinoma or melanoma) or skin cancer precursors (nevi or actinic keratosis).

While direct evidence is lacking, the USPSTF linked the evidence demonstrating that behavioral counseling interventions promote sun protective practices with the epidemiological data on UV exposure and skin cancer prevalence. This led to a recommendation for counseling children, adolescents, and young adults aged 6 months to 24 years and adults older than 24 years with fair skin on protective practices to reduce skin cancer.[23]

Topical Treatment to Prevent Skin Cancer—Sunscreen

Sunscreen use has been shown to decrease the rate of developing new actinic keratoses [24] and to increase the remission rate of existing lesions.[25] Another trial found no difference in keratinocyte cancers in daily versus discretionary sunscreen users.[26] An 8-year observational posttrial follow-up showed reductions in both squamous cancers [27] and melanomas [28] associated with sunscreen use, but the confidence intervals (CIs) were very wide, and the participation outside the initial trial introduced uncertainty.

A meta-analysis of 18 studies that explored the association between melanoma risk and previous sunscreen use illustrated widely differing study qualities and suggested little or no association.[29] A systematic review of the association between sunscreen use and the development of melanocytic nevi in children reported similar issues with study quality and heterogeneity, hindering conclusive assessments. However, of the 15 studies that met inclusion criteria, 12 found either an increased incidence or no association.[30]

Systemic Medications to Prevent Skin Cancer

Nonsteroidal anti-inflammatory drugs (NSAIDS)

A randomized controlled trial (RCT) included 240 people at high risk of skin cancer (each with 10–40 actinic keratoses and a history of previous skin cancer) who were given celecoxib 200 mg twice daily or a placebo for 9 months. The trial found no difference in the incidence of actinic keratosis, but a post hoc analysis revealed a statistically significant difference in the mean number of keratinocyte carcinomas per patient (rate ratio, 0.43; 95% CI, 0.24–0.75; absolute difference, 0.2 lesions per patient).[31] A meta-analysis of nine studies (five case-control, three cohort, and one intervention) reported a small reduction in squamous cell carcinoma (SCC) risk associated with the use of nonaspirin NSAIDs (relative risk [RR], 0.85; 95% CI, 0.78–0.94), with the effect seen particularly in those with previous actinic skin tumors.[32]

NSAIDs are associated with known adverse cardiovascular effects, gastrointestinal bleeding, and kidney damage.[33]

Nicotinamide (vitamin B3)

The effect of nicotinamide on the development of new actinic keratosis lesions has been studied with inadequate evidence for efficacy, even in higher-risk populations. Studies include a clinical trial of patients with four or fewer actinic keratosis lesions at baseline (Oral Nicotinamide to Reduce Actinic Cancer [ONTRAC]) [34] and a trial of immunosuppressed organ-transplant recipients (Oral Nicotinamide to Reduce Actinic Cancer after Transplant [ONTRANS]).[35] The ONTRAC trial showed a lower rate of new lesions while individuals received treatment, but not during the 6-month postintervention follow-up period.[36] The ONTRANS trial was impacted by slow recruitment and was stopped early but showed no efficacy in the limited sample size.

Isotretinoin and related systemic retinoids such as acitretin

Retinoids are vitamin A derivatives that are available in topical and oral preparations. Oral retinoids have been studied in high-risk populations, such as those with a history of multiple nonmelanoma skin cancers, genetic disorders such as xeroderma pigmentosum, transplant recipients, and those exposed to high cumulative levels of psoralen plus UV A (PUVA) therapy.[3743] However, side effects of oral retinoids, including hypertriglyceridemia and hepatic toxicity, are significant.

Topical tretinoin 0.1% cream was compared with a control for 1.5 to 5.5 years in an RCT. No difference was found in the proportions of patients who developed SCC or basal cell carcinoma (BCC) or actinic keratosis.[44]

Selenium

A multicenter, double-blind, randomized, placebo-controlled trial included 1,312 patients with a history of BCC or SCC and a mean follow-up of 6.4 years. The study showed that 200 µg of selenium (in brewer’s yeast tablets) did not have a statistically significant effect on the primary end point of BCC development, but selenium instead increased the risk of SCC and total keratinocyte carcinomas (unadjusted RR, 1.27; 95% CI, 1.11–1.45).[45,46]

Beta carotene

In the Physicians’ Health Study, 21,884 male physicians with no reported history of BCC or SCC were randomly assigned to take 50 mg doses of daily oral beta carotene versus placebo in a 2 × 2 factorial trial of beta carotene and aspirin.[47] After 12 years, there was no difference in incidence of either BCC or SCC between the beta carotene and placebo groups. Similar findings were noted in 10 years and 14 years of follow-up among the participants in the Nurses’ Health Study and the Health Professionals Follow-up Study.[48] Reanalysis of data from these two cohorts after an additional 16 years of follow-up noted higher intake of some carotenoids was associated with a modest reduction in SCC risk.[49] Data on the use of sun protection behaviors were not available, and as participants with higher intake tended to have higher levels of physical activity, lower smoking, and alcohol consumption, it is possible that there was a confounding effect of sun protection behaviors. RCTs of long-term treatment with beta carotene in individuals previously treated for keratinocyte carcinoma also showed no benefit in preventing the occurrence of new keratinocyte carcinomas.[26,50]

Several RCTs show that beta carotene supplementation can increase cardiovascular disease mortality and increase the risk of lung cancer.[51,52]

Alpha-difluoromethylornithine (DFMO)

An RCT of oral DFMO (500 mg/m2/day) versus placebo for up to 5 years (n = 250 participants) showed no difference in the number of new keratinocyte carcinomas.[53] A subset analysis showed a difference in BCC events favoring the DFMO group (0.28 vs. 0.40 per year; P = .03) but no difference in SCC rates. However, the DFMO group experienced greater hearing loss than the placebo group (4 dB vs. 2 dB, P = .003), resulting in a higher study drug discontinuation rate (10.8% vs. 4.5%).

References
  1. Henrikson NB, Morrison CC, Blasi PR, et al.: Behavioral Counseling for Skin Cancer Prevention: Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 319 (11): 1143-1157, 2018. [PUBMED Abstract]
  2. Crane LA, Deas A, Mokrohisky ST, et al.: A randomized intervention study of sun protection promotion in well-child care. Prev Med 42 (3): 162-70, 2006. [PUBMED Abstract]
  3. Glasser A, Shaheen M, Glenn BA, et al.: The sun sense study: an intervention to improve sun protection in children. Am J Health Behav 34 (4): 500-10, 2010 Jul-Aug. [PUBMED Abstract]
  4. Norman GJ, Adams MA, Calfas KJ, et al.: A randomized trial of a multicomponent intervention for adolescent sun protection behaviors. Arch Pediatr Adolesc Med 161 (2): 146-52, 2007. [PUBMED Abstract]
  5. Crane LA, Asdigian NL, Barón AE, et al.: Mailed intervention to promote sun protection of children: a randomized controlled trial. Am J Prev Med 43 (4): 399-410, 2012. [PUBMED Abstract]
  6. Glanz K, Steffen AD, Schoenfeld E, et al.: Randomized trial of tailored skin cancer prevention for children: the Project SCAPE family study. J Health Commun 18 (11): 1368-83, 2013. [PUBMED Abstract]
  7. Youl PH, Soyer HP, Baade PD, et al.: Can skin cancer prevention and early detection be improved via mobile phone text messaging? A randomised, attention control trial. Prev Med 71: 50-6, 2015. [PUBMED Abstract]
  8. Prochaska JO, Velicer WF, Redding C, et al.: Stage-based expert systems to guide a population of primary care patients to quit smoking, eat healthier, prevent skin cancer, and receive regular mammograms. Prev Med 41 (2): 406-16, 2005. [PUBMED Abstract]
  9. Glanz K, Schoenfeld ER, Steffen A: A randomized trial of tailored skin cancer prevention messages for adults: Project SCAPE. Am J Public Health 100 (4): 735-41, 2010. [PUBMED Abstract]
  10. Janda M, Neale RE, Youl P, et al.: Impact of a video-based intervention to improve the prevalence of skin self-examination in men 50 years or older: the randomized skin awareness trial. Arch Dermatol 147 (7): 799-806, 2011. [PUBMED Abstract]
  11. Walton AE, Janda M, Youl PH, et al.: Uptake of skin self-examination and clinical examination behavior by outdoor workers. Arch Environ Occup Health 69 (4): 214-22, 2014. [PUBMED Abstract]
  12. Glazebrook C, Garrud P, Avery A, et al.: Impact of a multimedia intervention “Skinsafe” on patients’ knowledge and protective behaviors. Prev Med 42 (6): 449-54, 2006. [PUBMED Abstract]
  13. Heckman CJ, Darlow SD, Ritterband LM, et al.: Efficacy of an Intervention to Alter Skin Cancer Risk Behaviors in Young Adults. Am J Prev Med 51 (1): 1-11, 2016. [PUBMED Abstract]
  14. Mahler HI, Kulik JA, Gerrard M, et al.: Long-term effects of appearance-based interventions on sun protection behaviors. Health Psychol 26 (3): 350-60, 2007. [PUBMED Abstract]
  15. Weinstock MA, Risica PM, Martin RA, et al.: Melanoma early detection with thorough skin self-examination: the “Check It Out” randomized trial. Am J Prev Med 32 (6): 517-24, 2007. [PUBMED Abstract]
  16. Lin SW, Wheeler DC, Park Y, et al.: Prospective study of ultraviolet radiation exposure and risk of cancer in the United States. Int J Cancer 131 (6): E1015-23, 2012. [PUBMED Abstract]
  17. Lazovich D, Vogel RI, Berwick M, et al.: Melanoma risk in relation to use of sunscreen or other sun protection methods. Cancer Epidemiol Biomarkers Prev 20 (12): 2583-93, 2011. [PUBMED Abstract]
  18. Veierød MB, Couto E, Lund E, et al.: Host characteristics, sun exposure, indoor tanning and risk of squamous cell carcinoma of the skin. Int J Cancer 135 (2): 413-22, 2014. [PUBMED Abstract]
  19. Ferrucci LM, Vogel RI, Cartmel B, et al.: Indoor tanning in businesses and homes and risk of melanoma and nonmelanoma skin cancer in 2 US case-control studies. J Am Acad Dermatol 71 (5): 882-7, 2014. [PUBMED Abstract]
  20. Weinstock MA, Lott JP, Wang Q, et al.: Skin biopsy utilization and melanoma incidence among Medicare beneficiaries. Br J Dermatol 176 (4): 949-954, 2017. [PUBMED Abstract]
  21. Committee opinion no. 626: the transition from pediatric to adult health care: preventive care for young women aged 18-26 years. Obstet Gynecol 125 (3): 752-754, 2015. [PUBMED Abstract]
  22. American Academy of Dermatology: Skin Cancer. Washington, DC: American Academy of Dermatology Association, 2020. Available online. Last accessed April 8, 2025.
  23. Grossman DC, Curry SJ, Owens DK, et al.: Behavioral Counseling to Prevent Skin Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 319 (11): 1134-1142, 2018. [PUBMED Abstract]
  24. Naylor MF, Boyd A, Smith DW, et al.: High sun protection factor sunscreens in the suppression of actinic neoplasia. Arch Dermatol 131 (2): 170-5, 1995. [PUBMED Abstract]
  25. Thompson SC, Jolley D, Marks R: Reduction of solar keratoses by regular sunscreen use. N Engl J Med 329 (16): 1147-51, 1993. [PUBMED Abstract]
  26. Green A, Williams G, Neale R, et al.: Daily sunscreen application and betacarotene supplementation in prevention of basal-cell and squamous-cell carcinomas of the skin: a randomised controlled trial. Lancet 354 (9180): 723-9, 1999. [PUBMED Abstract]
  27. van der Pols JC, Williams GM, Pandeya N, et al.: Prolonged prevention of squamous cell carcinoma of the skin by regular sunscreen use. Cancer Epidemiol Biomarkers Prev 15 (12): 2546-8, 2006. [PUBMED Abstract]
  28. Green AC, Williams GM, Logan V, et al.: Reduced melanoma after regular sunscreen use: randomized trial follow-up. J Clin Oncol 29 (3): 257-63, 2011. [PUBMED Abstract]
  29. Dennis LK, Beane Freeman LE, VanBeek MJ: Sunscreen use and the risk for melanoma: a quantitative review. Ann Intern Med 139 (12): 966-78, 2003. [PUBMED Abstract]
  30. de Maleissye MF, Beauchet A, Saiag P, et al.: Sunscreen use and melanocytic nevi in children: a systematic review. Pediatr Dermatol 30 (1): 51-9, 2013 Jan-Feb. [PUBMED Abstract]
  31. Elmets CA, Viner JL, Pentland AP, et al.: Chemoprevention of nonmelanoma skin cancer with celecoxib: a randomized, double-blind, placebo-controlled trial. J Natl Cancer Inst 102 (24): 1835-44, 2010. [PUBMED Abstract]
  32. Solomon SD, McMurray JJ, Pfeffer MA, et al.: Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med 352 (11): 1071-80, 2005. [PUBMED Abstract]
  33. Bhala N, Emberson J, Merhi A, et al.: Vascular and upper gastrointestinal effects of non-steroidal anti-inflammatory drugs: meta-analyses of individual participant data from randomised trials. Lancet 382 (9894): 769-79, 2013. [PUBMED Abstract]
  34. Surjana D, Halliday GM, Martin AJ, et al.: Oral nicotinamide reduces actinic keratoses in phase II double-blinded randomized controlled trials. J Invest Dermatol 132 (5): 1497-500, 2012. [PUBMED Abstract]
  35. Allen NC, Martin AJ, Snaidr VA, et al.: Nicotinamide for Skin-Cancer Chemoprevention in Transplant Recipients. N Engl J Med 388 (9): 804-812, 2023. [PUBMED Abstract]
  36. Chen AC, Martin AJ, Choy B, et al.: A Phase 3 Randomized Trial of Nicotinamide for Skin-Cancer Chemoprevention. N Engl J Med 373 (17): 1618-26, 2015. [PUBMED Abstract]
  37. Nijsten TE, Stern RS: Oral retinoid use reduces cutaneous squamous cell carcinoma risk in patients with psoriasis treated with psoralen-UVA: a nested cohort study. J Am Acad Dermatol 49 (4): 644-50, 2003. [PUBMED Abstract]
  38. DiGiovanna JJ: Retinoid chemoprevention in patients at high risk for skin cancer. Med Pediatr Oncol 36 (5): 564-7, 2001. [PUBMED Abstract]
  39. Moon TE, Levine N, Cartmel B, et al.: Effect of retinol in preventing squamous cell skin cancer in moderate-risk subjects: a randomized, double-blind, controlled trial. Southwest Skin Cancer Prevention Study Group. Cancer Epidemiol Biomarkers Prev 6 (11): 949-56, 1997. [PUBMED Abstract]
  40. Kraemer KH, DiGiovanna JJ, Moshell AN, et al.: Prevention of skin cancer in xeroderma pigmentosum with the use of oral isotretinoin. N Engl J Med 318 (25): 1633-7, 1988. [PUBMED Abstract]
  41. Kraemer KH, DiGiovanna JJ, Peck GL: Chemoprevention of skin cancer in xeroderma pigmentosum. J Dermatol 19 (11): 715-8, 1992. [PUBMED Abstract]
  42. McKenna DB, Murphy GM: Skin cancer chemoprophylaxis in renal transplant recipients: 5 years of experience using low-dose acitretin. Br J Dermatol 140 (4): 656-60, 1999. [PUBMED Abstract]
  43. Harwood CA, Leedham-Green M, Leigh IM, et al.: Low-dose retinoids in the prevention of cutaneous squamous cell carcinomas in organ transplant recipients: a 16-year retrospective study. Arch Dermatol 141 (4): 456-64, 2005. [PUBMED Abstract]
  44. Weinstock MA, Bingham SF, Digiovanna JJ, et al.: Tretinoin and the prevention of keratinocyte carcinoma (Basal and squamous cell carcinoma of the skin): a veterans affairs randomized chemoprevention trial. J Invest Dermatol 132 (6): 1583-90, 2012. [PUBMED Abstract]
  45. Clark LC, Combs GF, Turnbull BW, et al.: Effects of selenium supplementation for cancer prevention in patients with carcinoma of the skin. A randomized controlled trial. Nutritional Prevention of Cancer Study Group. JAMA 276 (24): 1957-63, 1996. [PUBMED Abstract]
  46. Duffield-Lillico AJ, Slate EH, Reid ME, et al.: Selenium supplementation and secondary prevention of nonmelanoma skin cancer in a randomized trial. J Natl Cancer Inst 95 (19): 1477-81, 2003. [PUBMED Abstract]
  47. Frieling UM, Schaumberg DA, Kupper TS, et al.: A randomized, 12-year primary-prevention trial of beta carotene supplementation for nonmelanoma skin cancer in the physician’s health study. Arch Dermatol 136 (2): 179-84, 2000. [PUBMED Abstract]
  48. Fung TT, Spiegelman D, Egan KM, et al.: Vitamin and carotenoid intake and risk of squamous cell carcinoma of the skin. Int J Cancer 103 (1): 110-5, 2003. [PUBMED Abstract]
  49. Kim J, Park MK, Li WQ, et al.: Association of Vitamin A Intake With Cutaneous Squamous Cell Carcinoma Risk in the United States. JAMA Dermatol 155 (11): 1260-1268, 2019. [PUBMED Abstract]
  50. Greenberg ER, Baron JA, Stukel TA, et al.: A clinical trial of beta carotene to prevent basal-cell and squamous-cell cancers of the skin. The Skin Cancer Prevention Study Group. N Engl J Med 323 (12): 789-95, 1990. [PUBMED Abstract]
  51. Omenn GS, Goodman GE, Thornquist MD, et al.: Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med 334 (18): 1150-5, 1996. [PUBMED Abstract]
  52. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. N Engl J Med 330 (15): 1029-35, 1994. [PUBMED Abstract]
  53. Bailey HH, Kim K, Verma AK, et al.: A randomized, double-blind, placebo-controlled phase 3 skin cancer prevention study of {alpha}-difluoromethylornithine in subjects with previous history of skin cancer. Cancer Prev Res (Phila) 3 (1): 35-47, 2010. [PUBMED Abstract]

Latest Updates to This Summary (04/08/2025)

The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

Incidence and Mortality of Skin Cancer

Added American Cancer Society as reference 1.

Updated statistics with estimated new cases and deaths of melanoma 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 skin cancer prevention. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.

Reviewers and Updates

This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

Board members review recently published articles each month to determine whether an article should:

  • be discussed at a meeting,
  • be cited with text, or
  • replace or update an existing article that is already cited.

Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

Levels of Evidence

Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

Permission to Use This Summary

PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”

The preferred citation for this PDQ summary is:

PDQ® Screening and Prevention Editorial Board. PDQ Skin Cancer Prevention. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/skin/hp/skin-prevention-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389494]

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.

Mantle Cell Lymphoma Treatment (PDQ®)–Health Professional Version

Mantle Cell Lymphoma Treatment (PDQ®)–Health Professional Version

General Information About Mantle Cell Lymphoma

Incidence

Mantle cell lymphoma (MCL) is a less common type of B-cell non-Hodgkin lymphoma (NHL). With about 4,000 new cases each year,[1] MCL accounts for about 5% of all NHLs in the United States. The median age at diagnosis is approximately 65 years, with most cases occurring in men.

Anatomy

NHL usually originates in lymphoid tissues.

EnlargeLymphatic system; drawing shows the lymph vessels and lymph organs, including the lymph nodes, tonsils, thymus, spleen, and bone marrow. Also shown is the small intestine (one site of mucosal-associated lymphoid tissue). There are also two pullouts: one showing a close up of the inside structure of a lymph node and the attached artery, vein, and lymph vessels with arrows showing how the lymph (clear, watery fluid) moves into and out of the lymph node, and another showing a close up of bone marrow with blood cells.
The lymph system is part of the body’s immune system and is made up of tissues and organs that help protect the body from infection and disease. These include the tonsils, adenoids (not shown), thymus, spleen, bone marrow, lymph vessels, and lymph nodes. Lymph tissue is also found in many other parts of the body, including the small intestine.

Clinical Features

MCL presents in the lymph nodes, spleen, bone marrow, and sometimes as gastrointestinal polyposis (especially in the colon).[1] Most patients with MCL have stage III or IV disease at diagnosis. Like low-grade lymphomas, MCL is highly responsive to treatment, but not curable in most cases. MCL is characterized by CD5- and CD20-positive B cells derived from the mantle region of the lymphoid follicle, most often with a translocation of chromosomes 11 and 14 (t(11;14)(q13;q32)), resulting in overexpression of cyclin D1.[2] Histopathology typically shows CD5-positive, CD20-positive, cyclin D1-positive, CD10-negative, and CD23-negative or low disease. More than 95% of cases are cyclin D1-positive MCL with a classic IGH::CCND1 fusion.[3] Rarely, the free kappa (chromosome 2) or free lambda (chromosome 22) enhancer may partner with CCND1, and other times CCND2, CCND3, or CCNE may be the rearrangement partner.

MCL may be divided into two clinical subtypes: indolent (often a non-nodal leukemic version) and aggressive (a nodal version).[2]

Indolent MCL

The more indolent version occurs in 20% of patients with MCL and is also called indolent non-nodal leukemia. Indolent MCL characteristics include:[2]

  • Small (<3 cm) lymph nodes.
  • Leukemic presentation.
  • Early stage.
  • Lack of constitutional B symptoms (fever, recurrent night sweats, or weight loss).
  • Negative or low (<10%) SOX11 expression.
  • Hypermutation of IGHV.
  • CD23 and CD20 positivity.
  • Absence of ATM or CCND1 variants or deletions.

Isolated gastrointestinal polyposis also has an indolent course. These patients have a significantly better prognosis (with a median survival exceeding 15 years), and many can defer therapy on initial presentation and be followed with a watchful waiting approach (as is done with other indolent lymphomas, such as follicular lymphoma).[46]

Aggressive MCL

Most patients with MCL (80%) present with more aggressive disease, which is also called aggressive nodal leukemia. Patients have a median survival exceeding 8 to 10 years. Aggressive MCL characteristics include:[2,7]

  • Extensive enlarged lymph nodes.
  • Rapid progression.
  • Constitutional B symptoms.
  • High (≥10%) SOX11 expression.
  • Unmutated IGHV.
  • CCND1 or ATM variants or deletions, or other genomic complexity.

Patients with a worse prognosis (median survival, 4 to 7 years) can be identified by the presence of blastoid or pleomorphic variants by microscopy, a high Ki-67 (≥30%), and TP53 variants or deletions.[2,3,811] Age and comorbidities may impact the prognosis and treatment options for any patient with aggressive MCL. Any patient with indolent or moderately aggressive MCL may later convert to a blastoid or TP53 variant/deletion phenotype, which is resistant to treatment due to genomic instability or selection of resistant clones through destruction of the predominant sensitive cells after prior therapy.[8,9] Standard chemoimmunotherapy is particularly ineffective for patients with TP53 pathogenic variants. Targeted therapies for the B-cell receptor (Bruton tyrosine kinase inhibitors), surface antigens (like chimeric antigen receptor T-cell and bispecific antibodies), and BCL-2 inhibitors are more applicable.

Prognosis

MCL is not considered curable in the standard sense because eventual relapse is a certainty. However, many older patients achieve a functional cure, surviving until death from other causes while in MCL remission. There is no evidence that the distinction between the nodal and non-nodal subtypes maintains its relevance in patients with multiply relapsed or refractory disease.

References
  1. Armitage JO, Longo DL: Mantle-Cell Lymphoma. N Engl J Med 386 (26): 2495-2506, 2022. [PUBMED Abstract]
  2. Campo E, Jaffe ES, Cook JR, et al.: The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood 140 (11): 1229-1253, 2022. [PUBMED Abstract]
  3. Alaggio R, Amador C, Anagnostopoulos I, et al.: The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 36 (7): 1720-1748, 2022. [PUBMED Abstract]
  4. Clot G, Jares P, Giné E, et al.: A gene signature that distinguishes conventional and leukemic nonnodal mantle cell lymphoma helps predict outcome. Blood 132 (4): 413-422, 2018. [PUBMED Abstract]
  5. Fenske TS: Frontline Therapy in Mantle Cell Lymphoma: When Clinical Trial and Real-World Data Collide. J Clin Oncol 41 (3): 452-459, 2023. [PUBMED Abstract]
  6. Cohen JB, Han X, Jemal A, et al.: Deferred therapy is associated with improved overall survival in patients with newly diagnosed mantle cell lymphoma. Cancer 122 (15): 2356-63, 2016. [PUBMED Abstract]
  7. Greenwell IB, Staton AD, Lee MJ, et al.: Complex karyotype in patients with mantle cell lymphoma predicts inferior survival and poor response to intensive induction therapy. Cancer 124 (11): 2306-2315, 2018. [PUBMED Abstract]
  8. Dreyling M, Klapper W, Rule S: Blastoid and pleomorphic mantle cell lymphoma: still a diagnostic and therapeutic challenge! Blood 132 (26): 2722-2729, 2018. [PUBMED Abstract]
  9. Jain P, Dreyling M, Seymour JF, et al.: High-Risk Mantle Cell Lymphoma: Definition, Current Challenges, and Management. J Clin Oncol 38 (36): 4302-4316, 2020. [PUBMED Abstract]
  10. Lew TE, Minson A, Dickinson M, et al.: Treatment approaches for patients with TP53-mutated mantle cell lymphoma. Lancet Haematol 10 (2): e142-e154, 2023. [PUBMED Abstract]
  11. Jain P, Wang M: High-risk MCL: recognition and treatment. Blood 145 (7): 683-695, 2025. [PUBMED Abstract]

Stage Information for Mantle Cell Lymphoma

Stage is important in selecting a treatment for patients with mantle cell lymphoma (MCL). Positron emission tomography–computed tomography (PET-CT) is usually part of the staging evaluation for all patients with lymphoma.

Patients with MCL commonly have involvement of the following sites:

  • Contiguous or noncontiguous lymph nodes.
  • Gastrointestinal tract, especially colonic polyposis.
  • Extranodal presentations.
  • Bone marrow.
  • Spleen.

Rarely, cytological examination of cerebrospinal fluid may be positive in patients with MCL. Lumbar puncture is not a typical staging procedure, but it is considered for patients with mantle cell blastoid variant or 17p deletion/TP53-altered disease.

Most patients with MCL present with advanced (stage III or stage IV) disease, often identified by PET-CT scans or biopsies of the bone marrow when indicated by PET positivity. In a retrospective review of over 32,000 cases of lymphoma in France, up to 40% of diagnoses were made by core needle biopsy, and 60% were made by excisional biopsy.[1] After expert review, core needle biopsy provided a definite diagnosis in 92.3% of cases; excisional biopsy provided a definite diagnosis in 98.1% of cases (P < .0001). Laparoscopic biopsy or laparotomy is not required for staging but rarely may be necessary to establish a diagnosis or histological type.[2]

PET-CT scans with fluorine F 18-fludeoxyglucose are used for initial staging and may also be used for follow-up after therapy.[3] Multiple studies have demonstrated that routine interim PET scans after two to four cycles of therapy do not provide reliable prognostic information in aggressive lymphomas, and they are not recommended for MCL.[47]

For patients with MCL, a positive PET result after therapy confers a worse prognosis. However, it is unclear whether a positive PET result is predictive when alternative therapy is implemented.[8]

Staging Subclassification System

Lugano classification

The American Joint Committee on Cancer (AJCC) has adopted the Lugano classification to evaluate and stage lymphoma.[9] The Lugano classification system replaces the Ann Arbor classification system, which was adopted in 1971 at the Ann Arbor Conference,[10] with some modifications 18 years later from the Cotswolds meeting.[11,12]

Table 1. Lugano Classification for Non-Hodgkin Lymphoma (Including Mantle Cell Lymphoma)a
Stage Stage Description Illustration
CSF = cerebrospinal fluid; MCL = mantle cell lymphoma; NHL = non-Hodgkin lymphoma.
aHodgkin and Non-Hodgkin Lymphomas. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. New York, NY: Springer, 2017, pp. 937–58.
bStage II bulky may be considered either early or advanced stage based on lymphoma histology and prognostic factors.
cThe definition of disease bulk varies according to lymphoma histology. Precise measurements have not been determined for MCL, and proposals range from ≥5 cm to ≥10 cm.
Limited stage
I Involvement of a single lymphatic site (i.e., nodal region, Waldeyer’s ring, thymus, or spleen).
EnlargeStage I adult lymphoma; drawing shows cancer in one lymph node group and in the spleen. Also shown are the Waldeyer’s ring and the thymus. An inset shows a lymph node with a lymph vessel, an artery, and a vein. Cancer cells are shown in the lymph node.
IE Single extralymphatic site in the absence of nodal involvement.  
II Involvement of two or more lymph node regions on the same side of the diaphragm.
EnlargeStage II adult lymphoma; drawing shows cancer in two lymph node groups above the diaphragm and below the diaphragm. An inset shows a lymph node with a lymph vessel, an artery, and a vein. Cancer cells are shown in the lymph node.
IIE Contiguous extralymphatic extension from a nodal site with or without involvement of other lymph node regions on the same side of the diaphragm.
EnlargeStage IIE adult lymphoma; drawing shows cancer that has spread from a group of lymph nodes to a nearby area. Also shown is a lung and the diaphragm. An inset shows a lymph node with a lymph vessel, an artery, and a vein. Cancer cells are shown in the lymph node.
II bulkyb Stage II with disease bulk.c  
Advanced stage
III Involvement of lymph node regions on both sides of the diaphragm; nodes above the diaphragm with spleen involvement.
EnlargeStage III adult lymphoma; drawing shows the right and left sides of the body. The right side of the body shows cancer in a group of lymph nodes above the diaphragm and below the diaphragm. The left side of the body shows cancer in a group of lymph nodes above the diaphragm and cancer in the spleen.
IV Diffuse or disseminated involvement of one or more extralymphatic organs, with or without associated lymph node involvement; or noncontiguous extralymphatic organ involvement in conjunction with nodal stage II disease; or any extralymphatic organ involvement in nodal stage III disease. Stage IV includes any involvement of the CSF, bone marrow, liver, or multiple lung lesions (other than by direct extension in stage IIE disease).
EnlargeStage IV adult lymphoma; drawing shows four panels: (a) the top left panel shows cancer in the liver; (b) the top right panel shows cancer in the left lung and in two groups of lymph nodes below the diaphragm; (c) the bottom left panel shows cancer in the left lung and in a group of lymph nodes above the diaphragm and below the diaphragm; and (d) the bottom right panel shows cancer in both lungs, the liver, and the bone marrow (pullout). Also shown is primary cancer in the lymph nodes and a pullout of the brain with cerebrospinal fluid (in blue).
Note: Hodgkin lymphoma uses A or B designation with stage group. A/B is no longer used in NHL.

Occasionally, specialized staging systems are used. The physician should be aware of the system used in a specific report.

The E designation is used when extranodal lymphoid malignancies arise in tissues separate from, but near, the major lymphatic aggregates. Stage IV refers to disease that is diffusely spread throughout an extranodal site, such as the liver. If pathological proof of involvement of one or more extralymphatic sites has been documented, the symbol for the site of involvement, followed by a plus sign (+), is listed.

Table 2. Notation to Identify Specific Sites
N = nodes H = liver L = lung M = bone marrow
S = spleen P = pleura O = bone D = skin

Current practice assigns a clinical stage based on the findings of the clinical evaluation and a pathological stage based on the findings from invasive procedures beyond the initial biopsy.

Several other factors that are not included in the above staging system are important for the staging and prognosis of patients with MCL. These factors include:

  • Age.
  • Performance status.
  • Tumor size.
  • Lactate dehydrogenase level.
  • The number of extranodal sites.
  • TP53 status (pathogenic variant or deletion).
  • Ki-67 cell proliferation rate.
  • Complex karyotype or gene expression.
  • CCND1 or ATM pathogenic variants or deletions.
  • Hypermutated or unmutated IGHV.
  • Beta-2 microglobulin.

MCL has demonstrated heterogeneous and variable clinical courses. Many prognostic factors have been identified, and mantle cell international prognostic scores have been devised. While these indicators help designate the need for therapy, they have not proven useful for selection of treatment. The one exception is the poor performance of standard chemotherapeutic agents with immunotherapy in patients with the highest-risk disease, as described previously.

References
  1. Syrykh C, Chaouat C, Poullot E, et al.: Lymph node excisions provide more precise lymphoma diagnoses than core biopsies: a French Lymphopath network survey. Blood 140 (24): 2573-2583, 2022. [PUBMED Abstract]
  2. Mann GB, Conlon KC, LaQuaglia M, et al.: Emerging role of laparoscopy in the diagnosis of lymphoma. J Clin Oncol 16 (5): 1909-15, 1998. [PUBMED Abstract]
  3. Barrington SF, Mikhaeel NG, Kostakoglu L, et al.: Role of imaging in the staging and response assessment of lymphoma: consensus of the International Conference on Malignant Lymphomas Imaging Working Group. J Clin Oncol 32 (27): 3048-58, 2014. [PUBMED Abstract]
  4. Horning SJ, Juweid ME, Schöder H, et al.: Interim positron emission tomography scans in diffuse large B-cell lymphoma: an independent expert nuclear medicine evaluation of the Eastern Cooperative Oncology Group E3404 study. Blood 115 (4): 775-7; quiz 918, 2010. [PUBMED Abstract]
  5. Moskowitz CH, Schöder H, Teruya-Feldstein J, et al.: Risk-adapted dose-dense immunochemotherapy determined by interim FDG-PET in Advanced-stage diffuse large B-Cell lymphoma. J Clin Oncol 28 (11): 1896-903, 2010. [PUBMED Abstract]
  6. Pregno P, Chiappella A, Bellò M, et al.: Interim 18-FDG-PET/CT failed to predict the outcome in diffuse large B-cell lymphoma patients treated at the diagnosis with rituximab-CHOP. Blood 119 (9): 2066-73, 2012. [PUBMED Abstract]
  7. Sun N, Zhao J, Qiao W, et al.: Predictive value of interim PET/CT in DLBCL treated with R-CHOP: meta-analysis. Biomed Res Int 2015: 648572, 2015. [PUBMED Abstract]
  8. Pyo J, Won Kim K, Jacene HA, et al.: End-therapy positron emission tomography for treatment response assessment in follicular lymphoma: a systematic review and meta-analysis. Clin Cancer Res 19 (23): 6566-77, 2013. [PUBMED Abstract]
  9. Hodgkin and non-Hodgkin lymphoma. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp. 937–58.
  10. Carbone PP, Kaplan HS, Musshoff K, et al.: Report of the Committee on Hodgkin’s Disease Staging Classification. Cancer Res 31 (11): 1860-1, 1971. [PUBMED Abstract]
  11. Lister TA, Crowther D, Sutcliffe SB, et al.: Report of a committee convened to discuss the evaluation and staging of patients with Hodgkin’s disease: Cotswolds meeting. J Clin Oncol 7 (11): 1630-6, 1989. [PUBMED Abstract]
  12. National Cancer Institute sponsored study of classifications of non-Hodgkin’s lymphomas: summary and description of a working formulation for clinical usage. The Non-Hodgkin’s Lymphoma Pathologic Classification Project. Cancer 49 (10): 2112-35, 1982. [PUBMED Abstract]

Treatment Option Overview for Mantle Cell Lymphoma

Once the diagnosis of mantle cell lymphoma (MCL) is established and staging is completed (usually with positron emission tomography–computed tomography, although colonoscopy, bone marrow biopsy, or lumbar puncture may be indicated in selected cases), laboratory testing is performed. This testing allows a distinction to be made among the indolent non-nodal leukemic subtype (20% of patients with MCL), the more aggressive nodal subtype, or the hyperaggressive blastoid or TP53-altered subtype which confers the worst prognosis. Clinical judgment may be required for patients with both indolent and aggressive features. For more information about laboratory testing, see the sections on Clinical Features and Stage Information for Mantle Cell Lymphoma.

Table 3. Treatment Options for Previously Untreated Mantle Cell Lymphoma (MCL)
MCL Subtype Treatment Options
BR = bendamustine and rituximab; BTK = Bruton tyrosine kinase; CAR = chimeric antigen receptor; R2 regimen = rituximab and lenalidomide; R-CHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone; R-DHAP = rituximab, dexamethasone, high-dose cytarabine, and cisplatin; R-HCVAD = rituximab plus hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone; R-MA = rituximab, methotrexate, and cytarabine.
Non-nodal leukemic subtype: Early-stage (IA, IIA) stable or slowly progressing disease Watchful waiting
Radiation therapy
Anti-CD20 monoclonal antibodies (rituximab, obinutuzumab)
BTK inhibitors (acalabrutinib)
R2 regimen
BR
Nodal aggressive subtype: Advanced-stage lymph nodes (stage IIB, III, IV) BTK inhibitors with or without anti-CD20 monoclonal antibodies
Chemoimmunotherapy (BR; R-CHOP/R-DHAP; R-HCVAD/R-MA)
R2 regimen
Rituximab alone (in older patients with comorbidities who cannot take BTK inhibitors)
Hyperaggressive blastoid or TP53-altered subtype BTK inhibitor + rituximab (or obinutuzumab) + BCL-2 inhibitor (venetoclax)
Consolidation with T-cell directed therapy such as allogeneic stem cell transplant, CAR T-cell therapy, or bispecific antibody therapy
Clinical trials

Before beginning systemic therapy, patients should be screened for active hepatitis B, hepatitis C, or HIV.[1] Patients with detectable hepatitis B virus (HBV) benefit from prophylaxis with entecavir if their treatment plan includes rituximab, Bruton tyrosine kinase (BTK) inhibitors, or chemoimmunotherapy. Patients with a resolved HBV infection (defined as hepatitis B surface antigen-negative but hepatitis B core antibody-positive) are at risk of reactivation of HBV and require active monitoring of HBV DNA. For patients who received rituximab or obinutuzumab therapy, the risk of reactivation was 10% to 15%; prophylaxis reduced this risk to 2% in a retrospective study.[2] Similarly, prophylaxis for herpes zoster with valacyclovir or acyclovir and prophylaxis for pneumocystis with trimethoprim/sulfa or dapsone are usually given to all patients receiving systemic therapy.

Summary of Therapy for MCL

The following agents, alone or in combination, represent targeted biological therapy options that may enable chemotherapy-free treatment strategies for most patients with MCL:[3]

  • BTK inhibitors: acalabrutinib, zanubrutinib, ibrutinib, and pirtobrutinib.
  • Anti-CD20 monoclonal antibodies: rituximab and obinutuzumab.
  • BCL-2 inhibitor: venetoclax.
  • Immune stimulator: lenalidomide.

Chemoimmunotherapy with BR (bendamustine and rituximab) or with R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone)/R-DHAP (rituximab, dexamethasone, high-dose cytarabine, and cisplatin) is still considered a standard option for younger fit patients. Most patients can avoid autologous stem cell transplant (SCT), but maintenance therapy with rituximab for at least 2 to 3 years remains the standard of care after first-line induction therapy. The highest-risk patients may best respond to combinations of biological targeted therapies, followed by consolidation with allogeneic SCT or chimeric antigen receptor T-cell therapy.[4,5]

Routine administration of central nervous system (CNS) prophylaxis in patients with high-risk MCL has never been studied in a prospective randomized trial. The use of intrathecal or intravenous high-dose methotrexate or the use of systemic therapies with CNS penetration such as BTK inhibitors, high-dose cytarabine, or venetoclax have not been studied or proven efficacious in this situation.[6]

Outside the context of clinical trials, the use of measurable residual disease (MRD) testing has not been shown to be predictive for directing therapy for patients with MCL. In a retrospective analysis of a prospective randomized clinical trial, while MRD negativity after rituximab maintenance therapy was prognostic for a better outcome, continuation of maintenance rituximab prolonged progression-free survival and overall survival the most among patients with MRD-negative disease.[7][Level of evidence C1] Stopping maintenance rituximab was not indicated in patients with MRD-negative disease, negating any possible change in therapy based on that status.

References
  1. Dong HJ, Ni LN, Sheng GF, et al.: Risk of hepatitis B virus (HBV) reactivation in non-Hodgkin lymphoma patients receiving rituximab-chemotherapy: a meta-analysis. J Clin Virol 57 (3): 209-14, 2013. [PUBMED Abstract]
  2. Kusumoto S, Arcaini L, Hong X, et al.: Risk of HBV reactivation in patients with B-cell lymphomas receiving obinutuzumab or rituximab immunochemotherapy. Blood 133 (2): 137-146, 2019. [PUBMED Abstract]
  3. Martin P, Ruan J, Leonard JP: The potential for chemotherapy-free strategies in mantle cell lymphoma. Blood 130 (17): 1881-1888, 2017. [PUBMED Abstract]
  4. Fenske TS, Zhang MJ, Carreras J, et al.: Autologous or reduced-intensity conditioning allogeneic hematopoietic cell transplantation for chemotherapy-sensitive mantle-cell lymphoma: analysis of transplantation timing and modality. J Clin Oncol 32 (4): 273-81, 2014. [PUBMED Abstract]
  5. Jain P, Wang M: High-risk MCL: recognition and treatment. Blood 145 (7): 683-695, 2025. [PUBMED Abstract]
  6. Jain P, Dreyling M, Seymour JF, et al.: High-Risk Mantle Cell Lymphoma: Definition, Current Challenges, and Management. J Clin Oncol 38 (36): 4302-4316, 2020. [PUBMED Abstract]
  7. Hoster E, Delfau-Larue MH, Macintyre E, et al.: Predictive Value of Minimal Residual Disease for Efficacy of Rituximab Maintenance in Mantle Cell Lymphoma: Results From the European Mantle Cell Lymphoma Elderly Trial. J Clin Oncol 42 (5): 538-549, 2024. [PUBMED Abstract]

Treatment of Indolent Mantle Cell Lymphoma

Asymptomatic patients with indolent mantle cell lymphoma (MCL), a low burden of lymphadenopathy, and no significant splenomegaly or cytopenias may benefit from a watchful waiting approach. This approach has been demonstrated in several retrospective series.[1,2][Level of evidence C3] Because most patients with MCL are older than 60 years and MCL is not treated with curative intent, the quality-of life impact of treatment-related toxicities (both physical and financial) must be considered.

When patients with indolent MCL require therapy, a lower-intensity approach is preferred, but there is no standard approach due to the lack of clinical trials for this subgroup.

Induction chemotherapy regimens may be used for symptomatic progressive disease. These regimens range in intensity from rituximab alone to rituximab plus acalabrutinib (or ibrutinib or zanubrutinib), rituximab plus lenalidomide (R2), or bendamustine and rituximab (BR).

A prospective randomized trial included 373 patients with previously untreated MCL. A total of 87% of patients were aged 60 years or older. The study compared (1) BR versus BR plus bortezomib (BVR) as induction regimens and (2) rituximab versus R2 as maintenance regimens.[3]

  • With a median follow-up of 7.5 years, there was no difference in the median progression-free survival (PFS) for patients who received BR compared with patients who received BVR (5.5 vs. 6.4 years; hazard ratio [HR], 0.90; 90% confidence interval [CI], 0.70–1.16).[3][Level of evidence B1]
  • Independent of the induction therapy, there was no difference in the median PFS for patients who received rituximab versus patients who received R2 (5.9 vs. 7.2 years; HR, 0.84; 90% CI, 0.62–1.15).[3][Level of evidence B1]

Summary: BR induction therapy followed by maintenance therapy with rituximab remains a standard-of-care option. No benefit was noted in trials that incorporated early bortezomib or lenalidomide in the standard option. However, for many patients with indolent MCL, BR can be avoided by starting with rituximab alone and adding a Bruton tyrosine kinase inhibitor (such as acalabrutinib, ibrutinib, or zanubrutinib) if the response is not adequate after 4 to 8 weeks of therapy.

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. Martin P, Chadburn A, Christos P, et al.: Outcome of deferred initial therapy in mantle-cell lymphoma. J Clin Oncol 27 (8): 1209-13, 2009. [PUBMED Abstract]
  2. Cohen JB, Han X, Jemal A, et al.: Deferred therapy is associated with improved overall survival in patients with newly diagnosed mantle cell lymphoma. Cancer 122 (15): 2356-63, 2016. [PUBMED Abstract]
  3. Smith MR, Jegede OA, Martin P, et al.: Randomized study of induction with bendamustine-rituximab ± bortezomib and maintenance with rituximab ± lenalidomide for MCL. Blood 144 (10): 1083-1092, 2024. [PUBMED Abstract]

Treatment of Aggressive Mantle Cell Lymphoma

Clinical trials have not determined which therapeutic option offers the best long-term survival for patients with previously untreated mantle cell lymphoma (MCL). The situation is unclear because MCL is a relatively rare disease (4,000 new cases per year in the United States), and study evidence has accrued slowly over the past decade. A historical perspective may help to explain the state of the evidence.

MCL was first described in the 1980s as a distinct entity from small lymphocytic lymphoma/chronic lymphocytic lymphoma or follicular lymphoma. When treated with oral alkylators and infusional cytotoxic agents available at the time, MCL appeared to relapse sooner and more frequently than other indolent lymphomas. When purine analogues also proved ineffective in the 1990s, MCL was viewed as an aggressive lymphoma without a discernible cure. This view ultimately led to an aggressive treatment paradigm that incorporated all available modalities in the early 2000s: R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) followed by high-dose cytarabine with or without a platinum agent, plus autologous stem cell transplant (SCT) plus rituximab maintenance.[1,2]

Chemoimmunotherapy

Evidence (chemoimmunotherapy):

  1. In a prospective trial, 560 patients older than 60 years and not eligible for SCT were randomly assigned to receive induction therapy with either R-CHOP or R-FC (rituximab, fludarabine, cyclophosphamide) for six to eight cycles. Patients with disease response (n = 316) were then randomly assigned to receive maintenance therapy with either rituximab or interferon alfa.[3]
    • Focusing on the randomized induction therapy (n = 560), with a median follow-up of 7.6 years, the median overall survival (OS) was 6.4 years in the R-CHOP group and 3.9 years in the R-FC group (P = .0054).[3][Level of evidence A1]
    • Focusing on the randomized maintenance therapy (n = 316 responders), with a median follow-up of 8 years, the median OS was 9.8 years in the rituximab group and 7.1 years in the interferon alfa group (P = .009).[3][Level of evidence A1]
  2. A randomized trial compared bendamustine and rituximab (BR) with R-CHOP. Progression free-survival (PFS) improved in patients who received BR (35 months) compared with patients who received R-CHOP (22 months) (hazard ratio [HR], 0.49; 95% confidence interval [CI], 0.28–0.79; P = .004). There was no difference in OS.[4][Level of evidence B1]
    • This trial failed to show any benefit for rituximab maintenance therapy after BR.
  3. A prospective randomized trial of 487 patients compared VR-CAP (bortezomib, rituximab, cyclophosphamide, doxorubicin, prednisone) with R-CHOP.[5]
    • With a median follow-up of 82 months, the median OS was longer in the VR-CAP group (90.7 months) than in the R-CHOP group (55.7 months) (HR, 0.66; 95% CI, 0.51−0.85; P = .001).[5][Level of evidence A1]
  4. A prospective randomized trial from the European MCL Network included 497 patients younger than 65 years. The trial compared six cycles of R-CHOP with six cycles of alternating R-CHOP and R-DHAP (rituximab, dexamethasone, cytarabine, and cisplatin), with both groups then receiving autologous SCT.[1,2][Level of evidence B1]
    • With a median follow-up of 10.6 years, the 10-year PFS rate was 73% for patients who received R-DHAP and 57% for patients who received R-CHOP (HR, 0.56; P = .038). There was no difference in the 10-year OS rates (60% [R-DHAP] vs. 55% [R-CHOP]; HR, 0.80; 95% CI, 0.61–1.06; P = .12).[6][Level of evidence B1]

    This trial is often referenced by subsequent articles to assert a role for cytarabine in induction therapy, but the ultimate lack of survival advantage casts doubt on this assertion. This regimen is clearly not applicable for older, less fit patients with comorbidities who are not eligible for transplant.

Summary: By the 2010s, the standard of care for older patients with comorbidities was the BR regimen. For younger fit patients with MCL, the standard of care was R-CHOP/R-DHAP, followed by autologous SCT consolidation and rituximab maintenance therapy, based on the results of the trial by the European MCL Network.[1,2]

Since 2015, clinical trials have focused on the necessity of autologous SCT consolidation and the use of high-dose cytarabine during induction therapy. As a result of randomized trials that incorporated the Bruton tyrosine kinase (BTK) inhibitors ibrutinib or acalabrutinib, sufficient evidence exists to avoid autologous SCT and high-dose cytarabine in most patients.

Treatment Options to Avoid Autologous SCT Consolidation

Evidence (treatment options to avoid autologous SCT consolidation):

  1. The prospective, randomized TRIANGLE trial (NCT02858258) trial included 870 patients aged 65 years or younger with previously untreated MCL who were fit enough for autologous SCT. There were three study arms. The primary end point was failure-free survival (FFS), and the secondary end point was OS.[7,8] The three arms included:
    1. Arm A: Standard therapy at the start of the trial with R-CHOP/R-DHAP followed by autologous SCT consolidation and rituximab maintenance therapy.
    2. Arm A+I: Ibrutinib plus R-CHOP/R-DHAP followed by autologous SCT consolidation and ibrutinib plus rituximab maintenance therapy.
    3. Arm I: Ibrutinib plus R-CHOP/R-DHAP with no consolidation and ibrutinib plus rituximab maintenance therapy.
    • With a median follow-up of 53 months, results were published in abstract form. The 3-year OS rate was 90% for arm A+I versus 85% for arm A (HR, 0.61; P = .0069), and the 3-year OS rate was 91% for arm I versus 85% for arm A (HR, 0.59; P = .0041).[8][Level of evidence A1]
    • Also published in abstract form, after a median follow-up of 53 months, the role of autologous SCT was evaluated by comparing the FFS of arm A+I (with autologous SCT) with the FFS of arm I (without autologous SCT) (86% vs. 85%, respectively; HR, 0.86; P = .56), which was not significantly different.[8][Level of evidence B1]
    • Toxicity was highest in the autologous SCT arms, as expected.

    Summary: When patients received ibrutinib and a high-dose chemoimmunotherapy regimen (including cytarabine), the addition of ibrutinib led to superior outcomes by 5% for OS. Autologous SCT did not add efficacy to the ibrutinib-containing regimens but did add toxicity.

  2. The ECOG 4151 trial (NCT03267433), published in abstract form, included 650 patients younger than 71 years with previously untreated MCL who were eligible for autologous SCT. The trial allowed any standard induction therapy regimen. Most patients received R-CHOP or R-DHAP induction therapy, and 27% received BR. All patients received rituximab maintenance therapy. Patients found to have measurable residual disease (MRD)–negative MCL in the blood and marrow (80%, n = 516) were randomly assigned to receive either autologous SCT plus 3 years of maintenance therapy with rituximab or 3 years of maintenance therapy with rituximab alone.[9]
    • With a median follow-up of 42 months, there was no difference in 3-year OS rates, at 82.1% for patients who received autologous SCT and 82.7% for patients who did not receive autologous SCT (HR, 1.11; 95% CI, 0.71–1.74; P = .66). This OS HR crossed the futility boundary.[9][Level of evidence A1]

    Summary: With the introduction of ibrutinib and other BTK inhibitors that can be used during induction therapy, maintenance therapy, or at relapse, most patients can avoid autologous SCT.

  3. A retrospective analysis included 1,265 patients aged 65 years or younger with MCL who were transplant-eligible. The analysis showed no benefit for autologous SCT in time-to-next treatment (HR, 0.84; 95% CI, 0.68–1.03) or OS (HR, 0.86; 95% CI, 0.63–1.18).[10][Level of evidence C3]

Treatment Options to Avoid High-Dose Cytarabine

Evidence (treatment options to avoid high-dose cytarabine):

  1. A three-arm prospective randomized trial, published in abstract form, included 359 patients with previously untreated MCL. The trial evaluated complete remission rates after induction therapy (defined as a complete metabolic response by positron emission tomography–computed tomography, undetectable MRD by blood and bone marrow, and PFS).[11] The three treatment arms included:
    1. BR for three cycles plus CR (high-dose cytarabine plus rituximab).
    2. BR plus CR plus A (acalabrutinib).
    3. BR plus A (omitting cytarabine).
    • With a median follow-up of 27.9 months, the 1-year PFS rate was 86% for BR plus CR, 89% for BR plus CR plus A, and 87% for BR plus A. The 1-year OS rate was 94% for BR plus CR, 98% for BR plus CR plus A, and 95% for BR plus A.
    • The trial was closed because of an interim futility analysis for superiority of any treatment arm. However, BR plus A was the least toxic arm.

    Summary: Although adding acalabrutinib to BR plus CR did not improve efficacy, adding acalabrutinib to BR (and avoiding cytarabine) was equally effective. Since the pivotal initial trial by the European Mantle Cell Lymphoma Network [1,2] failed to confirm OS benefit at 10 years (without post-hoc adjustments), this finding suggests that cytarabine is not a mandatory agent in some induction therapy regimens.

It remains unclear whether induction therapy that combines chemotherapy with BTK inhibitors can be replaced by BTK inhibitors alone or BTK inhibitors in combination with CD20-directed monoclonal antibodies like rituximab or obinutuzumab.

BTK inhibitors With or Without Other Drugs

Evidence (BTK inhibitors with or without other drugs):

  1. A prospective trial, published in abstract form, included 397 patients aged 60 years and older with previously untreated MCL. Patients were randomly assigned to receive either ibrutinib plus rituximab or BR.[12]
    • With a median follow-up of 47.9 months, the PFS rate was 65.3% for patients who received ibrutinib plus rituximab and 42.4% for patients who received BR (HR, 0.69; 95% CI, 0.52–0.90; P = .003).[12][Level of evidence B1]
  2. A prospective randomized trial (SHINE [NCT01776840]) included 523 patients aged 65 years and older with previously untreated MCL. Patients were randomly assigned to receive either ibrutinib plus BR or placebo plus BR. The primary end point was PFS.[13]
    • With a median follow-up of 84.7 months, the median PFS was 80.6 months for patients who received ibrutinib plus BR and 52.9 months for patients who received placebo plus BR (HR, 0.75; 95% CI, 0.59–0.96; P = .01). There was no difference in the 7-year OS rate (55.0% vs. 56.8%; HR, 1.07; 95% CI, 0.81–1.40).[13][Level of evidence B1]
    • The magnitude of benefit for PFS results contrasted with the lower 7-year OS may cast doubt on the long-term safety of the ibrutinib plus BR combination. Infectious deaths in the combination group contributed to the lack of survival advantage.
    • Further trials are required to determine if ibrutinib alone can achieve the same results without adding BR, potentially avoiding the increased toxicities and infectious deaths from including bendamustine.
  3. In a prospective randomized trial, 280 patients with relapsed or refractory MCL received either ibrutinib or temsirolimus.[14]
    • With a median follow-up of 15 months, the median PFS was 14.6 months in the ibrutinib group and 6.2 months in the temsirolimus group (HR, 0.43; 95% CI, 0.32–0.58; P < .0001).[14][Level of evidence B1]
  4. In a phase II trial of previously untreated patients older than 64 years with MCL, 50 patients received ibrutinib plus rituximab.[15]
    • With a median follow-up of 45 months, the overall response rate was 96%, the complete response rate was 71%, the 3-year PFS rate was 87%, and the 3-year OS rate was 94%.[15][Level of evidence C3]
  5. In a phase II trial of 131 previously untreated patients with MCL aged 65 years or younger, 1 year of ibrutinib plus 4 weeks of rituximab resulted in a complete response rate of 89% prior to any chemotherapy consolidation.[16][Level of evidence C3]
  6. A phase II trial using ibrutinib plus rituximab included asymptomatic patients with previously untreated MCL.[17]
  7. Ibrutinib was combined with another active agent, venetoclax, in a phase II study of 23 patients with relapsed or refractory MCL.[18]
    • With a median follow-up of 7 years 4 months, the 7-year PFS rate was 30% (95% CI, 14%–49%), and the 7-year OS rate was 43% (95% CI, 23%–26%).[18][Level of evidence C3]
  8. A randomized prospective trial (SYMPATICO [NCT03112174]) included 267 patients with relapsed or refractory MCL. The trial compared (1) ibrutinib plus venetoclax for 2 years followed by ibrutinib until disease progression versus (2) ibrutinib plus placebo followed by ibrutinib.[19]
    • With a median follow-up of 51.2 months (interquartile range, 48.2–55.3), the median PFS was 31.9 months (95% CI, 22.8–47.0) in the ibrutinib-venetoclax group and 22.1 months (16.5–29.5) in the ibrutinib-placebo group (HR, 0.65; 95% CI, 0.47–0.88; P = .0052).[19]
  9. Acalabrutinib was evaluated in a phase II study of 124 patients with relapsed or refractory MCL.[20]
    • There was an 81% overall response rate, 40% complete response rate, and 67% 1-year PFS rate.[20][Level of evidence C3]
  10. Zanubrutinib was evaluated in a phase II study of 86 patients with relapsed or refractory MCL.[21]
    • After a median follow-up of 35.3 months, the overall response rate was 84%, the complete response rate was 78%, and the median PFS was 33.0 months.[21][Level of evidence C3]

Summary: Ibrutinib and other BTK inhibitors such as acalabrutinib, zanubrutinib, and the noncovalent inhibitor pirtobrutinib are used in multiple clinical trials either alone or mostly in combination with rituximab, obinutuzumab, or venetoclax. Multiple agents are combined in clinical trials for the highest-risk patients with TP53 alterations, blastoid morphology, or high Ki-67. Further clinical trials may establish BTK inhibitors without chemotherapy as a standard first-line regimen for patients with standard-risk MCL.

Highest-Risk Patients With Blastoid Morphology and/or a TP53 Pathogenic Variant

Although the prior standard of care for untreated MCL was chemoimmunotherapy including high-dose cytarabine and autologous SCT, patients with TP53-altered MCL have had poor outcomes with this regimen, with a median PFS of under 1 year.[22,23] BTK inhibitors combined with other immunological or targeted molecules are particularly applicable for testing. Consolidation with allogeneic SCT or trials studying chimeric antigen receptor T cells or bispecific antibodies are also warranted after treatment response.[24]

Evidence (new combinations for highest-risk patients):

  1. A phase II trial included 25 patients with TP53 pathogenic variants who required therapy because of significant constitutional symptoms, cytopenias, symptomatic splenomegaly, progressive nodal involvement, or significant organ compression or involvement. Patients received zanubrutinib (the selective BTK inhibitor), obinutuzumab (the humanized anti-CD20 monoclonal antibody), and venetoclax (the BCL inhibitor, with a 5-week ramp-up beginning on day 1 of the third cycle).[25]
    • With a median follow-up of 28.2 months, the overall response rate was 96% and the complete response rate was 68% at the start of cycle three. The 2-year PFS rate was 72% (95% CI, 56%–92%), and the 2-year OS rate was 76% (95% CI, 79%–100%).[25][Level of evidence C3]

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. Hermine O, Hoster E, Walewski J, et al.: Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): a randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. Lancet 388 (10044): 565-75, 2016. [PUBMED Abstract]
  2. Hermine O, Jiang L, Walewski J, et al.: Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL younger): a long-term follow-up of the randomized, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. [Abstract] Blood 138 (Suppl 1); A-380, 2021.
  3. Kluin-Nelemans HC, Hoster E, Hermine O, et al.: Treatment of Older Patients With Mantle Cell Lymphoma (MCL): Long-Term Follow-Up of the Randomized European MCL Elderly Trial. J Clin Oncol 38 (3): 248-256, 2020. [PUBMED Abstract]
  4. Rummel MJ, Niederle N, Maschmeyer G, et al.: Bendamustine plus rituximab versus CHOP plus rituximab as first-line treatment for patients with indolent and mantle-cell lymphomas: an open-label, multicentre, randomised, phase 3 non-inferiority trial. Lancet 381 (9873): 1203-10, 2013. [PUBMED Abstract]
  5. Robak T, Jin J, Pylypenko H, et al.: Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: final overall survival results of a randomised, open-label, phase 3 study. Lancet Oncol 19 (11): 1449-1458, 2018. [PUBMED Abstract]
  6. Hermine O, Jiang L, Walewski J, et al.: High-Dose Cytarabine and Autologous Stem-Cell Transplantation in Mantle Cell Lymphoma: Long-Term Follow-Up of the Randomized Mantle Cell Lymphoma Younger Trial of the European Mantle Cell Lymphoma Network. J Clin Oncol 41 (3): 479-484, 2023. [PUBMED Abstract]
  7. Dreyling M, Doorduijn J, Giné E, et al.: Ibrutinib combined with immunochemotherapy with or without autologous stem-cell transplantation versus immunochemotherapy and autologous stem-cell transplantation in previously untreated patients with mantle cell lymphoma (TRIANGLE): a three-arm, randomised, open-label, phase 3 superiority trial of the European Mantle Cell Lymphoma Network. Lancet 403 (10441): 2293-2306, 2024. [PUBMED Abstract]
  8. Dreyling M, Doorduijn JK, Gine E, et al.: Role of autologous stem cell transplantation in the context of ibrutinib-containing first-line treatment in younger patients with mantle cell lymphoma: results from the randomized Triangle trial by the European MCL Network. [Abstract] Blood 144 (Suppl 1) A-240, 240-2, 2024.
  9. Fenske TS, Wang XV, Till BG, et al.: Lack of benefit of autologous hematopoietic cell transplantation (auto-HCT) in mantle cell lymphoma (MCL) patients (pts) in first complete remission (CR) with undetectable minimal residual disease (uMRD): initial report from the ECOG-ACRIN EA4151 phase 3 randomized trial. [Abstract] Blood 144 (Suppl 2): A-LBA-6, 2024.
  10. Martin P, Cohen JB, Wang M, et al.: Treatment Outcomes and Roles of Transplantation and Maintenance Rituximab in Patients With Previously Untreated Mantle Cell Lymphoma: Results From Large Real-World Cohorts. J Clin Oncol 41 (3): 541-554, 2023. [PUBMED Abstract]
  11. Wagner-Johnston N, Jegede O, Spurgeon SE, et al.: Addition or substitution of acalabrutinib in intensive frontline chemoimmunotherapy for patients ≤ 70 years old with mantle cell lymphoma: outcomes of the 3-arm randomized phase II intergroup trial ECOG-ACRIN EA4181. [Abstract] Blood 144 (Suppl 1): A-236, 2024.
  12. Lewis DJ, Jerkeman M, Sorrell L, et al.: Ibrutinib-rituximab is superior to rituximab-chemotherapy in previously untreated older mantle cell lymphoma patients: results from the international randomised controlled trial, Enrich. [Abstract] Blood 144 (Suppl 1): A-235, 2024.
  13. Wang ML, Jurczak W, Jerkeman M, et al.: Ibrutinib plus Bendamustine and Rituximab in Untreated Mantle-Cell Lymphoma. N Engl J Med 386 (26): 2482-2494, 2022. [PUBMED Abstract]
  14. Dreyling M, Jurczak W, Jerkeman M, et al.: Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet 387 (10020): 770-8, 2016. [PUBMED Abstract]
  15. Jain P, Zhao S, Lee HJ, et al.: Ibrutinib With Rituximab in First-Line Treatment of Older Patients With Mantle Cell Lymphoma. J Clin Oncol 40 (2): 202-212, 2022. [PUBMED Abstract]
  16. Wang ML, Jain P, Zhao S, et al.: Ibrutinib-rituximab followed by R-HCVAD as frontline treatment for young patients (≤65 years) with mantle cell lymphoma (WINDOW-1): a single-arm, phase 2 trial. Lancet Oncol 23 (3): 406-415, 2022. [PUBMED Abstract]
  17. Giné E, de la Cruz F, Jiménez Ubieto A, et al.: Ibrutinib in Combination With Rituximab for Indolent Clinical Forms of Mantle Cell Lymphoma (IMCL-2015): A Multicenter, Open-Label, Single-Arm, Phase II Trial. J Clin Oncol 40 (11): 1196-1205, 2022. [PUBMED Abstract]
  18. Handunnetti SM, Anderson MA, Burbury K, et al.: Seven-year outcomes of venetoclax-ibrutinib therapy in mantle cell lymphoma: durable responses and treatment-free remissions. Blood 144 (8): 867-872, 2024. [PUBMED Abstract]
  19. Wang M, Jurczak W, Trneny M, et al.: Ibrutinib plus venetoclax in relapsed or refractory mantle cell lymphoma (SYMPATICO): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 26 (2): 200-213, 2025. [PUBMED Abstract]
  20. Wang M, Rule S, Zinzani PL, et al.: Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet 391 (10121): 659-667, 2018. [PUBMED Abstract]
  21. Song Y, Zhou K, Zou D, et al.: Zanubrutinib in relapsed/refractory mantle cell lymphoma: long-term efficacy and safety results from a phase 2 study. Blood 139 (21): 3148-3158, 2022. [PUBMED Abstract]
  22. Eskelund CW, Dahl C, Hansen JW, et al.: TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 130 (17): 1903-1910, 2017. [PUBMED Abstract]
  23. Ferrero S, Rossi D, Rinaldi A, et al.: KMT2D mutations and TP53 disruptions are poor prognostic biomarkers in mantle cell lymphoma receiving high-dose therapy: a FIL study. Haematologica 105 (6): 1604-1612, 2020. [PUBMED Abstract]
  24. Fenske TS, Zhang MJ, Carreras J, et al.: Autologous or reduced-intensity conditioning allogeneic hematopoietic cell transplantation for chemotherapy-sensitive mantle-cell lymphoma: analysis of transplantation timing and modality. J Clin Oncol 32 (4): 273-81, 2014. [PUBMED Abstract]
  25. Kumar A, Soumerai J, Abramson JS, et al.: Zanubrutinib, obinutuzumab, and venetoclax for first-line treatment of mantle cell lymphoma with a TP53 mutation. Blood 145 (5): 497-507, 2025. [PUBMED Abstract]

Maintenance Therapy After Induction Therapy for Mantle Cell Lymphoma

The use of maintenance therapy with rituximab alone or combined with a Bruton tyrosine kinase (BTK) inhibitor after induction therapy or any consolidation has been the standard of care for mantle cell lymphoma (MCL). The duration of maintenance therapy has ranged from 3 years until time of disease relapse.

Rituximab Maintenance Therapy Alone or Combined With a BTK Inhibitor

Evidence (use of rituximab maintenance therapy alone or combined with a BTK inhibitor):

  1. A prospective randomized trial included 299 patients with MCL who underwent chemoimmunotherapy and autologous stem cell transplant (SCT) consolidation. Patients were then randomly assigned to receive either 3 years of rituximab maintenance therapy or observation.[1]
    • With a median follow-up of 50.2 months after autologous SCT, the overall survival (OS) rate was 89% (95% confidence interval [CI], 81%–94%) in the rituximab group and 80% (95% CI 72%–88%) in the observation group (hazard ratio [HR], 0.50; 95% CI, 0.26–0.99; P = .04).[1][Level of evidence A1]
    • The 4-year event-free survival rate was 79% (95% CI, 70%–86%) in the rituximab group and 61% (95% CI, 51%–70%) in the observation group (P = .001).[1]
    • The 4-year progression-free survival (PFS) rate was 83% (95% CI, 73%–88%) in the rituximab group and 64% (95% CI, 55%–73%) in the observation group (P < .001).[1]
  2. In a prospective trial of 299 patients with untreated MCL, 257 responders received four courses of R-DHAP (rituximab, dexamethasone, cytarabine, and cisplatin) and autologous SCT. These patients were then randomly assigned to receive either rituximab maintenance therapy for 3 years or no maintenance therapy.[2]
    • The 7-year PFS rate was significantly higher for the rituximab maintenance group at 78.5 % (95% CI, 69.9%–85.0%) versus 47.4% (95% CI, 39.9%–56.3%) for the group that did not receive maintenance therapy (HR, 0.36; 95% CI, 0.23–0.56; P < .0001).[2][Level of evidence B1]
    • After randomization, with a median follow-up of 7.5 years, the 7-year OS rate in the rituximab maintenance group was not significantly better than in the no-maintenance group (83.2% [95% CI, 74.7%–89.0%] vs. 72.2% [95% CI, 62.9%–79.5%]; HR, 0.63; 95% CI, 0.37–1.08).
  3. In a prospective randomized trial, 500 patients aged 60 years or older and not transplant-eligible received induction therapy with R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) or R-FC (rituximab, fludarabine, cyclophosphamide) for six to eight cycles. Responders were randomly assigned to receive either rituximab or interferon alfa maintenance therapy until disease relapse.[3]
    • With a median follow-up of 8.0 years for the 316 responding patients, the median OS was 9.8 years in the rituximab maintenance group and 7.1 years in the interferon alfa maintenance group (P = .0054).[3][Level of evidence A1]
  4. As previously described, the TRIANGLE trial (NCT02858258) was a prospective randomized trial that included 870 patients aged 65 years or younger with previously untreated MCL who were transplant-eligible.[4,5] Patients received one of three induction regimens with chemoimmunotherapy (R-CHOP/R-DHAP), with or without ibrutinib and with or without autologous SCT consolidation in the ibrutinib arms. All patients were recommended to have maintenance therapy with at least rituximab or with both rituximab and ibrutinib in the ibrutinib induction therapy arms.[6] Not all patients could tolerate or receive maintenance therapy, and some deferred the recommended maintenance therapy (33%–41% of patients in each group).
    • In a retrospective analysis with a median follow-up of 4.0 years, the 4-year PFS rate was 10% to 29% higher for patients who received rituximab maintenance (P ranged from .016 to < .001).[6][Level of evidence C3]
  5. A retrospective analysis of 1,265 patients aged 65 years and younger evaluated rituximab maintenance therapy after bendamustine and rituximab induction.[7]
    • A benefit was seen for rituximab maintenance therapy in time-to-next treatment (HR, 1.96; 95% CI, 1.61–2.38; P < .001) and OS (HR, 1.51; 95% CI, 1.19–1.92; P < .001).[7][Level of evidence C3]
  6. In a prospective randomized trial, 319 patients with follicular lymphoma or MCL received R-FCM (rituximab, fludarabine, cyclophosphamide, and mitoxantrone) or FCM (a subsequent analysis confirmed the superiority of R-FCM and all patients received that induction). The 267 patients with disease response were randomly assigned to either rituximab maintenance therapy or observation. Most patients had follicular lymphoma, but 47 patients with disease response had MCL.[8]
    • With a median follow-up of 26 months, the 2-year PFS rate was 45% in the rituximab maintenance group and 9% in the observation group (P = .049).[8][Level of evidence B1]

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. Le Gouill S, Thieblemont C, Oberic L, et al.: Rituximab after Autologous Stem-Cell Transplantation in Mantle-Cell Lymphoma. N Engl J Med 377 (13): 1250-1260, 2017. [PUBMED Abstract]
  2. Sarkozy C, Thieblemont C, Oberic L, et al.: Long-Term Follow-Up of Rituximab Maintenance in Young Patients With Mantle-Cell Lymphoma Included in the LYMA Trial: A LYSA Study. J Clin Oncol 42 (7): 769-773, 2024. [PUBMED Abstract]
  3. Kluin-Nelemans HC, Hoster E, Hermine O, et al.: Treatment of Older Patients With Mantle Cell Lymphoma (MCL): Long-Term Follow-Up of the Randomized European MCL Elderly Trial. J Clin Oncol 38 (3): 248-256, 2020. [PUBMED Abstract]
  4. Dreyling M, Doorduijn J, Giné E, et al.: Ibrutinib combined with immunochemotherapy with or without autologous stem-cell transplantation versus immunochemotherapy and autologous stem-cell transplantation in previously untreated patients with mantle cell lymphoma (TRIANGLE): a three-arm, randomised, open-label, phase 3 superiority trial of the European Mantle Cell Lymphoma Network. Lancet 403 (10441): 2293-2306, 2024. [PUBMED Abstract]
  5. Dreyling M, Doorduijn JK, Gine E, et al.: Role of autologous stem cell transplantation in the context of ibrutinib-containing first-line treatment in younger patients with mantle cell lymphoma: results from the randomized Triangle trial by the European MCL Network. [Abstract] Blood 144 (Suppl 1) A-240, 240-2, 2024.
  6. Ladetto M, Gutmair K, Doorduijn JK, et al.: Impact of rituximab maintenance added to ibrutinib-containing regimens with and without ASCT in younger, previously untreated MCL patients: an analysis of the Triangle data embedded in the Multiply Project. [Abstract] Blood 144 (Suppl 1): A-237, 2024.
  7. Martin P, Cohen JB, Wang M, et al.: Treatment Outcomes and Roles of Transplantation and Maintenance Rituximab in Patients With Previously Untreated Mantle Cell Lymphoma: Results From Large Real-World Cohorts. J Clin Oncol 41 (3): 541-554, 2023. [PUBMED Abstract]
  8. Forstpointner R, Unterhalt M, Dreyling M, et al.: Maintenance therapy with rituximab leads to a significant prolongation of response duration after salvage therapy with a combination of rituximab, fludarabine, cyclophosphamide, and mitoxantrone (R-FCM) in patients with recurring and refractory follicular and mantle cell lymphomas: Results of a prospective randomized study of the German Low Grade Lymphoma Study Group (GLSG). Blood 108 (13): 4003-8, 2006. [PUBMED Abstract]

Treatment of Relapsed/Refractory Mantle Cell Lymphoma

Patients with mantle cell lymphoma (MCL) whose disease relapses after standard chemoimmunotherapy with or without autologous stem cell transplant (SCT) typically receive a Bruton tyrosine kinase (BTK) inhibitor. A series of retrospective trials have shown that a BTK inhibitor has superior outcomes compared with repeat chemoimmunotherapy. However, it must be emphasized that some patients do respond well to repeat chemoimmunotherapy (e.g., BR [bendamustine plus rituximab] after R-CHOP [rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone]/R-DHAP [rituximab, dexamethasone, cytarabine, and cisplatin]).[1] Because of the relatively short durations of third and subsequent remissions, consolidation therapy should be considered with allogeneic SCT, chimeric antigen receptor (CAR) T-cell therapy, or clinical trials of bispecific antibodies.[1] Patients with disease relapse who are considered to be at highest risk include those with refractory disease, blastoid morphology, a TP53 pathogenic variant, a Ki-67 level of at least 30%, or progression of disease within 24 months.[1,2]

Treatment for Patients Who Have Not Received a Prior BTK Inhibitor

Evidence (patients who have not received a prior BTK inhibitor):

  1. An observational cohort study included 385 patients with MCL and disease relapse after 2 years. Patients received BTK inhibitors (usually ibrutinib) or chemoimmunotherapy in a nonrandomized fashion.[3]
    • With a median follow-up of 53 months, the median overall survival (OS) was better in patients who received BTK inhibitors (not reached [NR]) than in patients who received chemoimmunotherapy (56 months) (P = .03).[3][Level of evidence C1]
  2. Acalabrutinib, a selective BTK inhibitor, was studied in a phase II trial of 124 patients with relapsed/refractory MCL. The results of this trial led the U.S. Food and Drug Administration (FDA) to approve acalabrutinib in 2017, before the approval of ibrutinib.[4]
    • With a median follow-up of 15.2 months, the overall response rate was 81% (95% confidence interval [CI], 73%–87%), the complete response rate was 40% (95% CI, 31%–49%), and the 1-year progression-free survival (PFS) rate was 67% (95% CI, 58%–75%).[4][Level of evidence C2]
    • Acalabrutinib is a more selective BTK inhibitor with lower rates of atrial fibrillation than ibrutinib (3%–4% vs. 10%–12%).
  3. Zanubrutinib, a selective BTK inhibitor, was evaluated in a phase II trial of 86 patients with relapsed/refractory MCL.[5]
    • With a median follow-up of 35.3 months, the overall response rate was 84%, the complete response rate was 98%, and the median PFS was 33 months (95% CI, 19.4–not estimable [NE]).[5][Level of evidence C2]
    • Zanubrutinib is a more selective BTK inhibitor with lower rates of atrial fibrillation than ibrutinib (3%–4% vs. 10%–12%).
  4. Multiple phase II studies of ibrutinib in patients with relapsed or refractory MCL, including a pooled analysis of 370 patients, showed overall response rates of 66% to 68% and median PFS of 12.5 months to 13.9 months.[68] Long-term follow-up of the pooled analysis showed an OS of 61.6 months.[9]

Treatment for Patients Who Have Received a Prior BTK Inhibitor

Evidence (patients who have received a prior BTK inhibitor):

  1. The reversible, noncovalent BTK inhibitor pirtobrutinib was evaluated in a phase I/II trial of 164 patients with relapsed/refractory MCL.[10]
    • With a median follow-up of 12 months, among the 90 patients previously treated with covalent BTK inhibitors (ibrutinib, acalabrutinib, or zanubrutinib), the overall response rate was 57.8% (95% CI, 46.9%–68.1%), including a complete response rate of 20.0%.[10][Level of evidence C3]
    • The median duration of response was 2.6 months (95% CI, 7.5–NR).[10][Level of evidence C2]
    • Only 3% of patients discontinued therapy because of side effects, which included atrial fibrillation in 1.2% of patients, grade 3 or higher bleeding in 3.7%, dyspnea in 16.5%, diarrhea in 21.3%, and fatigue in 29.9%.

    The FDA approved pirtobrutinib for patients who received two prior lines of therapy, including a covalent BTK inhibitor.

  2. The combination of lenalidomide plus rituximab has been studied in several phase II trials, with an overall response rate of approximately 50% in patients with relapsed MCL.[1113][Level of evidence C3]
  3. A prospective phase II trial (ZUMA-2 [NCT02601313]) included 68 patients with relapsed or refractory MCL whose disease failed to respond to BTK inhibitors. Patients received CAR T-cell therapy with brexucabtagene autoleucel, which targets CD19.[14]
    • With a median follow-up of 36 months, the objective response rate was 91% (95% CI, 82%–97%), the complete response rate was 68% (95% CI, 55%–78%), the median PFS was 25.8 months (95% CI, 10–48), and the OS was 46.6 months (95% CI, 24.9–NE).[14][Level of evidence C3]
    • Grade 3 or higher cytokine release syndrome occurred in 15% of patients, and neurological events occurred in 31% of patients.
    • A retrospective evaluation at 16 institutions included 168 patients who received brexucabtagene autoleucel as part of the U.S. Lymphoma CAR-T Consortium. The study showed similar response rates and PFS as the ZUMA-2 trial.[15][Level of evidence C3]
  4. Patients with relapsed or refractory MCL who had received a median of three prior lines of therapy were enrolled in a phase I/II trial of lisocabtagene maraleucel, an anti-CD19 CAR T-cell therapy.[16]
    • With a median follow-up of 16.1 months, the objective response rate was 83.1% (95% CI, 73.3%–90.5%), and the complete response rate was 72.3% (95% CI, 61.4%–81.6%). The median duration of response was 15.7 months (95% CI, 6.2–24.0).[16][Level of evidence C3]
    • Grade 3 or higher cytokine release syndrome occurred in 1% of patients.
  5. Patients with relapsed or refractory MCL received the CD20 × CD3 bispecific antibody glofitamab in a phase I/II trial.[17]
    • With a median follow-up of 19.6 months, 60 patients were evaluable. The overall response rate was 85.0% (95% CI, 73.4%–92.9%), and the complete response rate was 78.3% (95% CI, 65.8%–87.9%).[17][Level of evidence C3]
    • The median duration of complete response was 15.4 months (95% CI, 12.7–NE), and the 1-year duration of complete response was 71.0% (95% CI, 56.8%–85.2%).[17][Level of evidence C3]
    • Grade 3 or higher cytokine release syndrome occurred in 25% of patients. No grade 3 or higher neurological symptoms were reported.

Treatment for Patients With Highest-Risk Disease

Evidence (treatment for patients with highest-risk disease with blastoid morphology, Ki-67 ≥30%, a TP53 pathogenic variant, or progression of disease <2 years after initial therapy):

  1. A prospective randomized trial of patients with relapsed or refractory MCL compared ibrutinib plus venetoclax (the BCL-2 inhibitor) versus ibrutinib alone. TP53 pathogenic variants were present in 49% of patients.[18]
    • With a median follow-up of 51.2 months, the PFS favored the ibrutinib combination compared with ibrutinib alone (31.9 months vs. 22.1 months; HR, 0.65; 95% CI, 0.47–0.88; P = .0052). This benefit was seen for the patients with blastoid morphology or TP53 pathogenic variants.[18][Level of evidence B1]
    • The complete response rate was 54% in the combination group and 32% in the ibrutinib-alone group (P = .004).[18][Level of evidence C3]
    • The median OS was 44.9 months for the ibrutinib-plus-venetoclax arm and 38.6 months for the ibrutinib-alone arm, but this was not statistically significant (P = .346).
    • The triple combination of obinutuzumab, ibrutinib, and venetoclax is being studied in clinical trials for the highest-risk patients with relapsed or refractory MCL.

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. Silkenstedt E, Dreyling M: Treatment of relapsed/refractory MCL. Blood 145 (7): 673-682, 2025. [PUBMED Abstract]
  2. Nadeu F, Martin-Garcia D, Clot G, et al.: Genomic and epigenomic insights into the origin, pathogenesis, and clinical behavior of mantle cell lymphoma subtypes. Blood 136 (12): 1419-1432, 2020. [PUBMED Abstract]
  3. Malinverni C, Bernardelli A, Glimelius I, et al.: Outcomes of younger patients with mantle cell lymphoma experiencing late relapse (>24 months): the LATE-POD study. Blood 144 (9): 1001-1009, 2024. [PUBMED Abstract]
  4. Wang M, Rule S, Zinzani PL, et al.: Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet 391 (10121): 659-667, 2018. [PUBMED Abstract]
  5. Song Y, Zhou K, Zou D, et al.: Zanubrutinib in relapsed/refractory mantle cell lymphoma: long-term efficacy and safety results from a phase 2 study. Blood 139 (21): 3148-3158, 2022. [PUBMED Abstract]
  6. Dreyling M, Jurczak W, Jerkeman M, et al.: Ibrutinib versus temsirolimus in patients with relapsed or refractory mantle-cell lymphoma: an international, randomised, open-label, phase 3 study. Lancet 387 (10020): 770-8, 2016. [PUBMED Abstract]
  7. Visco C, Di Rocco A, Evangelista A, et al.: Outcomes in first relapsed-refractory younger patients with mantle cell lymphoma: results from the MANTLE-FIRST study. Leukemia 35 (3): 787-795, 2021. [PUBMED Abstract]
  8. Rule S, Dreyling M, Goy A, et al.: Outcomes in 370 patients with mantle cell lymphoma treated with ibrutinib: a pooled analysis from three open-label studies. Br J Haematol 179 (3): 430-438, 2017. [PUBMED Abstract]
  9. Dreyling M, Goy A, Hess G, et al.: Long-term Outcomes With Ibrutinib Treatment for Patients With Relapsed/Refractory Mantle Cell Lymphoma: A Pooled Analysis of 3 Clinical Trials With Nearly 10 Years of Follow-up. Hemasphere 6 (5): e712, 2022. [PUBMED Abstract]
  10. Wang ML, Jurczak W, Zinzani PL, et al.: Pirtobrutinib in Covalent Bruton Tyrosine Kinase Inhibitor Pretreated Mantle-Cell Lymphoma. J Clin Oncol 41 (24): 3988-3997, 2023. [PUBMED Abstract]
  11. Ruan J, Martin P, Shah B, et al.: Lenalidomide plus Rituximab as Initial Treatment for Mantle-Cell Lymphoma. N Engl J Med 373 (19): 1835-44, 2015. [PUBMED Abstract]
  12. Ruan J, Martin P, Christos P, et al.: Five-year follow-up of lenalidomide plus rituximab as initial treatment of mantle cell lymphoma. Blood 132 (19): 2016-2025, 2018. [PUBMED Abstract]
  13. Wang M, Fayad L, Wagner-Bartak N, et al.: Lenalidomide in combination with rituximab for patients with relapsed or refractory mantle-cell lymphoma: a phase 1/2 clinical trial. Lancet Oncol 13 (7): 716-23, 2012. [PUBMED Abstract]
  14. Wang M, Munoz J, Goy A, et al.: Three-Year Follow-Up of KTE-X19 in Patients With Relapsed/Refractory Mantle Cell Lymphoma, Including High-Risk Subgroups, in the ZUMA-2 Study. J Clin Oncol 41 (3): 555-567, 2023. [PUBMED Abstract]
  15. Wang Y, Jain P, Locke FL, et al.: Brexucabtagene Autoleucel for Relapsed or Refractory Mantle Cell Lymphoma in Standard-of-Care Practice: Results From the US Lymphoma CAR T Consortium. J Clin Oncol 41 (14): 2594-2606, 2023. [PUBMED Abstract]
  16. Wang M, Siddiqi T, Gordon LI, et al.: Lisocabtagene Maraleucel in Relapsed/Refractory Mantle Cell Lymphoma: Primary Analysis of the Mantle Cell Lymphoma Cohort From TRANSCEND NHL 001, a Phase I Multicenter Seamless Design Study. J Clin Oncol 42 (10): 1146-1157, 2024. [PUBMED Abstract]
  17. Phillips TJ, Carlo-Stella C, Morschhauser F, et al.: Glofitamab in Relapsed/Refractory Mantle Cell Lymphoma: Results From a Phase I/II Study. J Clin Oncol 43 (3): 318-328, 2025. [PUBMED Abstract]
  18. Sawalha Y, Goyal S, Switchenko JM, et al.: A multicenter analysis of the outcomes with venetoclax in patients with relapsed mantle cell lymphoma. Blood Adv 7 (13): 2983-2993, 2023. [PUBMED Abstract]

Key References for Mantle Cell Lymphoma

These references have been identified by members of the PDQ Adult Treatment Editorial Board as significant in the field of mantle cell lymphoma treatment. This list is provided to inform users of important studies that have helped shape the current understanding of and treatment options for mantle cell lymphoma. Listed after each reference are the sections within this summary where the reference is cited.

  • Dreyling M, Doorduijn J, Giné E, et al.: Ibrutinib combined with immunochemotherapy with or without autologous stem-cell transplantation versus immunochemotherapy and autologous stem-cell transplantation in previously untreated patients with mantle cell lymphoma (TRIANGLE): a three-arm, randomised, open-label, phase 3 superiority trial of the European Mantle Cell Lymphoma Network. Lancet 403 (10441): 2293-2306, 2024. [PUBMED Abstract]

    Cited in:

  • Dreyling M, Doorduijn JK, Gine E, et al.: Role of autologous stem cell transplantation in the context of ibrutinib-containing first-line treatment in younger patients with mantle cell lymphoma: results from the randomized Triangle trial by the European MCL Network. [Abstract] Blood 144 (Suppl 1) A-240, 240-2, 2024.

    Cited in:

  • Fenske TS, Wang XV, Till BG, et al.: Lack of benefit of autologous hematopoietic cell transplantation (auto-HCT) in mantle cell lymphoma (MCL) patients (pts) in first complete remission (CR) with undetectable minimal residual disease (uMRD): initial report from the ECOG-ACRIN EA4151 phase 3 randomized trial. [Abstract] Blood 144 (Suppl 2): A-LBA-6, 2024.

    Cited in:

  • Hermine O, Jiang L, Walewski J, et al.: High-Dose Cytarabine and Autologous Stem-Cell Transplantation in Mantle Cell Lymphoma: Long-Term Follow-Up of the Randomized Mantle Cell Lymphoma Younger Trial of the European Mantle Cell Lymphoma Network. J Clin Oncol 41 (3): 479-484, 2023. [PUBMED Abstract]

    Cited in:

  • Le Gouill S, Thieblemont C, Oberic L, et al.: Rituximab after Autologous Stem-Cell Transplantation in Mantle-Cell Lymphoma. N Engl J Med 377 (13): 1250-1260, 2017. [PUBMED Abstract]

    Cited in:

  • Lewis DJ, Jerkeman M, Sorrell L, et al.: Ibrutinib-rituximab is superior to rituximab-chemotherapy in previously untreated older mantle cell lymphoma patients: results from the international randomised controlled trial, Enrich. [Abstract] Blood 144 (Suppl 1): A-235, 2024.

    Cited in:

Latest Updates to This Summary (05/12/2025)

The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

This is a new 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 mantle cell lymphoma. 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 Mantle Cell Lymphoma Treatment are:

  • Eric J. Seifter, MD (Johns Hopkins University)
  • Cole H. Sterling, MD (Johns Hopkins University)

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 Mantle Cell Lymphoma Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/lymphoma/hp/mantle-cell-lymphoma-treatment. Accessed <MM/DD/YYYY>.

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.

Childhood Gastrointestinal Stromal Tumors (PDQ®)–Patient Version

Childhood Gastrointestinal Stromal Tumors (PDQ®)–Patient Version

What is childhood gastrointestinal stromal tumor?

Childhood gastrointestinal stromal tumor (GIST) is a cancer that forms in the tissues of the wall of the stomach or intestines. Childhood GIST usually occurs in the stomach. It is most common in girls and typically appears in the teen years.

The gastrointestinal (GI) tract is part of the body’s digestive system. It helps digest food and takes nutrients (vitamins, minerals, carbohydrates, fats, proteins, and water) from food so they can be used by the body. The GI tract is made up of the:

GISTs usually begin in cells in the tissues of the wall of the stomach or intestines that help food move along the digestive tract.

EnlargeDrawing of the gastrointestinal tract showing the stomach, small intestine, colon, and rectum.
Gastrointestinal stromal tumors (GISTs) are most common in the stomach and small intestine but may be found anywhere in or near the gastrointestinal tract.

GISTs in children are not the same as GISTs in adults. Children should be seen at centers that specialize in treating GISTs in children and adolescents.

Causes and risk factors for childhood gastrointestinal stromal tumor

Childhood GIST is caused by certain changes to the way the cells in the wall of the stomach and intestines 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. Not every child with a risk factor will develop a GIST. And it will develop in some children who don’t have a known risk factor.

GIST may occur as part of the following syndromes:

Children with neurofibromatosis type 1 (NF1) may also have an increased risk.

Talk with your child’s doctor if you think your child may be at risk.

Symptoms of childhood gastrointestinal stromal tumor

The symptoms of childhood GIST may not appear until the tumor grows bigger. It’s important to check with your child’s doctor if your child has:

  • anemia, which causes tiredness, dizziness, fast or irregular heartbeat, shortness of breath, or pale skin
  • blood (either bright red or very dark) in the stool
  • a lump in the abdomen
  • a blockage in the intestine, which causes cramping pain in the abdomen, nausea, vomiting, diarrhea, constipation, and swelling of the abdomen

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

Tests to diagnose childhood gastrointestinal stromal tumor

If your child has symptoms that suggest a stomach or intestinal tumor, 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, they may recommend other tests. If your child is diagnosed with a GIST, the results of these tests will help plan treatment.

The tests used to diagnose GIST may include:

Gene testing

Gene testing analyzes cells or tissues from the tumor to look for changes in the KIT, PDGFRA, and SDH genes. Knowing whether there are changes in these genes can help diagnose GIST and plan treatment.

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. 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.

PET scan

A 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.

X-ray

X-ray is a type of radiation that can go through the body and make pictures of areas inside the body, such as the abdomen or the area where the tumor formed.

Biopsy

Biopsy is the removal of a sample of tissue from the tumor so that a pathologist can view it under a microscope to check for cancer. The following types of biopsies may be used to check for GIST:

  • Fine-needle aspiration uses a thin needle to remove tissue from the tumor.
  • Endoscopy looks 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 anus. 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 cancer.

The following laboratory test may be done to study the tissue samples:

  • 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 a fluorescent dye. After the antibodies bind to a specific antigen in the tissue sample, the enzyme or dye is activated, and the antigen can then be seen under a microscope. This type of test looks for the enzyme SDH in the patient’s tissue. When SDH is not present, it is called SDH-deficient GIST. Knowing whether the cancer is SDH-deficient can help plan treatment.

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. They 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.

Who treats children with gastrointestinal stromal tumor?

A pediatric oncologist, a doctor who specializes in treating children with cancer, oversees the treatment of GIST. 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 of childhood gastrointestinal stromal tumor

There are different types of treatment for children and adolescents with GIST. 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, whether the tumor has changes to the KIT, PDGFRA, or SDH genes, 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.

Types of treatment your child might have include:

  • Children with a GIST that has changes in the KIT or PDGFRA gene are treated 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. Imatinib and sunitinib are targeted therapies approved for adults with GIST and may be used in children and adolescents. Learn more about Targeted Therapy to Treat Cancer.
  • Children with a GIST that is SDH-deficient are treated with surgery to remove the tumor. More surgery may be needed if an intestinal blockage or bleeding occurs.

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.

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.

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 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:

  • physical problems
  • changes in mood, feelings, thinking, learning, or memory
  • second cancers (new types of cancer) or other conditions

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).

To learn more about follow-up tests, visit Tests to diagnose childhood gastrointestinal stromal tumor.

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 gastrointestinal stromal tumors. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.

Reviewers and Updates

Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.

The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Pediatric Treatment Editorial Board.

Clinical Trial Information

A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.

Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).

Permission to Use This Summary

PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”

The best way to cite this PDQ summary is:

PDQ® Pediatric Treatment Editorial Board. PDQ Childhood Gastrointestinal Stromal Tumors. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/soft-tissue-sarcoma/patient/child-gist-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.

Colorectal Cancer—Health Professional Version

Colorectal Cancer—Health Professional Version

Supportive & Palliative Care

We offer evidence-based supportive and palliative care information for health professionals on the assessment and management of cancer-related symptoms and conditions.

Cancer Pain Nausea and Vomiting Nutrition in Cancer Care Transition to End-of-Life Care Last Days of Life View all Supportive and Palliative Care Summaries

Male Breast Cancer Treatment (PDQ®)–Health Professional Version

Male Breast Cancer Treatment (PDQ®)–Health Professional Version

General Information About Male Breast Cancer

Incidence and Mortality

Estimated new cases and deaths from breast cancer (men only) in the United States in 2025:[1]

  • New cases: 2,800.
  • Deaths: 510.

Male breast cancer is rare.[2] Fewer than 1% of all breast carcinomas occur in men.[3,4] The mean age at diagnosis is between 60 and 70 years; however, men of all ages can be affected by the disease.

Anatomy

EnlargeAnatomy of the male breast; drawing shows the lymph nodes, nipple, areola, chest wall, ribs, muscle, fatty tissue, and ducts.
Anatomy of the male breast. The nipple and areola are shown on the outside of the breast. The lymph nodes, fatty tissue, ducts, and other parts of the inside of the breast are also shown.

Risk Factors

Predisposing risk factors for male breast cancer appear to include:[5,6]

  • Radiation exposure to breast/chest.
  • Estrogen use.
  • Diseases associated with hyperestrogenism, such as cirrhosis or Klinefelter syndrome.
  • Family health history: Definite familial tendencies are evident, with an increased incidence seen in men who have a number of female relatives with breast cancer.
  • Major inheritance susceptibility: Increased male breast cancer risk has been reported in families with BRCA pathogenic variants, although risk appears to be higher with inherited BRCA2 variants than with BRCA1 variants.[7,8] At age 70 years, men have an estimated cumulative breast cancer risk of 1.2% if they have BRCA1 pathogenic variants and 6.8% if they have BRCA2 pathogenic variants.[9] Other genes may be involved in male breast cancer predisposition, including pathogenic variants in the PTEN tumor suppressor gene, TP53 (Li-Fraumeni syndrome), PALB2, and in mismatch repair genes associated with Lynch syndrome (also called hereditary nonpolyposis colorectal cancer).[1012] For more information, see the sections on High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes in Genetics of Breast and Gynecologic Cancers, and Male Breast Cancer Screening and Surveillance for BRCA1/2 Carriers in BRCA1 and BRCA2: Cancer Risks and Management.

Clinical Features

Most breast cancers in men present with a retroareolar mass. Other signs include:

  • Nipple retraction.
  • Bleeding from the nipple.
  • Skin ulceration.
  • Peau d’orange.
  • Palpable axillary adenopathy.

Because of delays in diagnosis, breast cancer in men is more likely to present at an advanced stage.[2,5,13]

Diagnostic Evaluation

Breast imaging should be performed when breast cancer is suspected. The American College of Radiology recommends ultrasonography as the first imaging modality in men younger than 25 years because breast cancer is highly unlikely. Mammography is performed if ultrasonography findings are suspicious.

For men aged 25 years or older, or those who have a highly concerning physical examination, mammography is recommended as the initial test and ultrasonography is useful if mammography is inconclusive or suspicious.[14] Suspicious findings should be confirmed with a core biopsy. If the presence of tumor is confirmed, estrogen receptor, progesterone receptor, and human epidermal growth factor type 2 (HER2) expression/amplification should be evaluated.[15]

For more information, see the Diagnosis section in Breast Cancer Treatment.

Histopathologic Classification

Infiltrating ductal cancer is the most common tumor type of breast cancer in men, while invasive lobular carcinoma is very rare.[16] Breast cancer in men is almost always hormone receptor positive. In a male breast cancer series, 99% of the tumors were estrogen receptor positive, 82% were progesterone receptor positive, 9% were HER2 positive, and 0.3% were triple negative.[16]

Prognosis and Predictive Factors

Tumor size, lymph node involvement, and grade are anatomical prognostic factors, while estrogen receptor, progesterone receptor, and HER2 status are predictive of response to therapy.

A more advanced stage at diagnosis confers a worse prognosis for men with breast cancer.[2,5,13] A study found that mortality after breast cancer diagnosis was higher in male patients than in female patients. This disparity appeared to persist after accounting for clinical characteristics, treatment factors, and access to care, suggesting that biological factors and treatment efficacy may play a role.[17]

References
  1. American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
  2. Giordano SH, Cohen DS, Buzdar AU, et al.: Breast carcinoma in men: a population-based study. Cancer 101 (1): 51-7, 2004. [PUBMED Abstract]
  3. Borgen PI, Wong GY, Vlamis V, et al.: Current management of male breast cancer. A review of 104 cases. Ann Surg 215 (5): 451-7; discussion 457-9, 1992. [PUBMED Abstract]
  4. Fentiman IS, Fourquet A, Hortobagyi GN: Male breast cancer. Lancet 367 (9510): 595-604, 2006. [PUBMED Abstract]
  5. Giordano SH, Buzdar AU, Hortobagyi GN: Breast cancer in men. Ann Intern Med 137 (8): 678-87, 2002. [PUBMED Abstract]
  6. Hultborn R, Hanson C, Köpf I, et al.: Prevalence of Klinefelter’s syndrome in male breast cancer patients. Anticancer Res 17 (6D): 4293-7, 1997 Nov-Dec. [PUBMED Abstract]
  7. Wooster R, Bignell G, Lancaster J, et al.: Identification of the breast cancer susceptibility gene BRCA2. Nature 378 (6559): 789-92, 1995 Dec 21-28. [PUBMED Abstract]
  8. Thorlacius S, Tryggvadottir L, Olafsdottir GH, et al.: Linkage to BRCA2 region in hereditary male breast cancer. Lancet 346 (8974): 544-5, 1995. [PUBMED Abstract]
  9. Tai YC, Domchek S, Parmigiani G, et al.: Breast cancer risk among male BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 99 (23): 1811-4, 2007. [PUBMED Abstract]
  10. Ding YC, Steele L, Kuan CJ, et al.: Mutations in BRCA2 and PALB2 in male breast cancer cases from the United States. Breast Cancer Res Treat 126 (3): 771-8, 2011. [PUBMED Abstract]
  11. Silvestri V, Rizzolo P, Zanna I, et al.: PALB2 mutations in male breast cancer: a population-based study in Central Italy. Breast Cancer Res Treat 122 (1): 299-301, 2010. [PUBMED Abstract]
  12. Boyd J, Rhei E, Federici MG, et al.: Male breast cancer in the hereditary nonpolyposis colorectal cancer syndrome. Breast Cancer Res Treat 53 (1): 87-91, 1999. [PUBMED Abstract]
  13. Ravandi-Kashani F, Hayes TG: Male breast cancer: a review of the literature. Eur J Cancer 34 (9): 1341-7, 1998. [PUBMED Abstract]
  14. Mainiero MB, Lourenco AP, Barke LD, et al.: ACR Appropriateness Criteria Evaluation of the Symptomatic Male Breast. J Am Coll Radiol 12 (7): 678-82, 2015. [PUBMED Abstract]
  15. Giordano SH: A review of the diagnosis and management of male breast cancer. Oncologist 10 (7): 471-9, 2005. [PUBMED Abstract]
  16. Cardoso F, Paluch-Shimon S, Senkus E, et al.: 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann Oncol 31 (12): 1623-1649, 2020. [PUBMED Abstract]
  17. Wang F, Shu X, Meszoely I, et al.: Overall Mortality After Diagnosis of Breast Cancer in Men vs Women. JAMA Oncol 5 (11): 1589-1596, 2019. [PUBMED Abstract]

Stage Information for Male Breast Cancer

Staging for male breast cancer is identical to staging for female breast cancer. For more information, see the TNM Definitions section in Breast Cancer Treatment.

Treatment Option Overview for Male Breast Cancer

The approach to the treatment of men with breast cancer is similar to that for women. Because male breast cancer is rare, there is a lack of randomized data to support specific treatment modalities. Treatment options for men with breast cancer are described in Table 1.

Table 1. Treatment Options for Male Breast Cancer
Stage (TNM Definitions) Treatment Options
T = primary tumor; N = regional lymph node; M = distant metastasis; GnRH = gonadotropin-releasing hormone.
Early/localized/operable male breast cancer Surgery with or without radiation therapy
Adjuvant therapy
Locally advanced male breast cancer Neoadjuvant chemotherapy
Surgical excision
Radiation therapy and endocrine therapy
Metastatic male breast cancer Aromatase inhibitor therapy in conjunction with a GnRH agonist

Treatment of Early/Localized/Operable Male Breast Cancer

As in women, treatment options for men with early-stage breast cancer include:

Surgery With or Without Radiation Therapy

Primary treatment is a mastectomy with axillary lymph node dissection.[13] Responses in men are generally similar to those seen in women with breast cancer.[2] Breast conservation surgery with lumpectomy and radiation therapy has also been used and can be offered if standard criteria for breast conservation therapy are met. Results in men have been similar to those seen in women with breast cancer.[4]

For more information, see the Surgical Treatment for Breast Cancer section in Breast Cancer Treatment.

Adjuvant Therapy

The optimal systemic treatment in men with breast cancer has not been studied in randomized clinical trials. Adjuvant therapy should be administered according to the same criteria used for women. Adjuvant therapies used to treat early/localized/operable male breast cancer are outlined in Table 2. For more information, see the Systemic Therapy for Stages I, II, and III Breast Cancer section in Breast Cancer Treatment.

Table 2. Adjuvant Therapy Used to Treat Early/Localized/Operable Male Breast Cancer
Type of Adjuvant Therapy Agents Used
HER2 = human epidermal growth factor receptor 2; LHRH = luteinizing hormone-releasing hormone.
Chemotherapy Docetaxel and cyclophosphamide
Doxorubicin plus cyclophosphamide with or without paclitaxel
Endocrine therapy Tamoxifen [5]
Aromatase inhibitors with LHRH agonist [59]
HER2-directed therapy Trastuzumab [1,5]
Pertuzumab

Tamoxifen

Evidence (tamoxifen):

  1. A retrospective analysis of 257 men with stage I to stage III breast cancer included 50 men who were treated with an aromatase inhibitor (AI) and 207 men who were treated with tamoxifen.[10]
    • With a median follow-up of 42 months, treatment with an AI was associated with a higher risk of death compared with tamoxifen (32% with AI vs. 18% with tamoxifen; hazard ratio, 1.55; 95% confidence interval, 1.13–2.13).

In men with contraindications for tamoxifen, single-agent AI therapy is not recommended. AIs should be combined with gonadotropin-releasing hormone (GnRH) analogues.[6]

In male breast cancer patients, tamoxifen use is associated with a high rate of treatment-limiting symptoms such as hot flashes and impotence.[11]

The German Breast Group conducted a randomized phase II clinical trial (NCT01638247) of tamoxifen with or without a GnRH analogue versus AI plus a GnRH analogue in men with early-stage, hormone receptor–positive breast cancer. Results of this trial are pending.

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. Borgen PI, Wong GY, Vlamis V, et al.: Current management of male breast cancer. A review of 104 cases. Ann Surg 215 (5): 451-7; discussion 457-9, 1992. [PUBMED Abstract]
  2. Giordano SH, Buzdar AU, Hortobagyi GN: Breast cancer in men. Ann Intern Med 137 (8): 678-87, 2002. [PUBMED Abstract]
  3. Kinne DW: Management of male breast cancer. Oncology (Huntingt) 5 (3): 45-7; discussion 47-8, 1991. [PUBMED Abstract]
  4. Golshan M, Rusby J, Dominguez F, et al.: Breast conservation for male breast carcinoma. Breast 16 (6): 653-6, 2007. [PUBMED Abstract]
  5. Giordano SH: A review of the diagnosis and management of male breast cancer. Oncologist 10 (7): 471-9, 2005. [PUBMED Abstract]
  6. Giordano SH, Hortobagyi GN: Leuprolide acetate plus aromatase inhibition for male breast cancer. J Clin Oncol 24 (21): e42-3, 2006. [PUBMED Abstract]
  7. Cocconi G, Bisagni G, Ceci G, et al.: Low-dose aminoglutethimide with and without hydrocortisone replacement as a first-line endocrine treatment in advanced breast cancer: a prospective randomized trial of the Italian Oncology Group for Clinical Research. J Clin Oncol 10 (6): 984-9, 1992. [PUBMED Abstract]
  8. Gale KE, Andersen JW, Tormey DC, et al.: Hormonal treatment for metastatic breast cancer. An Eastern Cooperative Oncology Group Phase III trial comparing aminoglutethimide to tamoxifen. Cancer 73 (2): 354-61, 1994. [PUBMED Abstract]
  9. Zagouri F, Sergentanis TN, Koutoulidis V, et al.: Aromatase inhibitors with or without gonadotropin-releasing hormone analogue in metastatic male breast cancer: a case series. Br J Cancer 108 (11): 2259-63, 2013. [PUBMED Abstract]
  10. Eggemann H, Ignatov A, Smith BJ, et al.: Adjuvant therapy with tamoxifen compared to aromatase inhibitors for 257 male breast cancer patients. Breast Cancer Res Treat 137 (2): 465-70, 2013. [PUBMED Abstract]
  11. Anelli TF, Anelli A, Tran KN, et al.: Tamoxifen administration is associated with a high rate of treatment-limiting symptoms in male breast cancer patients. Cancer 74 (1): 74-7, 1994. [PUBMED Abstract]

Treatment of Locally Advanced Male Breast Cancer

Treatment options for men with locally advanced breast cancer include:[1]

  • Neoadjuvant chemotherapy.
  • Surgical excision.
  • Radiation therapy and endocrine therapy.

The decisions regarding the order and choice of treatments in men are guided by the same principles used for the treatment of breast cancer in women (in particular, evaluation of pathological response).[1,2]

For more information, see the Treatment of Locoregional Recurrent Breast Cancer section in Breast Cancer Treatment.

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. Giordano SH, Buzdar AU, Hortobagyi GN: Breast cancer in men. Ann Intern Med 137 (8): 678-87, 2002. [PUBMED Abstract]
  2. Kamila C, Jenny B, Per H, et al.: How to treat male breast cancer. Breast 16 (Suppl 2): S147-54, 2007. [PUBMED Abstract]

Treatment of Metastatic Male Breast Cancer

Treatment options for men with metastatic breast cancer include:

  • Aromatase inhibitor (AI) therapy in conjunction with a gonadotropin-releasing hormone (GnRH) agonist.

The management of metastatic hormone receptor–positive male breast cancer relies on the same treatment options used in women. However, data regarding the activity of AIs with GnRH agonists and fulvestrant in men are limited to case series.[14] The administration of an AI in conjunction with a GnRH agonist is recommended on the basis of the adjuvant data. There are no data comparing the activity of fulvestrant alone with fulvestrant in combination with a GnRH agonist.

Based on real world data and limited studies, it is reasonable to extrapolate the use of additional treatment options for men. These treatment options include cyclin-dependent kinase (CDK) 4/6 inhibitors, mammalian target of rapamycin (mTOR) inhibitors, and phosphatidylinositol-3 kinase (PI3K) inhibitors, used in combination with endocrine therapy.

The use of chemotherapy, human epidermal growth factor receptor 2 (HER2)-targeted therapy, immunotherapy, and poly (ADP-ribose) polymerase (PARP) inhibitors in men with metastatic breast cancer is guided by similar treatment principles as in women.[5,6]

For more information, see the Treatment of Metastatic Breast Cancer section in Breast Cancer Treatment.

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. Di Lauro L, Vici P, Barba M, et al.: Antiandrogen therapy in metastatic male breast cancer: results from an updated analysis in an expanded case series. Breast Cancer Res Treat 148 (1): 73-80, 2014. [PUBMED Abstract]
  2. Zagouri F, Sergentanis TN, Chrysikos D, et al.: Fulvestrant and male breast cancer: a pooled analysis. Breast Cancer Res Treat 149 (1): 269-75, 2015. [PUBMED Abstract]
  3. Zagouri F, Sergentanis TN, Koutoulidis V, et al.: Aromatase inhibitors with or without gonadotropin-releasing hormone analogue in metastatic male breast cancer: a case series. Br J Cancer 108 (11): 2259-63, 2013. [PUBMED Abstract]
  4. Di Lauro L, Vici P, Del Medico P, et al.: Letrozole combined with gonadotropin-releasing hormone analog for metastatic male breast cancer. Breast Cancer Res Treat 141 (1): 119-23, 2013. [PUBMED Abstract]
  5. Giordano SH, Buzdar AU, Hortobagyi GN: Breast cancer in men. Ann Intern Med 137 (8): 678-87, 2002. [PUBMED Abstract]
  6. Kamila C, Jenny B, Per H, et al.: How to treat male breast cancer. Breast 16 (Suppl 2): S147-54, 2007. [PUBMED Abstract]

Latest Updates to This Summary (02/28/2025)

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 Male Breast Cancer

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

This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.

About This PDQ Summary

Purpose of This Summary

This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of male breast 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 Male Breast Cancer Treatment are:

  • Fumiko Chino, MD (MD Anderson Cancer Center)
  • Tarek Hijal, MD (McGill University Health Centre)
  • Joseph L. Pater, MD (NCIC-Clinical Trials Group)
  • Carol Tweed, MD (Maryland Oncology Hematology)

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 Male Breast Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/breast/hp/male-breast-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389234]

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.

Male Breast Cancer Treatment (PDQ®)–Patient Version

Male Breast Cancer Treatment (PDQ®)–Patient Version

General Information about Male Breast Cancer

Key Points

  • Male breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast.
  • A family history of breast cancer and other factors can increase a man’s risk of breast cancer.
  • Male breast cancer is sometimes caused by inherited gene mutations (changes).
  • Men with breast cancer usually have lumps that can be felt.
  • Tests that examine the breasts are used to diagnose breast cancer in men.
  • If cancer is found, tests are done to study the cancer cells.
  • Survival for men with breast cancer is similar to survival for women with breast cancer.
  • Certain factors affect prognosis (chance of recovery) and treatment options.

Male breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast.

Breast cancer may occur in men. Breast cancer may occur in men at any age, but it usually occurs in men between 60 and 70 years of age. Male breast cancer makes up less than 1% of all cases of breast cancer.

The following types of breast cancer are found in men:

Lobular carcinoma in situ (abnormal cells found in one of the lobes or sections of the breast), which sometimes occurs in women, has not been seen in men.

EnlargeAnatomy of the male breast; drawing shows the lymph nodes, nipple, areola, chest wall, ribs, muscle, fatty tissue, and ducts.
Anatomy of the male breast. The nipple and areola are shown on the outside of the breast. The lymph nodes, fatty tissue, ducts, and other parts of the inside of the breast are also shown.

A family history of breast cancer and other factors can increase a man’s risk of breast cancer.

Anything that increases your risk of getting a disease is called a risk factor. Having a risk factor does not mean that you will get cancer; not having risk factors doesn’t mean that you will not get cancer. Talk with your doctor if you think you may be at risk. Risk factors for breast cancer in men may include the following:

Male breast cancer is sometimes caused by inherited gene mutations (changes).

The genes in cells carry the hereditary information that is received from a person’s parents. Hereditary breast cancer makes up about 5% to 10% of all breast cancer. Some mutated genes related to breast cancer, such as BRCA2, are more common in certain ethnic groups. Men who have a mutated gene related to breast cancer have an increased risk of this disease.

There are tests that can detect (find) mutated genes. These genetic tests are sometimes done for members of families with a high risk of cancer. See the following PDQ summaries for more information:

Men with breast cancer usually have lumps that can be felt.

Lumps and other signs may be caused by male breast cancer or by other conditions. Check with your doctor if you have any of the following:

  • A lump or thickening in or near the breast or in the underarm area.
  • A change in the size or shape of the breast.
  • A dimple or puckering in the skin of the breast.
  • A nipple turned inward into the breast.
  • Fluid from the nipple, especially if it’s bloody.
  • Scaly, red, or swollen skin on the breast, nipple, or areola (the dark area of skin around the nipple).
  • Dimples in the breast that look like the skin of an orange, called peau d’orange.

Tests that examine the breasts are used to diagnose breast cancer in men.

The following tests and procedures may be used:

  • Physical exam and health history: An exam of the body to check general signs of health, including checking for signs of disease, such as lumps or anything else that seems unusual. A history of the patient’s health habits and past illnesses and treatments will also be taken.
  • Clinical breast exam (CBE): An exam of the breast by a doctor or other health professional. The doctor will carefully feel the breasts and under the arms for lumps or anything else that seems unusual.
  • Mammogram: An x-ray of the breast.
  • Ultrasound exam: A procedure in which high-energy sound waves (ultrasound) are bounced off internal tissues or organs and make echoes. The echoes form a picture of body tissues called a sonogram. The picture can be printed to be looked at later.
  • MRI (magnetic resonance imaging): A procedure that uses a magnet, radio waves, and a computer to make a series of detailed pictures of both breasts. This procedure is also called nuclear magnetic resonance imaging (NMRI).
  • Blood chemistry studies: A procedure in which a blood sample is checked to measure the amounts of certain substances released into the blood by organs and tissues in the body. An unusual (higher or lower than normal) amount of a substance can be a sign of disease.
  • 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 four types of biopsies to check for breast cancer:

If cancer is found, tests are done to study the cancer cells.

Decisions about the best treatment are based on the results of these tests. The tests give information about:

  • How quickly the cancer may grow.
  • How likely it is that the cancer will spread through the body.
  • How well certain treatments might work.
  • How likely the cancer is to recur (come back).

Tests include the following:

Survival for men with breast cancer is similar to survival for women with breast cancer.

Survival for men with breast cancer is similar to that for women with breast cancer when their stage at diagnosis is the same. Breast cancer in men, however, is often diagnosed at a later stage. Cancer found at a later stage may be less likely to be cured.

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

The prognosis and treatment options depend on the following:

  • The stage of the cancer (the size of the tumor and whether it is in the breast only or has spread to lymph nodes or other places in the body).
  • The type of breast cancer.
  • Estrogen-receptor and progesterone-receptor levels in the tumor tissue.
  • Whether the cancer is also found in the other breast.
  • The man’s age and general health.
  • Whether the cancer has just been diagnosed or has recurred (come back).

Stages of Male Breast Cancer

Key Points

  • After breast cancer has been diagnosed, tests are done to find out if cancer cells have spread within the breast 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.
  • In breast cancer, stage is based on the size and location of the primary tumor, the spread of cancer to nearby lymph nodes or other parts of the body, tumor grade, and whether certain biomarkers are present.
  • The TNM system is used to describe the size of the primary tumor and the spread of cancer to nearby lymph nodes or other parts of the body.
    • Tumor (T). The size and location of the tumor.
    • Lymph Node (N). The size and location of lymph nodes where cancer has spread.
    • Metastasis (M). The spread of cancer to other parts of the body.
  • The grading system is used to describe how quickly a breast tumor is likely to grow and spread.
  • Biomarker testing is used to find out whether breast cancer cells have certain receptors.
  • The TNM system, the grading system, and biomarker status are combined to find out the breast cancer stage.
  • Talk to your doctor to find out what your breast cancer stage is and how it is used to plan the best treatment for you.
  • The treatment of male breast cancer depends partly on the stage of the disease.

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

After breast cancer has been diagnosed, tests are done to find out if cancer cells have spread within the breast or to other parts of the body. This process 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. Breast cancer in men is staged the same as it is in women. The spread of cancer from the breast to lymph nodes and other parts of the body appears to be similar in men and women.

The following tests and procedures may be used in the staging process:

  • Sentinel lymph node biopsy: The removal of the sentinel lymph node during surgery. The sentinel lymph node is the first lymph node in a group of lymph nodes to receive lymphatic drainage from the primary tumor. It is the first lymph node the cancer is likely to spread to from the primary tumor. A radioactive substance and/or blue dye is injected near the tumor. The substance or dye flows through the lymph ducts to the lymph nodes. The first lymph node to receive the substance or dye is removed. A pathologist views the tissue under a microscope to look for cancer cells. If cancer cells are not found, it may not be necessary to remove more lymph nodes. Sometimes, a sentinel lymph node is found in more than one group of nodes.
  • 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.
  • 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.
  • 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.
  • 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.

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 breast cancer spreads to the bone, the cancer cells in the bone are actually breast cancer cells. The disease is metastatic breast cancer, not bone 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.

In breast cancer, stage is based on the size and location of the primary tumor, the spread of cancer to nearby lymph nodes or other parts of the body, tumor grade, and whether certain biomarkers are present.

To plan the best treatment and understand your prognosis, it is important to know the breast cancer stage.

There are 3 types of breast cancer stage groups:

  • Clinical Prognostic Stage is used first to assign a stage for all patients based on health history, physical exam, imaging tests (if done), and biopsies. The Clinical Prognostic Stage is described by the TNM system, tumor grade, and biomarker status (ER, PR, HER2). In clinical staging, mammography or ultrasound is used to check the lymph nodes for signs of cancer.
  • Pathological Prognostic Stage is then used for patients who have surgery as their first treatment. The Pathological Prognostic Stage is based on all clinical information, biomarker status, and laboratory test results from breast tissue and lymph nodes removed during surgery.
  • Anatomic Stage is based on the size and the spread of cancer as described by the TNM system. The Anatomic Stage is used in parts of the world where biomarker testing is not available. It is not used in the United States.

The TNM system is used to describe the size of the primary tumor and the spread of cancer to nearby lymph nodes or other parts of the body.

For breast cancer, the TNM system describes the tumor as follows:

Tumor (T). The size and location of the tumor.

EnlargeDrawing shows different sizes of common items in millimeters (mm): a sharp pencil point (1 mm), a new crayon point (2 mm), a pencil-top eraser (5 mm), a pea (10 mm), a peanut (20 mm), and a lime (50 mm). Also shown is a 2-centimeter (cm) ruler that shows 10 mm is equal to 1 cm.
Tumor sizes are often measured in millimeters (mm) or centimeters. Common items that can be used to show tumor size in mm include: a sharp pencil point (1 mm), a new crayon point (2 mm), a pencil-top eraser (5 mm), a pea (10 mm), a peanut (20 mm), and a lime (50 mm).
  • TX: Primary tumor cannot be assessed.
  • T0: No sign of a primary tumor in the breast.
  • Tis: Carcinoma in situ. There are 2 types of breast carcinoma in situ:
    • Tis (DCIS): DCIS is a condition in which abnormal cells are found in the lining of a breast duct. The abnormal cells have not spread outside the duct to other tissues in the breast. In some cases, DCIS may become invasive breast cancer that is able to spread to other tissues. At this time, there is no way to know which lesions can become invasive.
    • Tis (Paget disease): Paget disease of the nipple is a condition in which abnormal cells are found in the skin cells of the nipple and may spread to the areola. It is not staged according to the TNM system. If Paget disease AND an invasive breast cancer are present, the TNM system is used to stage the invasive breast cancer.
  • T1: The tumor is 20 millimeters or smaller. There are 4 subtypes of a T1 tumor depending on the size of the tumor:
    • T1mi: the tumor is 1 millimeter or smaller.
    • T1a: the tumor is larger than 1 millimeter but not larger than 5 millimeters.
    • T1b: the tumor is larger than 5 millimeters but not larger than 10 millimeters.
    • T1c: the tumor is larger than 10 millimeters but not larger than 20 millimeters.
  • T2: The tumor is larger than 20 millimeters but not larger than 50 millimeters.
  • T3: The tumor is larger than 50 millimeters.
  • T4: The tumor is described as one of the following:
    • T4a: the tumor has grown into the chest wall.
    • T4b: the tumor has grown into the skin—an ulcer has formed on the surface of the skin on the breast, small tumor nodules have formed in the same breast as the primary tumor, and/or there is swelling of the skin on the breast.
    • T4c: the tumor has grown into the chest wall and the skin.
    • T4d: inflammatory breast cancer—one-third or more of the skin on the breast is red and swollen (called peau d’orange).

Lymph Node (N). The size and location of lymph nodes where cancer has spread.

When the lymph nodes are removed by surgery and studied under a microscope by a pathologist, pathologic staging is used to describe the lymph nodes. The pathologic staging of lymph nodes is described below.

  • NX: The lymph nodes cannot be assessed.
  • N0: No sign of cancer in the lymph nodes, or tiny clusters of cancer cells not larger than 0.2 millimeters in the lymph nodes.
  • N1: Cancer is described as one of the following:
    • N1mi: cancer has spread to the axillary (armpit area) lymph nodes and is larger than 0.2 millimeters but not larger than 2 millimeters.
    • N1a: cancer has spread to 1 to 3 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters.
    • N1b: cancer has spread to lymph nodes near the breastbone on the same side of the body as the primary tumor, and the cancer is larger than 0.2 millimeters and is found by sentinel lymph node biopsy. Cancer is not found in the axillary lymph nodes.
    • N1c: cancer has spread to 1 to 3 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters. Cancer is also found by sentinel lymph node biopsy in the lymph nodes near the breastbone on the same side of the body as the primary tumor.
  • N2: Cancer is described as one of the following:
    • N2a: cancer has spread to 4 to 9 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters.
    • N2b: cancer has spread to lymph nodes near the breastbone and the cancer is found by imaging tests. Cancer is not found in the axillary lymph nodes by sentinel lymph node biopsy or lymph node dissection.
  • N3: Cancer is described as one of the following:
    • N3a: cancer has spread to 10 or more axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters, or cancer has spread to lymph nodes below the collarbone.
    • N3b: cancer has spread to 1 to 9 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters. Cancer has also spread to lymph nodes near the breastbone and the cancer is found by imaging tests;

      or

      cancer has spread to 4 to 9 axillary lymph nodes and cancer in at least one of the lymph nodes is larger than 2 millimeters. Cancer has also spread to lymph nodes near the breastbone on the same side of the body as the primary tumor, and the cancer is larger than 0.2 millimeters and is found by sentinel lymph node biopsy.

    • N3c: cancer has spread to lymph nodes above the collarbone on the same side of the body as the primary tumor.

When the lymph nodes are checked using mammography or ultrasound, it is called clinical staging. The clinical staging of lymph nodes is not described here.

Metastasis (M). The spread of cancer to other parts of the body.

  • M0: There is no sign that cancer has spread to other parts of the body.
  • M1: Cancer has spread to other parts of the body, most often the bones, lungs, liver, or brain. If cancer has spread to distant lymph nodes, the cancer in the lymph nodes is larger than 0.2 millimeters. The cancer is called metastatic breast cancer.

The grading system is used to describe how quickly a breast tumor is likely to grow and spread.

The grading system describes a tumor based on how abnormal the cancer cells and tissue look under a microscope and how quickly the cancer cells are likely to grow and spread. Low-grade cancer cells look more like normal cells and tend to grow and spread more slowly than high-grade cancer cells. To describe how abnormal the cancer cells and tissue are, the pathologist will assess the following three features:

  • How much of the tumor tissue has normal breast ducts.
  • The size and shape of the nuclei in the tumor cells.
  • How many dividing cells are present, which is a measure of how fast the tumor cells are growing and dividing.

For each feature, the pathologist assigns a score of 1 to 3; a score of “1” means the cells and tumor tissue look the most like normal cells and tissue, and a score of “3” means the cells and tissue look the most abnormal. The scores for each feature are added together to get a total score between 3 and 9.

Three grades are possible:

  • Total score of 3 to 5: G1 (Low grade or well differentiated).
  • Total score of 6 to 7: G2 (Intermediate grade or moderately differentiated).
  • Total score of 8 to 9: G3 (High grade or poorly differentiated).

Biomarker testing is used to find out whether breast cancer cells have certain receptors.

Healthy breast cells, and some breast cancer cells, have receptors (biomarkers) that attach to the hormones estrogen and progesterone. These hormones are needed for healthy cells, and some breast cancer cells, to grow and divide. To check for these biomarkers, samples of tissue containing breast cancer cells are removed during a biopsy or surgery. The samples are tested in a laboratory to see whether the breast cancer cells have estrogen or progesterone receptors.

Another type of receptor (biomarker) that is found on the surface of all breast cancer cells is called HER2. HER2 receptors are needed for the breast cancer cells to grow and divide.

For breast cancer, biomarker testing includes the following:

  • Estrogen receptor (ER). If the breast cancer cells have estrogen receptors, the cancer cells are called ER positive (ER+). If the breast cancer cells do not have estrogen receptors, the cancer cells are called ER negative (ER-).
  • Progesterone receptor (PR). If the breast cancer cells have progesterone receptors, the cancer cells are called PR positive (PR+). If the breast cancer cells do not have progesterone receptors, the cancer cells are called PR negative (PR-).
  • Human epidermal growth factor type 2 receptor (HER2/neu or HER2). If the breast cancer cells have larger than normal amounts of HER2 receptors on their surface, the cancer cells are called HER2 positive (HER2+). If the breast cancer cells have a normal amount of HER2 on their surface, the cancer cells are called HER2 negative (HER2-). HER2+ breast cancer is more likely to grow and divide faster than HER2- breast cancer.

Sometimes the breast cancer cells will be described as triple negative or triple positive.

  • Triple negative. If the breast cancer cells do not have estrogen receptors, progesterone receptors, or a larger than normal amount of HER2 receptors, the cancer cells are called triple negative.
  • Triple positive. If the breast cancer cells do have estrogen receptors, progesterone receptors, and a larger than normal amount of HER2 receptors, the cancer cells are called triple positive.

It is important to know the estrogen receptor, progesterone receptor, and HER2 receptor status to choose the best treatment. There are drugs that can stop the receptors from attaching to the hormones estrogen and progesterone and stop the cancer from growing. Other drugs may be used to block the HER2 receptors on the surface of the breast cancer cells and stop the cancer from growing.

The TNM system, the grading system, and biomarker status are combined to find out the breast cancer stage.

Here are 3 examples that combine the TNM system, the grading system, and the biomarker status to find out the Pathological Prognostic breast cancer stage for a woman whose first treatment was surgery:

If the tumor size is 30 millimeters (T2), has not spread to nearby lymph nodes (N0), has not spread to distant parts of the body (M0), and is:

  • Grade 1
  • HER2+
  • ER-
  • PR-

The cancer is stage IIA.

If the tumor size is 53 millimeters (T3), has spread to 4 to 9 axillary lymph nodes (N2), has not spread to other parts of the body (M0), and is:

  • Grade 2
  • HER2+
  • ER+
  • PR-

The tumor is stage IIIA.

If the tumor size is 65 millimeters (T3), has spread to 3 axillary lymph nodes (N1a), has spread to the lungs (M1), and is:

  • Grade 1
  • HER2+
  • ER-
  • PR-

The cancer is stage IV (metastatic breast cancer).

Talk to your doctor to find out what your breast cancer stage is and how it is used to plan the best treatment for you.

After surgery, your doctor will receive a pathology report that describes the size and location of the primary tumor, the spread of cancer to nearby lymph nodes, tumor grade, and whether certain biomarkers are present. The pathology report and other test results are used to determine your breast cancer stage.

You are likely to have many questions. Ask your doctor to explain how staging is used to decide the best options to treat your cancer and whether there are clinical trials that might be right for you.

The treatment of male breast cancer depends partly on the stage of the disease.

For treatment options for stage I, stage II, stage IIIA, and operable stage IIIC breast cancer, see Treatment of Early/Localized/Operable Male Breast Cancer.

For treatment options for cancer that has recurred (come back) near the area where it first formed, see Treatment of Locoregional Recurrent Male Breast Cancer.

For treatment options for stage IV (metastatic) breast cancer or breast cancer that has recurred in other parts of the body, see Treatment of Metastatic Male Breast Cancer.

Inflammatory Male Breast Cancer

In inflammatory breast cancer, cancer has spread to the skin of the breast and the breast looks red and swollen and feels warm. The redness and warmth occur because the cancer cells block the lymph vessels in the skin. The skin of the breast may also show the dimpled appearance called peau d’orange (like the skin of an orange). There may not be any lumps in the breast that can be felt. Inflammatory breast cancer may be stage IIIB, stage IIIC, or stage IV.

Treatment Option Overview

Key Points

  • There are different types of treatment for men with breast cancer.
  • Five types of standard treatment are used to treat men with breast cancer:
    • Surgery
    • Chemotherapy
    • Hormone therapy
    • Radiation therapy
    • Targeted therapy
  • Treatment for male breast cancer may cause side effects.

There are different types of treatment for men with breast cancer.

Different types of treatment are available for men with breast 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.

For some patients, taking part in a clinical trial may be the best treatment choice. 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.

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 is available from the NCI website. Choosing the most appropriate cancer treatment is a decision that ideally involves the patient, family, and health care team.

Five types of standard treatment are used to treat men with breast cancer:

Surgery

Surgery for men with breast cancer is usually a modified radical mastectomy, surgery to remove the whole breast that has cancer. This may include removal of the nipple, areola (the dark-colored skin around the nipple), and skin over the breast. Most of the lymph nodes under the arm are also removed.

EnlargeModified radical mastectomy; the drawing on the left shows the removal of the whole breast, including the lymph nodes under the arm. The drawing on the right shows a cross-section of the breast, including the fatty tissue and chest wall (ribs and muscle). A tumor is also shown in the breast.
Modified radical mastectomy. The whole breast and most of the lymph nodes under the arm are removed.

Breast-conserving surgery, an operation to remove the cancer but not the breast itself, is also used for some men with breast cancer. A lumpectomy is done to remove the tumor (lump) and a small amount of normal tissue around it. Radiation therapy is given after surgery to kill any cancer cells that are left.

EnlargeLumpectomy; the drawing on the left shows removal of the tumor and some of the normal tissue around it. The drawing on the right shows removal of some of the lymph nodes under the arm and removal of the tumor and part of the chest wall lining near the tumor. Also shown is fatty tissue in the breast.
Lumpectomy. The tumor and some normal tissue around it are removed, but not the breast itself. Some lymph nodes under the arm may also be removed. If the cancer is near the chest wall, part of the chest wall lining may be removed as well.

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).

See Drugs Approved for Breast Cancer for more information.

Hormone therapy

Hormone therapy is a cancer treatment that removes hormones or blocks their action and stops cancer cells from growing. Hormones are substances made by glands in the body and circulated in the bloodstream. Some hormones can cause certain cancers to grow. If tests show that the cancer cells have places where hormones can attach (receptors), drugs, surgery, or radiation therapy is used to reduce the production of hormones or block them from working.

Hormone therapy with tamoxifen is often given to patients with estrogen-receptor and progesterone-receptor positive breast cancer and to patients with metastatic breast cancer (cancer that has spread to other parts of the body).

Hormone therapy with an aromatase inhibitor is given to some men who have metastatic breast cancer. Aromatase inhibitors decrease the body’s estrogen by blocking an enzyme called aromatase from turning androgen into estrogen. Anastrozole, letrozole, and exemestane are types of aromatase inhibitors.

Hormone therapy with a luteinizing hormone-releasing hormone (LHRH) agonist is given to some men who have metastatic breast cancer. LHRH agonists affect the pituitary gland, which controls how much testosterone is made by the testicles. In men who are taking LHRH agonists, the pituitary gland tells the testicles to make less testosterone. Leuprolide and goserelin are types of LHRH agonists.

Other types of hormone therapy include megestrol acetate or anti-estrogen therapy, such as fulvestrant.

See Drugs Approved for Breast Cancer for more information.

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.

Targeted therapy

Targeted therapy is a type of treatment that uses drugs or other substances to identify and attack specific cancer cells. Targeted therapies usually cause less harm to normal cells than chemotherapy or radiation therapy do. Monoclonal antibody therapy, tyrosine kinase inhibitors, cyclin-dependent kinase inhibitors, and mammalian target of rapamycin (mTOR) inhibitors are types of targeted therapies used to treat men with breast cancer.

Monoclonal antibodies are immune system proteins made in the laboratory to treat many diseases, including cancer. As a cancer treatment, these antibodies can attach to a specific target on cancer cells or other cells that may help cancer cells grow. The antibodies are able to then kill the cancer cells, block their growth, or keep them from spreading. Monoclonal antibodies are given by infusion. They may be used alone or to carry drugs, toxins, or radioactive material directly to cancer cells.

Types of monoclonal antibody therapy include the following:

How do monoclonal antibodies work to treat cancer? This video shows how monoclonal antibodies, such as trastuzumab, pembrolizumab, and rituximab, block molecules cancer cells need to grow, flag cancer cells for destruction by the body’s immune system, or deliver harmful substances to cancer cells.

Tyrosine kinase inhibitors are targeted therapy drugs that block signals needed for tumors to grow. Lapatinib is a tyrosine kinase inhibitor that may be used to treat men with metastatic breast cancer.

Cyclin-dependent kinase inhibitors are targeted therapy drugs that block proteins called cyclin-dependent kinases, which cause the growth of cancer cells. Palbociclib is a cyclin-dependent kinase inhibitor used to treat men with metastatic breast cancer.

Mammalian target of rapamycin (mTOR) inhibitors block a protein called mTOR, which may keep cancer cells from growing and prevent the growth of new blood vessels that tumors need to grow.

See Drugs Approved for Breast Cancer for more information.

Treatment for male breast cancer may cause side effects.

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

Treatment of Early/Localized/Operable Male Breast Cancer

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

Treatment of early, localized, or operable breast cancer may include the following:

Initial Surgery

Treatment for men diagnosed with breast cancer is usually modified radical mastectomy.

Breast-conserving surgery with lumpectomy followed by radiation therapy may be used for some men.

Adjuvant Therapy

Therapy given after an operation when cancer cells can no longer be seen is called adjuvant therapy. Even if the doctor removes all the cancer that can be seen at the time of the operation, the patient may be given radiation therapy, chemotherapy, hormone therapy, and/or targeted therapy after surgery, to try to kill any cancer cells that may be left.

  • Node-negative: For men whose cancer is node-negative (cancer has not spread to the lymph nodes), adjuvant therapy should be considered on the same basis as for a woman with breast cancer because there is no evidence that response to therapy is different for men and women.
  • Node-positive: For men whose cancer is node-positive (cancer has spread to the lymph nodes), adjuvant therapy may include the following:

These treatments appear to increase survival in men as they do in women. The patient’s response to hormone therapy depends on whether there are hormone receptors (proteins) in the tumor. Most breast cancers in men have these receptors. Hormone therapy is usually recommended for male breast cancer patients, but it can have many side effects, including hot flashes and impotence (the inability to have an erection adequate for sexual intercourse).

Treatment of Locoregional Recurrent Male Breast Cancer

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

For men with locally recurrent disease (cancer that has come back in a limited area after treatment), treatment options include:

Treatment of Metastatic Male Breast Cancer

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

Treatment options for metastatic breast cancer (cancer that has spread to distant parts of the body) may include the following:

Hormone therapy

In men who have just been diagnosed with metastatic breast cancer that is hormone receptor positive or if the hormone receptor status is not known, treatment may include:

In men whose tumors are hormone receptor positive or hormone receptor unknown, with spread to the bone or soft tissue only, and who have been treated with tamoxifen, treatment may include:

Targeted therapy

In men with metastatic breast cancer that is hormone receptor positive and has not responded to other treatments, options may include targeted therapy such as:

In men with metastatic breast cancer that is HER2/neu positive, treatment may include:

  • Targeted therapy such as trastuzumab, pertuzumab, ado-trastuzumab emtansine, or lapatinib.

Chemotherapy

In men with metastatic breast cancer that is hormone receptor negative, has not responded to hormone therapy, has spread to other organs or has caused symptoms, treatment may include:

Surgery

  • Total mastectomy for men with open or painful breast lesions. Radiation therapy may be given after surgery.
  • Surgery to remove cancer that has spread to the brain or spine. Radiation therapy may be given after surgery.
  • Surgery to remove cancer that has spread to the lung.
  • Surgery to repair or help support weak or broken bones. Radiation therapy may be given after surgery.
  • Surgery to remove fluid that has collected around the lungs or heart.

Radiation therapy

Other treatment options

Other treatment options for metastatic breast cancer include:

To Learn More About Male Breast 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 male breast 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 Male Breast Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/breast/patient/male-breast-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389417]

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.

Breast Cancer Treatment During Pregnancy (PDQ®)–Patient Version

Breast Cancer Treatment During Pregnancy (PDQ®)–Patient Version

General Information About Breast Cancer Treatment During Pregnancy

Key Points

  • Breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast.
  • Sometimes breast cancer occurs in women who are pregnant or have just given birth.
  • Signs of breast cancer include a lump or change in the breast.
  • It may be difficult to detect (find) breast cancer early in pregnant or nursing women.
  • Breast exams should be part of prenatal and postnatal care.
  • Tests that examine the breasts are used to diagnose breast cancer.
  • If cancer is found, tests are done to study the cancer cells.
  • Certain factors affect prognosis (chance of recovery) and treatment options.

Breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast.

The breast is made up of lobes and ducts. Each breast has 15 to 20 sections called lobes. Each lobe has many smaller sections called lobules. Lobules end in dozens of tiny bulbs that can make milk. The lobes, lobules, and bulbs are linked by thin tubes called ducts.

EnlargeIllustration of the female breast anatomy. On the left, a front view shows lymph nodes inside the breast going from the breast to the armpit. On the right, a cross-section shows the chest wall, ribs, fatty tissue, lobes, ducts, and lobules. Also shown in both panels are the muscle, nipple, and areola.
The female breast contains lobes, lobules, and ducts that produce and transport milk to the nipple. Fatty tissue gives the breast its shape, while muscles and the chest wall provide support. The lymphatic system, including lymph nodes, filter lymph and store white blood cells that help fight infection and disease.

Each breast also has blood vessels and lymph vessels. The lymph vessels carry an almost colorless, watery fluid called lymph. Lymph vessels carry lymph between lymph nodes. Lymph nodes are small, bean-shaped structures found throughout the body. They filter lymph and store white blood cells that help fight infection and disease. Groups of lymph nodes are found near the breast in the axilla (under the arm), above the collarbone, and in the chest.

Sometimes breast cancer occurs in women who are pregnant or have just given birth.

Breast cancer occurs about once in every 3,000 pregnancies. It occurs most often in women aged 32 to 38 years. Because many women are choosing to delay having children, it is likely that the number of new cases of breast cancer during pregnancy will increase.

Signs of breast cancer include a lump or change in the breast.

These and other signs may be caused by breast cancer or by other conditions. Check with your doctor if you have any of the following:

  • A lump or thickening in or near the breast or in the underarm area.
  • A change in the size or shape of the breast.
  • A dimple or puckering in the skin of the breast.
  • A nipple turned inward into the breast.
  • Fluid, other than breast milk, from the nipple, especially if it’s bloody.
  • Scaly, red, or swollen skin on the breast, nipple, or areola (the dark area of skin around the nipple).
  • Dimples in the breast that look like the skin of an orange, called peau d’orange.

It may be difficult to detect (find) breast cancer early in pregnant or nursing women.

The breasts usually get larger, tender, or lumpy in women who are pregnant, nursing, or have just given birth. This occurs because of normal hormone changes that take place during pregnancy. These changes can make small lumps difficult to detect. The breasts may also become denser. It is more difficult to detect breast cancer in women with dense breasts using mammography. Because these breast changes can delay diagnosis, breast cancer is often found at a later stage in these women.

Breast exams should be part of prenatal and postnatal care.

To detect breast cancer, pregnant and nursing women should examine their breasts themselves. Women should also receive clinical breast exams during their regular prenatal and postnatal check-ups. Talk to your doctor if you notice any changes in your breasts that you do not expect or that worry you.

Tests that examine the breasts are used to diagnose breast cancer.

The following tests and procedures may be used:

  • Physical exam and health history: An exam of the body to check general signs of health, including checking for signs of disease, such as lumps or anything else that seems unusual. A history of the patient’s health habits and past illnesses and treatments will also be taken.
  • Clinical breast exam (CBE): An exam of the breast by a doctor or other health professional. The doctor will carefully feel the breasts and under the arms for lumps or anything else that seems unusual.
  • Ultrasound exam: A procedure in which high-energy sound waves (ultrasound) are bounced off internal tissues or organs and make echoes. The echoes form a picture of body tissues called a sonogram. The picture can be printed to look at later.
  • Mammogram: An x-ray of the breast. A mammogram can be done with little risk to the fetus. Mammograms in pregnant women may appear negative even though cancer is present.
    EnlargeDrawing of a woman standing with her left breast pressed between two plates of a mammography machine. Behind her, a health professional uses an X-ray machine to take pictures of the breast. An inset shows the X-ray film image with an arrow pointed at abnormal tissue.
    Mammography is an imaging test used to screen for and diagnose breast cancer. It can detect abnormal breast tissue, including cancer, sometimes before symptoms appear.
  • Biopsy: The removal of cells or tissues so they can be viewed under a microscope by a pathologist to check for signs of cancer. If a lump in the breast is found, a biopsy may be done.

    There are three types of breast biopsies:

If cancer is found, tests are done to study the cancer cells.

Decisions about the best treatment are based on the results of these tests and the trimester of the pregnancy. The tests give information about:

  • How quickly the cancer may grow.
  • How likely it is that the cancer will spread to other parts of the body.
  • How well certain treatments might work.
  • How likely the cancer is to recur (come back).

Tests may include the following:

  • Estrogen and progesterone receptor test: A test to measure the amount of estrogen and progesterone (hormones) receptors in cancer tissue. If there are more estrogen or progesterone receptors than normal, the cancer is called estrogen receptor positive or progesterone receptor positive. This type of breast cancer may grow more quickly. The test results show whether treatment to block estrogen and progesterone given after the baby is born may stop the cancer from growing.
  • Human epidermal growth factor type 2 receptor (HER2/neu) test: A laboratory test to measure how many HER2/neu genes there are and how much HER2/neu protein is made in a sample of tissue. If there are more HER2/neu genes or higher levels of HER2/neu protein than normal, the cancer is called HER2/neu positive. This type of breast cancer may grow more quickly and is more likely to spread to other parts of the body. The cancer may be treated with drugs that target the HER2/neu protein, such as trastuzumab and pertuzumab, after the baby is born.
  • Multigene tests: Tests in which samples of tissue are studied to look at the activity of many genes at the same time. These tests may help predict whether cancer will spread to other parts of the body or recur (come back).
    • Oncotype DX: This test helps predict whether stage I or stage II breast cancer that is estrogen receptor positive and node-negative will spread to other parts of the body. If the risk of the cancer spreading is high, chemotherapy may be given to lower the risk.
    • MammaPrint: A laboratory test in which the activity of 70 different genes is looked at in the breast cancer tissue of women who have early-stage invasive breast cancer that has not spread to lymph nodes or has spread to 3 or fewer lymph nodes. The activity level of these genes helps predict whether breast cancer will spread to other parts of the body or come back. If the test shows that the risk that the cancer will spread or come back is high, chemotherapy may be given to lower the risk.

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

The prognosis and treatment options depend on the following:

  • The stage of the cancer (the size of the tumor and whether it is in the breast only or has spread to other parts of the body).
  • The type of breast cancer.
  • The trimester of the pregnancy.
  • Whether there are signs or symptoms.
  • The patient’s general health.

Stages of Breast Cancer

Key Points

  • After breast cancer has been diagnosed, tests are done to find out if cancer cells have spread within the breast 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.
  • In breast cancer, stage is based on the size and location of the primary tumor, the spread of cancer to nearby lymph nodes or other parts of the body, tumor grade, and whether certain biomarkers are present.
  • The TNM system is used to describe the size of the primary tumor and the spread of cancer to nearby lymph nodes or other parts of the body.
    • Tumor (T). The size and location of the tumor.
    • Lymph Node (N). The size and location of lymph nodes where cancer has spread.
    • Metastasis (M). The spread of cancer to other parts of the body.
  • The grading system is used to describe how quickly a breast tumor is likely to grow and spread.
  • Biomarker testing is used to find out whether breast cancer cells have certain receptors.
  • The TNM system, the grading system, and biomarker status are combined to find out the breast cancer stage.
  • Talk to your doctor to find out what your breast cancer stage is and how it is used to plan the best treatment for you.

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

The process used to find out if the cancer has spread within the breast 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.

Some procedures may expose the fetus to harmful radiation or dyes. These procedures are done only if absolutely necessary. Certain actions, such as using a lead-lined shield to cover the abdomen, are used to help protect the fetus from radiation as much as possible.

The following tests and procedures may be used to stage breast cancer during pregnancy:

  • 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.
  • 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 bones with cancer and is detected by a scanner.
  • Ultrasound exam: A procedure in which high-energy sound waves (ultrasound) are bounced off internal tissues or organs, such as the liver, and make echoes. The echoes form a picture of body tissues called a sonogram. The picture can be printed to be looked at later.
  • 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, such as the brain. This procedure is also called nuclear magnetic resonance imaging (NMRI).

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 breast cancer spreads to the bone, the cancer cells in the bone are actually breast cancer cells. The disease is metastatic breast cancer, not bone 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.

In breast cancer, stage is based on the size and location of the primary tumor, the spread of cancer to nearby lymph nodes or other parts of the body, tumor grade, and whether certain biomarkers are present.

To plan the best treatment and understand your prognosis, it is important to know the breast cancer stage.

There are 3 types of breast cancer stage groups:

  • Clinical Prognostic Stage is used first to assign a stage for all patients based on health history, physical exam, imaging tests (if done), and biopsies. The Clinical Prognostic Stage is described by the TNM system, tumor grade, and biomarker status (ER, PR, HER2). In clinical staging, mammography or ultrasound is used to check the lymph nodes for signs of cancer.
  • Pathological Prognostic Stage is then used for patients who have surgery as their first treatment. The Pathological Prognostic Stage is based on all clinical information, biomarker status, and laboratory test results from breast tissue and lymph nodes removed during surgery.
  • Anatomic Stage is based on the size and the spread of cancer as described by the TNM system. The Anatomic Stage is used in parts of the world where biomarker testing is not available. It is not used in the United States.

The TNM system is used to describe the size of the primary tumor and the spread of cancer to nearby lymph nodes or other parts of the body.

For breast cancer, the TNM system describes the tumor as follows:

Tumor (T). The size and location of the tumor.

EnlargeDrawing shows different sizes of common items in millimeters (mm): a sharp pencil point (1 mm), a new crayon point (2 mm), a pencil-top eraser (5 mm), a pea (10 mm), a peanut (20 mm), and a lime (50 mm). Also shown is a 2-centimeter (cm) ruler that shows 10 mm is equal to 1 cm.
Tumor sizes are often measured in millimeters (mm) or centimeters. Common items that can be used to show tumor size in mm include: a sharp pencil point (1 mm), a new crayon point (2 mm), a pencil-top eraser (5 mm), a pea (10 mm), a peanut (20 mm), and a lime (50 mm).
  • TX: Primary tumor cannot be assessed.
  • T0: No sign of a primary tumor in the breast.
  • Tis: Carcinoma in situ. There are 2 types of breast carcinoma in situ:
    • Tis (DCIS): DCIS is a condition in which abnormal cells are found in the lining of a breast duct. The abnormal cells have not spread outside the duct to other tissues in the breast. In some cases, DCIS may become invasive breast cancer that is able to spread to other tissues. At this time, there is no way to know which lesions can become invasive.
    • Tis (Paget disease): Paget disease of the nipple is a condition in which abnormal cells are found in the skin cells of the nipple and may spread to the areola. It is not staged according to the TNM system. If Paget disease AND an invasive breast cancer are present, the TNM system is used to stage the invasive breast cancer.
  • T1: The tumor is 20 millimeters or smaller. There are 4 subtypes of a T1 tumor depending on the size of the tumor:
    • T1mi: the tumor is 1 millimeter or smaller.
    • T1a: the tumor is larger than 1 millimeter but not larger than 5 millimeters.
    • T1b: the tumor is larger than 5 millimeters but not larger than 10 millimeters.
    • T1c: the tumor is larger than 10 millimeters but not larger than 20 millimeters.
  • T2: The tumor is larger than 20 millimeters but not larger than 50 millimeters.
  • T3: The tumor is larger than 50 millimeters.
  • T4: The tumor is described as one of the following:
    • T4a: the tumor has grown into the chest wall.
    • T4b: the tumor has grown into the skin—an ulcer has formed on the surface of the skin on the breast, small tumor nodules have formed in the same breast as the primary tumor, and/or there is swelling of the skin on the breast.
    • T4c: the tumor has grown into the chest wall and the skin.
    • T4d: inflammatory breast cancer—one-third or more of the skin on the breast is red and swollen (called peau d’orange).

Lymph Node (N). The size and location of lymph nodes where cancer has spread.

When the lymph nodes are removed by surgery and studied under a microscope by a pathologist, pathologic staging is used to describe the lymph nodes. The pathologic staging of lymph nodes is described below.

  • NX: The lymph nodes cannot be assessed.
  • N0: No sign of cancer in the lymph nodes, or tiny clusters of cancer cells not larger than 0.2 millimeters in the lymph nodes.
  • N1: Cancer is described as one of the following:
    • N1mi: cancer has spread to the axillary (armpit area) lymph nodes and is larger than 0.2 millimeters but not larger than 2 millimeters.
    • N1a: cancer has spread to 1 to 3 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters.
    • N1b: cancer has spread to lymph nodes near the breastbone on the same side of the body as the primary tumor, and the cancer is larger than 0.2 millimeters and is found by sentinel lymph node biopsy. Cancer is not found in the axillary lymph nodes.
    • N1c: cancer has spread to 1 to 3 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters. Cancer is also found by sentinel lymph node biopsy in the lymph nodes near the breastbone on the same side of the body as the primary tumor.
  • N2: Cancer is described as one of the following:
    • N2a: cancer has spread to 4 to 9 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters.
    • N2b: cancer has spread to lymph nodes near the breastbone and the cancer is found by imaging tests. Cancer is not found in the axillary lymph nodes by sentinel lymph node biopsy or lymph node dissection.
  • N3: Cancer is described as one of the following:
    • N3a: cancer has spread to 10 or more axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters, or cancer has spread to lymph nodes below the collarbone.
    • N3b: cancer has spread to 1 to 9 axillary lymph nodes and the cancer in at least one of the lymph nodes is larger than 2 millimeters. Cancer has also spread to lymph nodes near the breastbone and the cancer is found by imaging tests;

      or

      cancer has spread to 4 to 9 axillary lymph nodes and cancer in at least one of the lymph nodes is larger than 2 millimeters. Cancer has also spread to lymph nodes near the breastbone on the same side of the body as the primary tumor, and the cancer is larger than 0.2 millimeters and is found by sentinel lymph node biopsy.

    • N3c: cancer has spread to lymph nodes above the collarbone on the same side of the body as the primary tumor.

When the lymph nodes are checked using mammography or ultrasound, it is called clinical staging. The clinical staging of lymph nodes is not described here.

Metastasis (M). The spread of cancer to other parts of the body.

  • M0: There is no sign that cancer has spread to other parts of the body.
  • M1: Cancer has spread to other parts of the body, most often the bones, lungs, liver, or brain. If cancer has spread to distant lymph nodes, the cancer in the lymph nodes is larger than 0.2 millimeters. The cancer is called metastatic breast cancer.

The grading system is used to describe how quickly a breast tumor is likely to grow and spread.

The grading system describes a tumor based on how abnormal the cancer cells and tissue look under a microscope and how quickly the cancer cells are likely to grow and spread. Low-grade cancer cells look more like normal cells and tend to grow and spread more slowly than high-grade cancer cells. To describe how abnormal the cancer cells and tissue are, the pathologist will assess the following three features:

  • How much of the tumor tissue has normal breast ducts.
  • The size and shape of the nuclei in the tumor cells.
  • How many dividing cells are present, which is a measure of how fast the tumor cells are growing and dividing.

For each feature, the pathologist assigns a score of 1 to 3; a score of “1” means the cells and tumor tissue look the most like normal cells and tissue, and a score of “3” means the cells and tissue look the most abnormal. The scores for each feature are added together to get a total score between 3 and 9.

Three grades are possible:

  • Total score of 3 to 5: G1 (Low grade or well differentiated).
  • Total score of 6 to 7: G2 (Intermediate grade or moderately differentiated).
  • Total score of 8 to 9: G3 (High grade or poorly differentiated).

Biomarker testing is used to find out whether breast cancer cells have certain receptors.

Healthy breast cells, and some breast cancer cells, have receptors (biomarkers) that attach to the hormones estrogen and progesterone. These hormones are needed for healthy cells, and some breast cancer cells, to grow and divide. To check for these biomarkers, samples of tissue containing breast cancer cells are removed during a biopsy or surgery. The samples are tested in a laboratory to see whether the breast cancer cells have estrogen or progesterone receptors.

Another type of receptor (biomarker) that is found on the surface of all breast cancer cells is called HER2. HER2 receptors are needed for the breast cancer cells to grow and divide.

For breast cancer, biomarker testing includes the following:

  • Estrogen receptor (ER). If the breast cancer cells have estrogen receptors, the cancer cells are called ER positive (ER+). If the breast cancer cells do not have estrogen receptors, the cancer cells are called ER negative (ER-).
  • Progesterone receptor (PR). If the breast cancer cells have progesterone receptors, the cancer cells are called PR positive (PR+). If the breast cancer cells do not have progesterone receptors, the cancer cells are called PR negative (PR-).
  • Human epidermal growth factor type 2 receptor (HER2/neu or HER2). If the breast cancer cells have larger than normal amounts of HER2 receptors on their surface, the cancer cells are called HER2 positive (HER2+). If the breast cancer cells have a normal amount of HER2 on their surface, the cancer cells are called HER2 negative (HER2-). HER2+ breast cancer is more likely to grow and divide faster than HER2- breast cancer.

Sometimes the breast cancer cells will be described as triple negative or triple positive.

  • Triple negative. If the breast cancer cells do not have estrogen receptors, progesterone receptors, or a larger than normal amount of HER2 receptors, the cancer cells are called triple negative.
  • Triple positive. If the breast cancer cells do have estrogen receptors, progesterone receptors, and a larger than normal amount of HER2 receptors, the cancer cells are called triple positive.

It is important to know the estrogen receptor, progesterone receptor, and HER2 receptor status to choose the best treatment. There are drugs that can stop the receptors from attaching to the hormones estrogen and progesterone and stop the cancer from growing. Other drugs may be used to block the HER2 receptors on the surface of the breast cancer cells and stop the cancer from growing.

The TNM system, the grading system, and biomarker status are combined to find out the breast cancer stage.

Here are 3 examples that combine the TNM system, the grading system, and the biomarker status to find out the Pathological Prognostic breast cancer stage for a woman whose first treatment was surgery:

If the tumor size is 30 millimeters (T2), has not spread to nearby lymph nodes (N0), has not spread to distant parts of the body (M0), and is:

  • Grade 1
  • HER2+
  • ER-
  • PR-

The cancer is stage IIA.

If the tumor size is 53 millimeters (T3), has spread to 4 to 9 axillary lymph nodes (N2), has not spread to other parts of the body (M0), and is:

  • Grade 2
  • HER2+
  • ER+
  • PR-

The tumor is stage IIIA.

If the tumor size is 65 millimeters (T3), has spread to 3 axillary lymph nodes (N1a), has spread to the lungs (M1), and is:

  • Grade 1
  • HER2+
  • ER-
  • PR-

The cancer is stage IV (metastatic breast cancer).

Talk to your doctor to find out what your breast cancer stage is and how it is used to plan the best treatment for you.

After surgery, your doctor will receive a pathology report that describes the size and location of the primary tumor, the spread of cancer to nearby lymph nodes, tumor grade, and whether certain biomarkers are present. The pathology report and other test results are used to determine your breast cancer stage.

You are likely to have many questions. Ask your doctor to explain how staging is used to decide the best options to treat your cancer and whether there are clinical trials that might be right for you.

Treatment Option Overview

Key Points

  • Treatment options for pregnant women depend on the stage of the disease and the trimester of the pregnancy.
  • Three types of standard treatment are used:
    • Surgery
    • Radiation therapy
    • Chemotherapy
  • Ending the pregnancy does not seem to improve the mother’s chance of survival.
  • Treatment for breast cancer may cause side effects.

Treatment options for pregnant women depend on the stage of the disease and the trimester of the pregnancy.

Three types of standard treatment are used:

Surgery

Most pregnant women with breast cancer have surgery to remove the breast. Some of the lymph nodes under the arm may be removed so they can be checked under a microscope by a pathologist for signs of cancer.

Types of surgery to remove the cancer include:

  • Modified radical mastectomy: Surgery to remove the whole breast that has cancer. This may include removal of the nipple, areola (the dark-colored skin around the nipple), and skin over the breast. Most of the lymph nodes under the arm are also removed.
    EnlargeModified radical mastectomy; the drawing on the left shows the removal of the whole breast, including the lymph nodes under the arm. The drawing on the right shows a cross-section of the breast, including the fatty tissue and chest wall (ribs and muscle). A tumor in the breast is also shown.
    Modified radical mastectomy. The whole breast and most of the lymph nodes under the arm are removed.
  • Breast-conserving surgery: Surgery to remove the cancer and some normal tissue around it, but not the breast itself. Part of the chest wall lining may also be removed if the cancer is near it. This type of surgery may also be called lumpectomy, partial mastectomy, segmental mastectomy, quadrantectomy, or breast-sparing surgery.
    EnlargeLumpectomy; the drawing on the left shows removal of the tumor and some of the normal tissue around it. The drawing on the right shows removal of some of the lymph nodes under the arm and removal of the tumor and part of the chest wall lining near the tumor. Also shown is fatty tissue in the breast.
    Lumpectomy. The tumor and some normal tissue around it are removed, but not the breast itself. Some lymph nodes under the arm may also be removed. If the cancer is near the chest wall, part of the chest wall lining may be removed as well.

After the doctor removes all of the cancer that can be seen at the time of surgery, some patients may be given chemotherapy or radiation therapy after surgery to kill any cancer cells that are left. For pregnant women with early-stage breast cancer, radiation therapy and hormone therapy are given after the baby is born. Treatment given after 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.

External radiation therapy may be given to pregnant women with early stage (stage I or II) breast cancer after the baby is born. Women with late stage (stage III or IV) breast cancer may be given external radiation therapy after the first 3 months of pregnancy or, if possible, radiation therapy is delayed until after the baby is born.

Chemotherapy

Chemotherapy is a cancer treatment that uses drugs to stop the growth of cancer cells, either by killing the cells or by stopping the cells from dividing. When chemotherapy is taken by mouth or injected into a vein or muscle, the drugs enter the bloodstream and can reach cancer cells throughout the body (systemic chemotherapy).

Chemotherapy is usually not given during the first 3 months of pregnancy. Chemotherapy given after this time does not usually harm the fetus but may cause early labor or low birth weight.

See Drugs Approved for Breast Cancer for more information.

Ending the pregnancy does not seem to improve the mother’s chance of survival.

Because ending the pregnancy is not likely to improve the mother’s chance of survival, it is not usually a treatment option.

Treatment for breast cancer may cause side effects.

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

Treatment of Early Stage Breast Cancer During Pregnancy

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

Pregnant women with early-stage breast cancer (stage I and stage II) are usually treated in the same way as patients who are not pregnant, with some changes to protect the fetus. Treatment may include the following:

Hormone therapy and trastuzumab should not be given during pregnancy.

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 Late-Stage Breast Cancer During Pregnancy

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

There is no standard treatment for patients with late-stage breast cancer (stage III or stage IV) during pregnancy. Treatment may include the following:

Radiation therapy and chemotherapy should not be given during the first 3 months of pregnancy.

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.

Special Issues About Breast Cancer During Pregnancy

Key Points

  • Lactation (breast milk production) and breast-feeding should be stopped if surgery or chemotherapy is planned.
  • Breast cancer does not appear to harm the fetus.
  • Pregnancy does not seem to affect the survival of women who have had breast cancer in the past.

Lactation (breast milk production) and breast-feeding should be stopped if surgery or chemotherapy is planned.

If surgery is planned, breast-feeding should be stopped to reduce blood flow in the breasts and make them smaller. Many chemotherapy drugs, especially cyclophosphamide and methotrexate, may occur in high levels in breast milk and may harm the nursing baby. Women receiving chemotherapy should not breast-feed.

Stopping lactation does not improve the mother’s prognosis.

Breast cancer does not appear to harm the fetus.

Breast cancer cells do not seem to pass from the mother to the fetus.

Pregnancy does not seem to affect the survival of women who have had breast cancer in the past.

For women who have had breast cancer, pregnancy does not seem to affect their survival. However, some doctors recommend that a woman wait 2 years after treatment for breast cancer before trying to have a baby, so that any early return of the cancer would be detected. This may affect a woman’s decision to become pregnant.

To Learn More About Breast Cancer During Pregnancy

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 breast cancer during pregnancy. 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 Breast Cancer Treatment During Pregnancy. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/breast/patient/pregnancy-breast-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389161]

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.

Breast Cancer Treatment During Pregnancy (PDQ®)–Health Professional Version

Breast Cancer Treatment During Pregnancy (PDQ®)–Health Professional Version

General Information About Breast Cancer Treatment During Pregnancy

Incidence

Breast cancer is the most common cancer in pregnant and postpartum women and occurs in about 1 in 3,000 pregnant women. The average patient is between the ages of 32 years and 38 years. Because many women are choosing to delay childbearing, it is likely that the incidence of breast cancer during pregnancy will increase.

Anatomy

EnlargeIllustration of the female breast anatomy. On the left, a front view shows lymph nodes inside the breast going from the breast to the armpit. On the right, a cross-section shows the chest wall, ribs, fatty tissue, lobes, ducts, and lobules. Also shown in both panels are the muscle, nipple, and areola.
The female breast contains lobes, lobules, and ducts that produce and transport milk to the nipple. Fatty tissue gives the breast its shape, while muscles and the chest wall provide support. The lymphatic system, including lymph nodes, filter lymph and store white blood cells that help fight infection and disease.

Diagnostic Evaluation

The natural tenderness and engorgement of the breasts of pregnant and lactating women may hinder detection of discrete masses and early diagnosis of breast cancer. Delays in diagnosis are common, with an average reported delay of 5 to 15 months from the onset of symptoms.[14] Because of this delay, cancers are typically detected at a later stage than in a nonpregnant, age-matched population.[5]

The following tests and procedures may be used to diagnose breast cancer during pregnancy:

  • Breast self-examination.
  • Clinical breast examination.
  • Ultrasound.
  • Biopsy and hormone receptor assays.
  • Mammography.

To detect breast cancer, pregnant and lactating women should consider practicing self-examination and undergo a clinical breast examination as part of the routine prenatal examination by a doctor. If an abnormality is found, diagnostic approaches such as ultrasound and mammography may be used. With proper shielding, mammography poses little risk of radiation exposure to the fetus.[6] However, mammograms are only used to evaluate dominant masses and to locate occult carcinomas in the presence of other suspicious physical findings.[6]

Because at least 25% of mammograms in pregnancy may be negative in the presence of cancer, a biopsy is essential for the diagnosis of any palpable mass. Diagnosis may be safely accomplished with a fine-needle aspiration, core biopsy, or excisional biopsy under local anesthesia. To avoid a false-positive diagnosis as a result of misinterpretation of pregnancy-related changes, the pathologist should be advised that the patient is pregnant.[7]

Breast cancer pathology is similar in age-matched pregnant and nonpregnant women. Hormone receptor assays using a competitive binding assay are usually negative in pregnant patients with breast cancer, but this may be the result of receptor binding by high serum estrogen levels associated with the pregnancy. Enzyme immunocytochemical receptor assays are more sensitive than competitive binding assays. A study that used both assay methods indicated similar receptor positivity between pregnant and nonpregnant women with breast cancer.[8] The study concluded that increased estrogen levels during pregnancy could result in a higher incidence of receptor positivity detected with immunohistochemistry than is detected by radiolabeled ligand-binding assay because of competitive inhibition by high levels of endogenous estrogen.

For more information, see the Diagnosis section in Breast Cancer Treatment.

Prognosis

The overall survival of pregnant women with breast cancer may be worse than that of nonpregnant women at all stages.[6] However, this discrepancy may be primarily the result of delayed diagnosis.[9] Termination of pregnancy has not shown any beneficial effect on breast cancer outcome and is not usually considered as a therapeutic option.[1,2,4,10,11]

References
  1. Hoover HC: Breast cancer during pregnancy and lactation. Surg Clin North Am 70 (5): 1151-63, 1990. [PUBMED Abstract]
  2. Gwyn K, Theriault R: Breast cancer during pregnancy. Oncology (Huntingt) 15 (1): 39-46; discussion 46, 49-51, 2001. [PUBMED Abstract]
  3. Moore HC, Foster RS: Breast cancer and pregnancy. Semin Oncol 27 (6): 646-53, 2000. [PUBMED Abstract]
  4. Rugo HS: Management of breast cancer diagnosed during pregnancy. Curr Treat Options Oncol 4 (2): 165-73, 2003. [PUBMED Abstract]
  5. Clark RM, Chua T: Breast cancer and pregnancy: the ultimate challenge. Clin Oncol (R Coll Radiol) 1 (1): 11-8, 1989. [PUBMED Abstract]
  6. Yang WT, Dryden MJ, Gwyn K, et al.: Imaging of breast cancer diagnosed and treated with chemotherapy during pregnancy. Radiology 239 (1): 52-60, 2006. [PUBMED Abstract]
  7. Middleton LP, Amin M, Gwyn K, et al.: Breast carcinoma in pregnant women: assessment of clinicopathologic and immunohistochemical features. Cancer 98 (5): 1055-60, 2003. [PUBMED Abstract]
  8. Elledge RM, Ciocca DR, Langone G, et al.: Estrogen receptor, progesterone receptor, and HER-2/neu protein in breast cancers from pregnant patients. Cancer 71 (8): 2499-506, 1993. [PUBMED Abstract]
  9. Petrek JA, Dukoff R, Rogatko A: Prognosis of pregnancy-associated breast cancer. Cancer 67 (4): 869-72, 1991. [PUBMED Abstract]
  10. Barnavon Y, Wallack MK: Management of the pregnant patient with carcinoma of the breast. Surg Gynecol Obstet 171 (4): 347-52, 1990. [PUBMED Abstract]
  11. Gallenberg MM, Loprinzi CL: Breast cancer and pregnancy. Semin Oncol 16 (5): 369-76, 1989. [PUBMED Abstract]

Stage Information for Breast Cancer Treatment and Pregnancy

Staging Evaluation

The following procedures are used to determine the extent of the cancer:

  • Chest x-ray.
  • Bone scan.
  • Ultrasound of the liver.
  • Magnetic resonance imaging (MRI) of the brain.

Procedures used for determining the stage of breast cancer are modified for pregnant women to avoid radiation exposure to the fetus. Nuclear scans cause fetal radiation exposure.[1] If such scans are essential for evaluation, hydration and Foley catheter drainage of the bladder can be used to prevent retention of radioactivity. Timing of the exposure to radiation relative to the gestational age of the fetus may be more critical than the actual dose of radiation delivered.[2] Radiation exposure during the first trimester (>0.1 Gy) may lead to congenital malformations, intellectual disability, and increased relative risk of carcinogenesis.

Chest x-rays with abdominal shielding are considered safe, but as with all radiological procedures, they are used only when essential for making treatment decisions.[1,3] A chest x-ray delivers 0.00008 Gy.[4]

For the diagnosis of bone metastases, a bone scan is preferable to a skeletal series because the bone scan delivers a smaller amount of radiation and is more sensitive. A bone scan delivers 0.001 Gy.[5]

Evaluation of the liver can be performed with ultrasound, and brain metastases can be diagnosed with an MRI scan. Data on MRI during pregnancy are not available, but gadolinium crosses the placenta and is associated with fetal abnormalities in rats.[5]

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

For more information, see the Stage Information for Breast Cancer section in Breast Cancer Treatment.

References
  1. Gwyn K, Theriault R: Breast cancer during pregnancy. Oncology (Huntingt) 15 (1): 39-46; discussion 46, 49-51, 2001. [PUBMED Abstract]
  2. Barnavon Y, Wallack MK: Management of the pregnant patient with carcinoma of the breast. Surg Gynecol Obstet 171 (4): 347-52, 1990. [PUBMED Abstract]
  3. Nicklas AH, Baker ME: Imaging strategies in the pregnant cancer patient. Semin Oncol 27 (6): 623-32, 2000. [PUBMED Abstract]
  4. Gallenberg MM, Loprinzi CL: Breast cancer and pregnancy. Semin Oncol 16 (5): 369-76, 1989. [PUBMED Abstract]
  5. Yang WT, Dryden MJ, Gwyn K, et al.: Imaging of breast cancer diagnosed and treated with chemotherapy during pregnancy. Radiology 239 (1): 52-60, 2006. [PUBMED Abstract]

Treatment of Early/Localized/Operable Breast Cancer During Pregnancy

Generally, pregnant women with stage I or stage II breast cancer are treated in the same way as nonpregnant patients, with some modifications to protect the fetus.

Treatment options for early/localized/operable breast cancer in pregnant women include:

  1. Surgery. Postpartum radiation therapy may also be given to women diagnosed with breast cancer late in pregnancy.
  2. Chemotherapy (after the first trimester).
  3. Endocrine therapy (after delivery).

The use of trastuzumab during pregnancy is contraindicated.

Surgery

Surgery is recommended as the primary treatment of breast cancer in pregnant women.

The data regarding safety of sentinel lymph node biopsy in pregnant patients are limited to several retrospective case series. One study examined sentinel lymph node biopsy in eight patients in the first trimester, nine patients in the second trimester, and eight patients in the third trimester. Technetium Tc 99m alone was used in 16 patients, methylene blue dye alone was used in seven patients, and two patients had unknown mapping methods. All 25 patients had live-born infants, of whom 24 were healthy, and one had a cleft palate (in the setting of other maternal risk factors).[1]

Because radiation in therapeutic doses may expose the fetus to potentially harmful scatter radiation,[2] modified radical mastectomy is the treatment of choice if the breast cancer was diagnosed early in pregnancy. If diagnosed late in pregnancy, breast-conserving surgery with postpartum radiation therapy has been used for breast preservation.[3] An analysis has been performed that helps to predict the risk of waiting to have radiation.[4,5]

Chemotherapy

Data suggest that it is safe to administer certain chemotherapeutic drugs after the first trimester, with most pregnancies resulting in live births with low rates of morbidity in the newborns.

Anthracycline-based chemotherapy (doxorubicin plus cyclophosphamide or fluorouracil, doxorubicin, and cyclophosphamide [FAC]) appears to be safe to administer during the second and/or third trimester on the basis of limited prospective data.[68] Safety data on the use of taxanes during pregnancy are limited.

Evidence (use of chemotherapy during the second and/or third trimester of pregnancy):

  1. A multicenter case-control study compared pediatric outcomes of 129 children whose mothers had breast cancer with matched children of women without cancer. In the pregnancy study group, 96 children (74.4%) were exposed to chemotherapy, 11 (8.5%) to radiation therapy, 13 (10.1%) to surgery alone, 2 (1.7%) to other drug treatments, and 14 (10.9%) to no treatment.[9]
    • The study showed that there was no significant difference in birth weight below the 10th percentile (22% in the breast cancer treatment‒exposed group vs. 15.2% in the control group, P = .16) or in cognitive development based on the Bayley score (P = .08). The gestational age at birth was correlated with cognitive outcome in the two study groups.
    • Evaluation of cardiac function among 47 children, who were age 36 months in the study group, showed normal cardiac findings.
  2. In a prospective single-arm study, 57 pregnant patients with breast cancer were treated with FAC in the adjuvant or neoadjuvant setting.[6]
    • Survey data collected when the children were aged 2 months to 157 months revealed that no stillbirths, miscarriages, or perinatal deaths occurred.
    • One child born vaginally at a gestational age of 38 weeks had a subarachnoid hemorrhage on day 2 postpartum, one child had Down syndrome, and two children had congenital anomalies (club foot and bilateral ureteral reflux).
  3. The findings of the prospective single-arm study above were consistent with other smaller retrospective series of anthracycline-based chemotherapy.[7,8]
  4. A systematic review studied 40 case reports of taxane administration during the second or third trimesters of pregnancy.[10]
    • Minimal maternal, fetal, or neonatal toxicity was observed.

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.[11,12] 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.[1113] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[1416] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[17] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[18]

Endocrine Therapy

Endocrine therapy is generally avoided until after delivery. Case reports and a literature review of tamoxifen during pregnancy show that tamoxifen administration during pregnancy is associated with vaginal bleeding, miscarriage, congenital abnormalities such as Goldenhar syndrome, and fetal death.[1921] Breastfeeding is also not recommended concurrently with endocrine therapy.[22]

Targeted Therapy

The use of trastuzumab during pregnancy is contraindicated based on results of a systematic review of 17 studies (18 pregnancies, 19 newborns).[23] Of the fetal complications noted, occurrence of oligohydramnios/anhydramnios was the most common (61.1%) adverse event. Of the pregnancies exposed to trastuzumab during the second or third trimester, 73.3% of the pregnancies were complicated with oligohydramnios/anhydramnios. Of the pregnancies exposed to trastuzumab exclusively during the first trimester, 0% (P = .043) of the pregnancies were complicated with oligohydramnios/anhydramnios. The mean gestational age at delivery was 33.8 weeks, and the mean weight of newborns at delivery was 2,261 grams or 4.984 pounds. In 52.6% of cases, a healthy neonate was born. At the long-term evaluation, all children who were without complications at birth were healthy, with a median follow-up of 9 months, and four of nine children with complications at birth had died within an interval ranging from birth to 5.25 months. All children exposed to trastuzumab in utero exclusively in the first trimester were completely healthy at birth. The data suggest that for women who become pregnant during trastuzumab administration and wish to continue pregnancy, trastuzumab should be stopped and pregnancy would be allowed to continue.

References
  1. Gropper AB, Calvillo KZ, Dominici L, et al.: Sentinel lymph node biopsy in pregnant women with breast cancer. Ann Surg Oncol 21 (8): 2506-11, 2014. [PUBMED Abstract]
  2. Kal HB, Struikmans H: Radiotherapy during pregnancy: fact and fiction. Lancet Oncol 6 (5): 328-33, 2005. [PUBMED Abstract]
  3. Gwyn K, Theriault R: Breast cancer during pregnancy. Oncology (Huntingt) 15 (1): 39-46; discussion 46, 49-51, 2001. [PUBMED Abstract]
  4. Nettleton J, Long J, Kuban D, et al.: Breast cancer during pregnancy: quantifying the risk of treatment delay. Obstet Gynecol 87 (3): 414-8, 1996. [PUBMED Abstract]
  5. Kuerer HM, Gwyn K, Ames FC, et al.: Conservative surgery and chemotherapy for breast carcinoma during pregnancy. Surgery 131 (1): 108-10, 2002. [PUBMED Abstract]
  6. Hahn KM, Johnson PH, Gordon N, et al.: Treatment of pregnant breast cancer patients and outcomes of children exposed to chemotherapy in utero. Cancer 107 (6): 1219-26, 2006. [PUBMED Abstract]
  7. Turchi JJ, Villasis C: Anthracyclines in the treatment of malignancy in pregnancy. Cancer 61 (3): 435-40, 1988. [PUBMED Abstract]
  8. Zemlickis D, Lishner M, Degendorfer P, et al.: Fetal outcome after in utero exposure to cancer chemotherapy. Arch Intern Med 152 (3): 573-6, 1992. [PUBMED Abstract]
  9. Amant F, Vandenbroucke T, Verheecke M, et al.: Pediatric Outcome after Maternal Cancer Diagnosed during Pregnancy. N Engl J Med 373 (19): 1824-34, 2015. [PUBMED Abstract]
  10. Mir O, Berveiller P, Goffinet F, et al.: Taxanes for breast cancer during pregnancy: a systematic review. Ann Oncol 21 (2): 425-6, 2010. [PUBMED Abstract]
  11. 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]
  12. Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
  13. 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]
  14. 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]
  15. 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]
  16. 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]
  17. 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]
  18. 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]
  19. Cullins SL, Pridjian G, Sutherland CM: Goldenhar’s syndrome associated with tamoxifen given to the mother during gestation. JAMA 271 (24): 1905-6, 1994 Jun 22-29. [PUBMED Abstract]
  20. Tewari K, Bonebrake RG, Asrat T, et al.: Ambiguous genitalia in infant exposed to tamoxifen in utero. Lancet 350 (9072): 183, 1997. [PUBMED Abstract]
  21. Isaacs RJ, Hunter W, Clark K: Tamoxifen as systemic treatment of advanced breast cancer during pregnancy–case report and literature review. Gynecol Oncol 80 (3): 405-8, 2001. [PUBMED Abstract]
  22. Helewa M, Lévesque P, Provencher D, et al.: Breast cancer, pregnancy, and breastfeeding. J Obstet Gynaecol Can 24 (2): 164-80; quiz 181-4, 2002. [PUBMED Abstract]
  23. Zagouri F, Sergentanis TN, Chrysikos D, et al.: Trastuzumab administration during pregnancy: a systematic review and meta-analysis. Breast Cancer Res Treat 137 (2): 349-57, 2013. [PUBMED Abstract]

Treatment of Advanced Breast Cancer During Pregnancy

There is no standard treatment for patients with advanced (stage III or stage IV) breast cancer during pregnancy. Most studies show a 5-year survival rate of 10% in pregnant patients with stage III or IV disease.

First-trimester radiation therapy should be avoided. Chemotherapy may be given after the first trimester as discussed in the section on Treatment of Early/Localized/Operable Breast Cancer During Pregnancy.

Because the mother’s life span may be limited, and there is a risk of fetal damage with treatment during the first trimester,[1,2] issues regarding continuation of the pregnancy should be discussed with the patient and her family. Therapeutic abortion does not improve prognosis.[15]

References
  1. Hoover HC: Breast cancer during pregnancy and lactation. Surg Clin North Am 70 (5): 1151-63, 1990. [PUBMED Abstract]
  2. Rugo HS: Management of breast cancer diagnosed during pregnancy. Curr Treat Options Oncol 4 (2): 165-73, 2003. [PUBMED Abstract]
  3. Gwyn K, Theriault R: Breast cancer during pregnancy. Oncology (Huntingt) 15 (1): 39-46; discussion 46, 49-51, 2001. [PUBMED Abstract]
  4. Clark RM, Chua T: Breast cancer and pregnancy: the ultimate challenge. Clin Oncol (R Coll Radiol) 1 (1): 11-8, 1989. [PUBMED Abstract]
  5. Barnavon Y, Wallack MK: Management of the pregnant patient with carcinoma of the breast. Surg Gynecol Obstet 171 (4): 347-52, 1990. [PUBMED Abstract]

Special Considerations for Pregnancy and Breast Cancer

Lactation

Suppression of lactation does not improve prognosis. If surgery is planned, however, lactation is suppressed to decrease the size and vascularity of the breasts. If chemotherapy is to be given, lactation is also suppressed because many antineoplastic agents (i.e., cyclophosphamide and methotrexate), when given systemically, may occur in high levels in breast milk and would affect the nursing baby. Women receiving chemotherapy should not breastfeed.[1]

Fetal Consequences of Maternal Breast Cancer

No damaging effects on the fetus from maternal breast cancer have been demonstrated,[2] and there are no reported cases of maternal-fetal transfer of breast cancer cells.

Pregnancy in Patients With a History of Breast Cancer

Based on limited retrospective data, pregnancy does not appear to compromise the survival of women with a previous history of breast cancer, and no deleterious effects have been demonstrated in the fetus.[311] Some physicians recommend that patients wait 2 years after diagnosis before attempting to conceive. This allows early recurrence to become manifest, which may influence the decision to become a parent.

Little is known about pregnancy after bone marrow transplant and high-dose chemotherapy with or without total-body irradiation. In one report of pregnancies after bone marrow transplant for hematologic disorders, a 25% incidence of preterm labor and low birth weight for gestational-age infants was noted.[12]

References
  1. Helewa M, Lévesque P, Provencher D, et al.: Breast cancer, pregnancy, and breastfeeding. J Obstet Gynaecol Can 24 (2): 164-80; quiz 181-4, 2002. [PUBMED Abstract]
  2. Amant F, Vandenbroucke T, Verheecke M, et al.: Pediatric Outcome after Maternal Cancer Diagnosed during Pregnancy. N Engl J Med 373 (19): 1824-34, 2015. [PUBMED Abstract]
  3. Clark RM, Chua T: Breast cancer and pregnancy: the ultimate challenge. Clin Oncol (R Coll Radiol) 1 (1): 11-8, 1989. [PUBMED Abstract]
  4. Harvey JC, Rosen PP, Ashikari R, et al.: The effect of pregnancy on the prognosis of carcinoma of the breast following radical mastectomy. Surg Gynecol Obstet 153 (5): 723-5, 1981. [PUBMED Abstract]
  5. Petrek JA: Pregnancy safety after breast cancer. Cancer 74 (1 Suppl): 528-31, 1994. [PUBMED Abstract]
  6. von Schoultz E, Johansson H, Wilking N, et al.: Influence of prior and subsequent pregnancy on breast cancer prognosis. J Clin Oncol 13 (2): 430-4, 1995. [PUBMED Abstract]
  7. Kroman N, Mouridsen HT: Prognostic influence of pregnancy before, around, and after diagnosis of breast cancer. Breast 12 (6): 516-21, 2003. [PUBMED Abstract]
  8. Malamos NA, Stathopoulos GP, Keramopoulos A, et al.: Pregnancy and offspring after the appearance of breast cancer. Oncology 53 (6): 471-5, 1996 Nov-Dec. [PUBMED Abstract]
  9. Gelber S, Coates AS, Goldhirsch A, et al.: Effect of pregnancy on overall survival after the diagnosis of early-stage breast cancer. J Clin Oncol 19 (6): 1671-5, 2001. [PUBMED Abstract]
  10. Gwyn K, Theriault R: Breast cancer during pregnancy. Oncology (Huntingt) 15 (1): 39-46; discussion 46, 49-51, 2001. [PUBMED Abstract]
  11. Rugo HS: Management of breast cancer diagnosed during pregnancy. Curr Treat Options Oncol 4 (2): 165-73, 2003. [PUBMED Abstract]
  12. Sanders JE, Hawley J, Levy W, et al.: Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation. Blood 87 (7): 3045-52, 1996. [PUBMED Abstract]

Latest Updates to This Summary (12/04/2024)

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.

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 breast cancer during pregnancy. 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 Breast Cancer Treatment During Pregnancy are:

  • Fumiko Chino, MD (MD Anderson Cancer Center)
  • Tarek Hijal, MD (McGill University Health Centre)
  • Joseph L. Pater, MD (NCIC-Clinical Trials Group)
  • Carol Tweed, MD (Maryland Oncology Hematology)

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 Breast Cancer Treatment During Pregnancy. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/breast/hp/pregnancy-breast-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389427]

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.

Breast Cancer Screening (PDQ®)–Patient Version

Breast Cancer Screening (PDQ®)–Patient Version

What Is Screening?

Screening is looking for signs of disease, such as breast cancer, before a person has symptoms. The goal of screening tests is to find cancer at an early stage when it can be treated and may be cured. Sometimes a screening test finds cancer that is very small or very slow growing. These cancers are unlikely to cause death or illness during a person’s lifetime.

Scientists are trying to better understand which people are more likely to get certain types of cancer. For example, they look at a person’s age, their family history, and certain exposures during their lifetime. This information helps doctors recommend who should be screened for cancer, which screening tests should be used, and how often the tests should be done.

It is important to remember that your doctor does not necessarily think you have cancer if he or she suggests a screening test. Screening tests are done when you have no cancer symptoms. Women who have a strong family history or a personal history of cancer or other risk factors may also be offered genetic testing.

If a screening test result is abnormal, you may need to have more tests done to find out if you have cancer. These are called diagnostic tests, rather than screening tests.

To learn more about cancer screening, visit Cancer Screening Overview.

General Information About Breast Cancer

Key Points

  • Breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast.
  • Breast cancer is the second leading cause of death from cancer in American women.
  • Different factors increase or decrease the risk of getting breast cancer.

Breast cancer is a disease in which malignant (cancer) cells form in the tissues of the breast.

The breast is made up of lobes and ducts. Each breast has 15 to 20 sections called lobes, which have many smaller sections called lobules. Lobules end in dozens of tiny bulbs that can produce milk. The lobes, lobules, and bulbs are linked by thin tubes called ducts.

EnlargeIllustration of the female breast anatomy. On the left, a front view shows lymph nodes inside the breast going from the breast to the armpit. On the right, a cross-section shows the chest wall, ribs, fatty tissue, lobes, ducts, and lobules. Also shown in both panels are the muscle, nipple, and areola.
The female breast contains lobes, lobules, and ducts that produce and transport milk to the nipple. Fatty tissue gives the breast its shape, while muscles and the chest wall provide support. The lymphatic system, including lymph nodes, filter lymph and store white blood cells that help fight infection and disease.

Each breast also has blood vessels and lymph vessels. The lymph vessels carry an almost colorless, watery fluid called lymph. Lymph vessels carry lymph between lymph nodes. Lymph nodes are small, bean-shaped structures that filter lymph and store white blood cells that help fight infection and disease. Groups of lymph nodes are found near the breast in the axilla (under the arm), above the collarbone, and in the chest.

Other PDQ summaries containing information related to breast cancer include:

Breast cancer is the second leading cause of death from cancer in American women.

Women in the United States get breast cancer more than any other type of cancer except for skin cancer.

Breast cancer is more likely to occur as a woman ages. It occurs more often in White women than in Black women, but Black women die from breast cancer more often than White women. However, this difference could be due to factors, such as quality of the screening test, how long women wait to follow up after getting an abnormal test result, the quality of treatment, and the type of tumor.

Breast cancer rarely occurs in men. Because men with breast cancer usually have a lump that can be felt, screening tests are not likely to be helpful.

Different factors increase or decrease the risk of getting breast cancer.

Anything that increases your chance of getting a disease is called a risk factor. Anything that decreases your chance of getting a disease is called a protective factor. Talk to your doctor if you think you may be at risk for breast cancer.

To learn more about risk factors and protective factors for breast cancer, visit Breast Cancer Prevention.

Breast Cancer Screening

Key Points

  • Tests are used to screen for different types of cancer when a person does not have symptoms.
  • Mammography is the most common screening test for breast cancer.
  • Magnetic resonance imaging (MRI) may be used to screen women who have a high risk of breast cancer.
  • Whether a woman should be screened for breast cancer and the screening test to use depends on certain factors.
  • Other screening tests have been or are being studied in clinical trials.
    • Breast exam
    • Thermography
    • Tissue sampling
    • Ultrasound exam
  • Screening tests for breast cancer are being studied in clinical trials.

Tests are used to screen for different types of cancer when a person does not have symptoms.

Scientists study screening tests to find those with the fewest harms and most benefits. Cancer screening trials also are meant to show whether early detection (finding cancer before it causes symptoms) helps a person live longer or decreases a person’s chance of dying from the disease. For some types of cancer, the chance of recovery is better if the disease is found and treated at an early stage.

Mammography is the most common screening test for breast cancer.

A mammogram is a picture of the inside of the breast. Mammography may find tumors that are too small to feel. It may also find ductal carcinoma in situ (DCIS). In DCIS, abnormal cells line the breast duct, and in some women may become invasive breast cancer.

There are different types of mammograms:

  • Screen-film mammography (SFM) is an x-ray picture of the breast.
  • Digital mammography (DM) is a computer picture of the breast.
  • Digital breast tomosynthesis (DBT) uses x-rays to take a series of pictures of the inside of the breast from many different angles. A computer is used to make 3-D pictures of the breast from these x-rays.
  • Synthetic 2-dimensional mammography (S2D) uses x-rays to take pictures of the inside of the breast, usually from two different angles. A computer or x-ray film is used to make 2-D pictures of the breast.

Digital breast tomosynthesis (DBT) was approved by the U.S. Food and Drug Administration (FDA) in 2018 and is now used in 3 out of 4 facilities. One recent study found that synthetic 2-dimensional mammography (S2D) combined with DBT improved tumor detection rates and lowered mammogram callbacks, radiation dose, and overall costs. More studies are being done to compare different types of breast cancer screening.

Mammography is less likely to find breast tumors in women with dense breast tissue. Because both tumors and dense breast tissue appear white on a mammogram, it can be harder to find a tumor when there is dense breast tissue. Younger women are more likely to have dense breast tissue. To learn more, visit Dense Breasts: Answers to Commonly Asked Questions.

EnlargeDrawing of a woman standing with her left breast pressed between two plates of a mammography machine. Behind her, a health professional uses an X-ray machine to take pictures of the breast. An inset shows the X-ray film image with an arrow pointed at abnormal tissue.
Mammography is an imaging test used to screen for and diagnose breast cancer. It can detect abnormal breast tissue, including cancer, sometimes before symptoms appear.

Many factors affect whether mammography is able to detect (find) breast cancer:

  • The age and weight of the patient.
  • The size and type of tumor.
  • Where the tumor has formed in the breast.
  • How sensitive the breast tissue is to hormones.
  • How dense the breast tissue is.
  • The timing of the mammography within the woman’s menstrual cycle.
  • The quality of the mammogram picture.
  • The skill of the radiologist in reading the mammogram.

Women aged 50 to 69 years who have screening mammograms have a lower chance of dying from breast cancer than women who do not have screening mammograms.

Fewer women are dying of breast cancer in the United States, but it is not known whether the lower risk of dying is because the cancer was found early by screening or whether the treatments were better.

Magnetic resonance imaging (MRI) may be used to screen women who have a high risk of breast cancer.

MRI is 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). MRI does not use any x-rays, and the woman is not exposed to radiation.

EnlargeMRI of the breast; drawing shows a person lying face down on a narrow, padded table with their arms above their head. The person’s breasts hang down into an opening in the table. The table slides into the MRI machine, which takes detailed pictures of the inside of the breast. An inset shows an MRI image of the insides of both breasts.
An MRI of the breast is a procedure that uses radio waves, a strong magnet, and a computer to create detailed pictures of the inside of the breast. A contrast dye may be injected into a vein (not shown) to make the breast tissues easier to see on the MRI pictures. An MRI may be used with other breast imaging tests to detect breast cancer or other abnormal changes in the breast. It may also be used to screen for breast cancer in some people who have a high risk of the disease. Note: The inset shows an MRI image of the insides of both breasts. Credit for inset: The Cancer Imaging Archive (TCIA).

MRI may be used as a screening test for women who have a high risk of breast cancer. Factors that put women at high risk include:

An MRI is more likely than mammography to find a breast mass (lump) that is not cancer.

Women with dense breasts who have supplemental screening (for example, an MRI) show higher rates of breast cancer detection, but there is limited evidence about whether this leads to better health outcomes.

Whether a woman should be screened for breast cancer and the screening test to use depends on certain factors.

Women with risk factors for breast cancer, such as certain changes in the BRCA1 or BRCA2 gene or certain genetic syndromes, may be screened at a younger age and more often.

Women who have had radiation treatment to the chest, especially at a young age, may start routine breast cancer screening at an earlier age. The benefits and risks of mammograms and MRIs for these women have not been studied.

Breast cancer screening has not been shown to benefit the following women:

  • Older women who, if diagnosed with breast cancer through screening, will usually die of other causes. Screening mammograms for those aged 66 to 79 years may find cancer in a very small percentage of women, but most of these cancers are low risk.
  • In women with an average risk of developing breast cancer, screening mammography before age 40 has not shown any benefit.
  • In women who are not expected to live for a long time and have other diseases or conditions, finding and treating early-stage breast cancer may reduce their quality of life without helping them live longer.

Other screening tests have been or are being studied in clinical trials.

Studies have been done to find out if the following breast cancer screening tests are useful in finding breast cancer or helping women with breast cancer live longer.

Breast exam

A clinical breast exam is an exam of the breast by a doctor or other health professional. He or she will carefully feel the breasts and under the arms for lumps or anything else that seems unusual. It is not known if having clinical breast exams decreases the chance of dying from breast cancer.

Breast self-exams may be done by women or men to check their breasts for lumps or other changes. If you feel any lumps or notice any other changes in your breasts, talk to your doctor. Doing regular breast self-exams has not been shown to decrease the chance of dying from breast cancer.

Thermography

Thermography is a procedure in which a special camera that senses heat is used to record the temperature of the skin that covers the breasts. Tumors can cause temperature changes that may show up on the thermogram.

There have been no randomized clinical trials of thermography to find out how well it detects breast cancer or the harms of the procedure.

Tissue sampling

Breast tissue sampling is taking cells from breast tissue to check under a microscope. Breast tissue sampling as a screening test has not been shown to decrease the risk of dying from breast cancer.

Ultrasound exam

A procedure in which high-energy sound waves are bounced off internal tissues or organs and make echoes. The echoes form a picture of body tissues called a sonogram.

An ultrasound is sometimes used as an additional screening test for women who are at increased risk of developing breast cancer (such as women with dense breasts). It is not known if supplemental screening with ultrasound leads to better health outcomes.

Screening tests for breast cancer are being studied in clinical trials.

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.

Harms of Breast Cancer Screening

Key Points

  • Screening tests can have harms.
  • The harms of mammography include:
    • False-positive test results can occur.
    • False-positive results can lead to extra testing and cause anxiety.
    • False-negative test results can delay diagnosis and treatment.
    • Finding breast cancer may lead to breast cancer treatment and side effects, but it may not improve a woman’s health or help her live longer.
    • Mammography exposes the breast to low doses of radiation.
    • There may be pain or x-ray discomfort during a mammogram.
  • Talk to your doctor about your risk of breast cancer and your need for screening tests.

Screening tests can have harms.

Not all breast cancers will cause death or illness in a woman’s lifetime, so they may not need to be found or treated.

Decisions about screening tests can be difficult. Not all screening tests are helpful and most have harms. Before having any screening test, you may want to discuss the test with your doctor. It is important to know the harms of the test and whether it has been proven to reduce the risk of dying from cancer.

The harms of mammography include:

False-positive test results can occur.

Screening test results may appear to be abnormal even though no cancer is present. A false-positive test result (one that shows there is cancer when there really isn’t) is usually followed by more tests (such as biopsy), which also have risks.

When a breast biopsy result is abnormal, getting a second opinion from a different pathologist may confirm a correct breast cancer diagnosis.

Most abnormal test results turn out not to be cancer. False-positive results are more common in:

  • Younger women (under age 50).
  • Women who have had previous breast biopsies.
  • Women with a family history of breast cancer.
  • Women who take hormones for menopause.

False-positive results are more likely the first time screening mammography is done than with later screenings. For every 10 women who have a single mammogram, 1 will have a false-positive result. The chance of having a false-positive result goes up the more mammograms a woman has. Comparing a current mammogram with a past mammogram lowers the risk of a false-positive result.

The skill of the radiologist also can affect the chance of a false-positive result.

False-positive results can lead to extra testing and cause anxiety.

If a mammogram is abnormal, more tests may be done to diagnose cancer. Women can become anxious during the diagnostic testing. Even if it is a false-positive test and cancer is not diagnosed, the result can lead to anxiety anywhere from a few days to years later.

Several studies show that women who feel anxiety after false-positive test results are more likely to schedule regular breast screening exams in the future.

False-negative test results can delay diagnosis and treatment.

Screening test results may appear to be normal even though breast cancer is present. This is called a false-negative test result. A woman who has a false-negative test result may delay seeking medical care even if she has symptoms. About 1 in 5 cancers are missed by mammography.

The chance of a false-negative test result is more common in women who:

Finding breast cancer may lead to breast cancer treatment and side effects, but it may not improve a woman’s health or help her live longer.

Some breast cancers found only by screening mammography may never cause health problems or become life-threatening. Finding these cancers is called overdiagnosis. When these cancers are found, having treatment may cause serious side effects and may not lead to a longer, healthier life.

Mammography exposes the breast to low doses of radiation.

Being exposed to high doses of radiation is a risk factor for breast cancer. The radiation dose used with a mammogram is very low. Women who start getting mammograms at age 50 and continue getting them every 2 years have a lower risk of developing breast cancer from radiation exposure during a mammogram than women who start getting mammograms at age 40 and continue getting them every year. Women with large breasts or with breast implants may be exposed to slightly higher radiation doses during screening mammography.

There may be pain or x-ray discomfort during a mammogram.

During a mammogram, the breast is placed between two plates that are pressed together. Pressing the breast helps to get a better image of the breast. Some women have pain or discomfort during a mammogram. The amount of pain may also depend on:

  • The phase of the woman’s menstrual cycle.
  • The woman’s anxiety level.
  • How much pain the woman expected.

Talk to your doctor about your risk of breast cancer and your need for screening tests.

Talk to your doctor or other health care provider about your risk of breast cancer, whether a screening test is right for you, and the benefits and harms of the screening test. You should take part in the decision about whether you want to have a screening test, based on what is best for you. To learn more, visit Cancer Screening Overview.

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 breast cancer screening. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.

Reviewers and Updates

Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.

The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Screening and Prevention Editorial Board.

Clinical Trial Information

A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.

Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).

Permission to Use This Summary

PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”

The best way to cite this PDQ summary is:

PDQ® Screening and Prevention Editorial Board. PDQ Breast Cancer Screening. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/breast/patient/breast-screening-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389160]

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.