Soft Tissue Tumors /Soft tissue sarcomas are cancerous (malignant) tumors that originate in the soft tissues of your body. This illustration shows a soft tissue sarcoma of the thigh muscle just above the knee. Soft tissue sarcoma is a rare type of cancer that begins in the tissues that connect, support, and surround other body structures
WHO classification of Soft Tissue Tumors
The World Health Organization (WHO) classification of soft tissue tumors is the most widely used pathologic classification system for such disorders. The current revision, part of the 5th edition of the WHO series, was published in 2020 and is reflected in the article below 1.
epithelioid hemangioendothelioma features two newly recognized subtypes
anastomosing hemangioma
tufted angioma and Kaposiform hemangioendothelioma are classified together
pericytic (perivascular) tumors
myopericytoma, including myofibroma – new subtype: cellular myofibroma/myopericytoma
smooth muscle tumors
EBV-associated smooth muscle tumor
inflammatory leiomyosarcoma
skeletal muscle tumors
spindle cell/sclerosing rhabdomyosarcoma – three subtypes
tumors of uncertain differentiation
NTRK-rearranged soft tissue neoplasms
Other changes
melanotic schwannoma – now renamed to malignant melanotic nerve sheath tumor
dedifferentiated liposarcoma – the adverse prognostic impact of myogenic and rhabdomyoblastic differentiation as well of a high grade on the FNCLCC (French Federation of Cancer Centers Sarcoma Group) grading system
solitary fibrous tumor – extrapleural’ has been removed
Pleomorphic liposarcoma (PLS )is a rare and aggressive, fast-growing tumor and high-grade malignancy with high recurrence, poor prognosis, and its treatment is still highly controversial. A rare, fast-growing type of cancer that begins in fat cells. It usually forms in the deep soft tissues of the arms or legs, but it may also form in the abdomen or chest. Pleomorphic liposarcoma often recurs (comes back) after treatment and spreads to other parts of the body, including the lungs.
Pleomorphic liposarcoma is characterized as the growth of a progressively painless mass, and it is easily ignored until the mass is big enough or there are some other compressive manifestations. It is known that this neoplasm is usually aggressive, occurring in adulthood, and usually in the limbs. Pleomorphic liposarcoma can occur in the mediastinum, liver, orbit, paratesticular region, and also as a purely dermal tumor [rx,rx,rx–rx]PLS could occur in various organs, but the most common sites are proximal extremities, especially in the lower extremities, and in other uncommon sites including the retroperitoneum, the abdominal wall, the chest wall, the mesentery, the pelvic cavity, the spermatic cord, the mediastinum, the parietal pleura, the pericardium, the foot, the spine, the head, and neck region.[rx,rx–rx]
Grading
According to the guidelines of the ADASP, pleomorphic liposarcoma is considered high grade
French Federation of Cancer Centers System grading scheme for adult sarcomas
Tumor differentiation score = 3 for pleomorphic liposarcoma
Mitotic index
Score 1 0-9 mitoses per 10 hpf (0.1744 sq mm)
Score 2 10-19 mitoses per 10 hpf
Score 3 >19 mitoses per 10 hpf
Tumor cell necrosis
Score 0 No necrosis on any slide (one slide per 2 cm tumor diameter)
Score 1 <50% of tumor is necrotic on slides examined
Score 2 >50% of tumor is necrotic on slides examined
Final Grade (add the three scores above)
Grade 1 Sum of scores = 2 or 3
Grade 2 Sum of scores = 4 or 5
Grade 3 Sum of scores = 6 or more
Symptoms of Pleomorphic Liposarcoma
As previously mentioned, most patients are diagnosed with pleomorphic liposarcoma do not have any early symptoms and it can go unnoticed during the initial and primary stages of the disease until the tumor has grown to a large enough size to compress neighboring tissues and cause pain or decreased function.
It can sometimes be noticed as a deep-seated mass to touch.
Anorexia and abdominal distension, whereas the other patient had persistent pain in the left lower abdomen due to the compression from the pelvic tumor. However, these symptoms were ignored until the mass was big enough or there were some other serious manifestations.
Pleomorphic liposarcoma, as with all other cancers, can present with non-specific symptoms such as fevers, chills, fatigue, night sweats, anorexia, and weight loss.
If the tumor is retroperitoneal in location, it can present with specific symptoms in the abdomen, including abdominal pain, constipation, gastritis, or flank pain, swelling, and constipation, or the sensation of feeling full sooner than expected after eating.
The well-differentiated type tumor is less aggressive and tends to be a large painless mass found in deeper tissues and in the retroperitoneum.
Pleomorphic liposarcoma, round cell, and pleomorphic types tend to be in the arms and legs, whereas dedifferentiated tend to be in the retroperitoneum and often associated with the well-differentiated variety.
Specifically, pleomorphic liposarcoma is the least common subtype with a high rate of recurrence and poor outcomes.
Patients usually present with progressive dysphagia with weight loss
The symptoms of myxoid liposarcoma depend on where the tumor is on your body, but they include:
A new or growing lump beneath your skin, especially around or behind your knees or on your thighs
Pain or swelling
Weakness in an arm or leg that has the lump
Feeling full soon after you start eating
A new lump anywhere on your body, or an existing lump that grows persistently
Painful swelling or numbness in the area around your lump
Blood in your stool, or black or tarry stool (an indication of blood)
Blood in your vomit
Abdominal pain or cramping
Constipation
Poop that has blood or looks black or tarry
Cramping
Bloody vomit
Your belly gets larger
Tumors in the retroperitoneal, abdominal, and pelvic cavities were larger than those near the body surface such as the subcutaneous and intermuscular tissues.
Diagnosis of Pleomorphic Liposarcoma
Accessory examinations such as CT, ultrasound, and magnetic resonance imaging are critical for surgical methods by assessing the size of the tumor and the degree of tumor infiltration into the surrounding tissues. However, it is difficult to distinguish PLS/Pleomorphic liposarcoma from other liposarcoma and sarcoma using these accessory examinations. The definite diagnosis of PLS still depends on the pathological examination, which reveals highly meta topic cells with granular and/or foamy small vacuoles in cytoplasm, mono-/poly-nuclear giant cells with a deep
Microscopic (histologic) description
Well circumscribed but non-encapsulated with infiltrative borders
At least focal typical liposarcomatous areas
Pleomorphic cells cover > 65% of cut surface with MFH-like, round cell liposarcoma-like (without vascular network), spindle cell liposarcoma-like or epithelioid cells (Mod Pathol 1999;12:722)
Usually high grade with an enlarged round to bizarre nuclei
Tumor necrosis common
Median 25 mitotic figures / 10 HPF
May have neutrophils within giant cells, hemangiopericytoma foci, extra- and intracellular hyaline droplets
Epithelioid variant often confused with carcinoma
Imaging
CT scan, magnetic resonance imaging (MRI) – If you have symptoms of MRCLS, your doctor will use imaging scans such as CT and MRI to look at where the tumor is and how big it is. They will also check for signs that the tumor has spread to other parts of the body. Although none of these techniques is specific, CT scan and MRI can be more helpful in narrowing down the differential diagnosis as both modalities can detect the percentage of a lipomatous/Pleomorphic liposarcoma component of the tumor. Higher fat content is associated with benign lipoma, while less fat is consistent with atypical lipoma or sarcoma. A definitive diagnosis can only be achieved by tissue examination. And in the majority of the cases, complete resection of the tumor is needed for a correct diagnosis [rx].
Removing a sample of tissue for testing – During a biopsy procedure, your doctor removes a small sample of tissue to test for cancer cells. Your tumor’s location determines how the tissue sample is removed.
Using advanced lab tests to determine the kinds of cells involved in cancer – Doctors who specialize in analyzing blood and body tissue (pathologists) will study your biopsy samples using specialized laboratory tests, such as immunohistochemistry, cytogenetic analysis, fluorescence in situ hybridization, and molecular genetic testing. These tests provide information about pleomorphic liposarcoma that helps your doctor determine your prognosis and your treatment options.
Biopsy – To check if the tumor is MRCLS, your doctor will do a biopsy, taking a small sample from the tumor with a needle. An expert, called a pathologist, will study cells from the sample under the microscope to see what kind of tumor it is.
Treatment of Pleomorphic Liposarcoma
Treatments for pleomorphic liposarcoma include
Surgery – The goal of surgery is to remove all of the cancer cells. Whenever possible, surgeons work to remove the entire pleomorphic liposarcoma. If a pleomorphic liposarcoma grows to involve nearby organs, removal of the entire pleomorphic liposarcoma may not be possible. In those situations, your doctor may recommend other treatments to shrink the liposarcoma to make it easier to remove during an operation.
Radiation therapy – Radiation therapy uses powerful energy beams, such as X-rays and protons, to kill cancer cells. Radiation may be used after surgery to kill any cancer cells that remain. Radiation may also be used before surgery to shrink a tumor in order to make it more likely that surgeons can remove the entire tumor.
Chemotherapy – Chemotherapy uses drugs to kill cancer cells. Not all types of pleomorphic liposarcoma are sensitive to chemotherapy drugs. Careful analysis of your cancer cells by an expert pathologist can determine whether chemotherapy is likely to help you. Chemotherapy may be used after surgery to kill any cancer cells that remain or before surgery to shrink a tumor. Chemotherapy is sometimes combined with radiation therapy.
Conventional chemotherapy pleomorphic liposarcoma is of low efficiency, so new drugs and target therapy might be good options. For example, eribulin and pazopanib are new treatment options for patients with metastatic STS.[rx,rx] Eribulin mesylate was reported to have selective activity in LPS.[rx] However, the adverse effects of eribulin were severe. In a phase III trial, treatment-emergent adverse events occurred in 224 (99%) of 226 patients who received eribulin. Grade 3/4 adverse events were 152 (67%) who received eribulin.[rx] The Food and Drug Administration (FDA) approved pazopanib as second-line chemotherapy for the treatment of patients with advanced nonlipogenic STS, but still not yet for LPS.[rx] In addition, eribulin and pazopanib have not been approved for clinical use in China.
Meanwhile, antiangiogenic targeted drugs such as sunitinib, sorafenib, and pazopanib all appeared to demonstrate acceptable antitumor activity in liposarcomas.[rx–rx] Moreover, several articles reported that apatinib had a good effect on angiosarcoma[rx] and round cell liposarcoma.[rx]
Newer drugs for
Halaven® (eribulin) and Yondelis® (trabedectin) are approved for people who have not responded to earlier treatment, have widespread liposarcoma, or have cancers that cannot be removed via surgery.
Ongoing and Upcoming Clinical Trials of Targeted Therapy and Immunotherapy for Liposarcoma
Myxoid liposarcoma (MLS) is a subtype of liposarcoma that represents a distinct pathological entity characterized morphologically by tumor cells within a myxoid stroma with a rich, branching thin-walled vasculature, and focal lipomatous differentiation.
Myxoid liposarcoma is the second most common subtype (MLs). It accounts for 15–20% of liposarcomas and represents about 5% of all soft tissue sarcomas in adults. Histologically MLS shows a continuous spectrum of lesions with low-grade forms and others poorly differentiated round cell forms [rx]. The tumoral site (upper limb, lower limb, and trunk) did not result in a significant risk factor, even though the few numbers of patients with trunk localization could have hampered statistical significance. It is an important factor of this usually slow-growing, deep-seated tumor mainly located in the lower extremities is the propensity to metastasize to nonpulmonary soft-tissues as the retroperitoneum, the bone, or the contralateral limb [rx]. Furthermore, myxoid liposarcoma is particularly radiosensitive thus neoadjuvant radiation protocols may be very effective [rx–rx].
Causes of Myxoid Liposarcoma
Low grade:
Atypical lipomatous tumor / well differentiated liposarcoma (ALT / WDL):
ALT / WDL can be myxoid and focally indistinguishable from myxoid liposarcoma
ALT / WDL usually has some degree of stromal atypia however and will lack the plexiform vasculature
ALT / WDL has amplification of chromosome 12q14 (including the MDM2 gene) vs. the FUS rearrangement seen in myxoid liposarcoma
Myxoid liposarcomas are also more likely to show a predilection for signet ring lipoblasts
Extraskeletal myxoid chondrosarcoma (EMC):
Composed of cords of epithelioid malignant cells set in a similar chondromyxoid matrix
There are no cytoplasmic fat vacuoles and less prominent vasculature
Immunohistochemical staining is not helpful (both are S100 positive)
Cytogenetics can be helpful but care must be taken
EMC have t(9;22)(q22;q12) gene rearrangements in most cases that result in an EWSR1-NR4A3 fusion
EWSR1 FISH will be positive but can lead to confusion with the 2 – 5% of myxoid liposarcomas that rarely have EWSR1 rearrangements
PCR applications can be particularly helpful in this setting
In the lung, primary pulmonary myxoid sarcoma should be considered (Pathology 2017;49:792)
Lipoblastoma / lipoblastomatosis:
Can show similar histology but usually are present in patients less than 5 years old
Will have PLAG1 gene rearrangements instead of FUS or EWSR1 rearrangements
Lipoblastoma-like tumor of the vulva:
This is a recently described entity that occurs in the vulva and shares remarkable histologic overlap with lipoblastoma and myxoid liposarcoma
These lesions have been shown to have Rb loss like the spindle cell lipoma family of tumors
There is a significant difference in treatment and clinical outcome (only a limited ability to locally recur) and these lesions should be distinguished wherever possible
Myxoid dermatofibrosarcoma protuberans (DFSP):
Typically these are located superficially, which is uncommon in myxoid liposarcoma
Look carefully to distinguish between entrapped fat versus true signet ring lip blasts
Immunohistochemical staining and molecular testing can help
DFSP will be CD34 positive, S100 negative, with the inverse seen in myxoid liposarcoma and DFSP, will harbor the t(17;22)(q22;q13) COL1A1-PDGFB gene fusion
Some large reference labs offer PDGFB as a break-apart FISH assay
Myxoma:
Extremely paucicellular, lacks a prominent vascular component and no lipoblasts are found
There are numerous round cell sarcomas that may morphologically resemble high-grade myxoid liposarcoma
Low-grade areas and lip blasts can be particularly informative
Immunohistochemical and molecular differences can also be exploited (Ewing sarcoma has different partner genes than does myxoid liposarcoma, a feature that can be taken advantage of via sequencing or PCR)
Symptoms of Myxoid Liposarcoma
Most patients with myxoid liposarcoma have no symptoms until the tumor is large and invades the neighboring organs or tissues, causing tenderness, pain, or functional problems.
As previously mentioned, most patients are diagnosed with myxoid liposarcoma do not have any early symptoms and it can go unnoticed during the initial and primary stages of the disease until the tumor has grown to a large enough size to compress neighboring tissues and cause pain or decreased function.
It can sometimes be noticed as a deep-seated mass to touch.
Myxoid liposarcoma, as with all other cancers, can present with non-specific symptoms such as fevers, chills, fatigue, night sweats, anorexia, and weight loss.
If the tumor is retroperitoneal in location, it can present with specific symptoms in the abdomen, including abdominal pain, constipation, gastritis, or flank pain, swelling, and constipation or the sensation of feeling full sooner than expected after eating.
The well-differentiated type tumor is less aggressive and tends to be a large painless mass found in deeper tissues and in the retroperitoneum.
Myxoid, round cell and pleomorphic types tend to be in the arms and legs, whereas dedifferentiated tend to be in the retroperitoneum and often associated with the well-differentiated variety.
Specifically, pleomorphic liposarcoma is the least common subtype with a high rate of recurrence and poor outcomes.
Patients usually present with progressive dysphagia with weight loss
The symptoms of myxoid liposarcoma depend on where the tumor is on your body, but they include:
A new or growing lump beneath your skin, especially around or behind your knees or on your thighs
Pain or swelling
Weakness in an arm or leg that has the lump
Feeling full soon after you start eating
A new lump anywhere on your body, or an existing lump that grows persistently
Painful swelling or numbness in the area around your lump
Blood in your stool, or black or tarry stool (an indication of blood)
Blood in your vomit
Abdominal pain or cramping
Constipation
Poop that has blood or looks black or tarry
Cramping
Bloody vomit
Your belly gets larger
Recurrent molecular alteration with either t(12;16)(q13;p11.2) FUS-DDIT3 or very rarely (~2%) t(12;22)(q13;q12) EWSR1-DDIT3 rearrangements
Includes a spectrum of disease including high-grade lesions, which were formerly regarded as “round cell liposarcoma”
Has an unusual propensity to present with the first metastasis to another soft tissue or bony site (such as from one leg to the contralateral leg or to the retroperitoneum or spine
Prominent myxoid stroma with branching vasculature (so-called chicken wire vasculature)
The majority of the tumor can be nonlipogenic with only scattered lip blasts that often have a characteristic signet ring morphology
Diagnosis of Myxoid Liposarcoma
Microscopic (histologic) description
Low grade:
Paucicellular with monomorphic, stellate, or fusiform shaped cells without atypia; striking in their blandness, so much so that any significant pleomorphism should cause one to pause
Prominent plexiform vasculature (delicate thin-walled arborizing and curving capillaries that form a network reminiscent of chicken wire fencing)
These are so striking because of the overall background paucicellularity and are still present but much less obvious in high-grade tumors
Numerous signet ring lip blasts, particularly at the periphery of lobules
This imparts a lipoblastoma-like appearance
Mucoid matrix is rich in hyaluronic acid that may form large mucoid pools (so called pulmonary edema pattern)
Will be positive for stromal mucin stains such as Alcian blue
Typically there is no significant mitotic activity
There are many rare morphologic variants; an excellent review of the spectrum of histologic features can be found at Am J Clin Pathol 2012;137:229
High grade:
Hypercellular solid sheets of back to back cells with round cell or primitive cytomorphology in greater than 5% of the sampled tumor
Cells can have a small amount of hypereosinophilic cytoplasm, a finding of no clinical significance but of significant diagnostic confusion, especially in a limited sample
Pitfalls and tips
Round cell features of high-grade tumors are so cellular you can typically “walk” across nuclei in a high power field without “stepping” in matrix
When in doubt, especially in a small sample, pursue molecular testing (typically fluorescent in situ hybridization) for FUS gene rearrangement
If you are particularly certain and FUS is negative, proceed to EWSR1
Sample these tumors extensively; you likely will not see small amounts of round cell progression grossly
Can contain large areas of mature adipocytic differentiation
If the clinical or radiologic picture is concerning, sample additional tumor or do molecular testing
Location (extremity) and age of the patient (young adult) can be helpful clues in the differential diagnosis
Most of the diagnostic clues are helpful in the appropriate context but individually can be seen in many other tumor types
Plexiform vasculature and cells that look like signet rings can be found in a diverse variety of tumors
Combination of a number of clinical, radiologic, histologic and if needed, molecular features will make the diagnosis
Imaging
CT scan, magnetic resonance imaging (MRI) – If you have symptoms of MRCLS, your doctor will use imaging scans such as CT and MRI to look at where the tumor is and how big it is. They will also check for signs that the tumor has spread to other parts of the body. Although none of these techniques is specific, CT scan and MRI can be more helpful in narrowing down the differential diagnosis as both modalities can detect the percentage of a lipomatous component of the tumor. Higher fat content is associated with benign lipoma, while less fat is consistent with atypical lipoma or sarcoma. A definitive diagnosis can only be achieved by tissue examination. And in the majority of the cases, complete resection of the tumor is needed for a correct diagnosis [rx].
Removing a sample of tissue for testing – During a biopsy procedure, your doctor removes a small sample of tissue to test for cancer cells. Your tumor’s location determines how the tissue sample is removed.
Using advanced lab tests to determine the kinds of cells involved in cancer – Doctors who specialize in analyzing blood and body tissue (pathologists) will study your biopsy samples using specialized laboratory tests, such as immunohistochemistry, cytogenetic analysis, fluorescence in situ hybridization, and molecular genetic testing. These tests provide information about your liposarcoma that helps your doctor determine your prognosis and your treatment options.
Biopsy – To check if the tumor is MRCLS, your doctor will do a biopsy, taking a small sample from the tumor with a needle. An expert, called a pathologist, will study cells from the sample under the microscope to see what kind of tumor it is.
Treatment of Myxoid Liposarcoma
A standard fraction schedule was used: 2 Gy per fraction, 5 days a week.
Chemotherapy was performed in patients with more than two of these unfavorable prognostic factors: high-grade disease, tumor size > 5 cm, deep sited tumors, and positive surgical margins. Chemotherapy consisted of three or five cycles of epirubicin (60 mg/m2, Days 1-2) and ifosfamide (3 g/m2, Days 1–3) administered every 21 days.
Treatments for liposarcoma include
Surgery – The goal of surgery is to remove all of the cancer cells. Whenever possible, surgeons work to remove the entire liposarcoma. If a liposarcoma grows to involve nearby organs, removal of the entire liposarcoma may not be possible. In those situations, your doctor may recommend other treatments to shrink the liposarcoma to make it easier to remove during an operation.
Radiation therapy – Radiation therapy uses powerful energy beams, such as X-rays and protons, to kill cancer cells. Radiation may be used after surgery to kill any cancer cells that remain. Radiation may also be used before surgery to shrink a tumor in order to make it more likely that surgeons can remove the entire tumor.
Chemotherapy – Chemotherapy uses drugs to kill cancer cells. Not all types of liposarcoma are sensitive to chemotherapy drugs. Careful analysis of your cancer cells by an expert pathologist can determine whether chemotherapy is likely to help you. Chemotherapy may be used after surgery to kill any cancer cells that remain or before surgery to shrink a tumor. Chemotherapy is sometimes combined with radiation therapy.
Newer drugs for
Halaven® (eribulin) and Yondelis® (trabedectin) are approved for people who have not responded to earlier treatment, have widespread liposarcoma, or have cancers that cannot be removed via surgery.
Ongoing and Upcoming Clinical Trials of Targeted Therapy and Immunotherapy for Liposarcoma
Liposarcoma is a malignant tumor of mesenchymal origin a significant tissue diversity tumor of lip blasts, rare mesenchymal neoplasm and involves deep soft tissues including retroperitoneum and popliteal fossa [rx]. It is cancer that arises in the fat cells in soft tissue, such as that inside the thigh or in the retroperitoneum portion of the body.[rx] The relative frequency of liposarcoma in various body sites and it is much dependent on the tumor subtypes. For example, dedifferentiated liposarcoma is much the most common in retroperitoneal locations, while myxoid liposarcoma occurs in the lower extremities maximum times[rx]. Liposarcoma is exceedingly rare in the esophagus [rx][rx]. Esophageal liposarcoma typically behaves as a slow-growing tumor and involves the upper part of the throat.
Liposarcoma is a tumor that arises from fat tissue in the human body. This tumor most often occurs in the thigh, legs, behind the knee, or in the abdomen, but it can be found in other parts of the body frequently, in the retroperitoneum; and, less often, in the head and neck area. Their primary occurrence in the skin is rarer. Because liposarcoma may grow into surrounding tissues or organs in the body, it is considered a malignant tumor frequently.[rx]
Although surgical removal of the tumor is the curative treatment, some patients may benefit from chemotherapy and radiation.[rx]
Types of Liposarcoma
The World Health Organization classification of soft tissue tumors recognizes 5 types of liposarcomas. There are five types of liposarcoma, each with its own unique characteristics and behaviors.
Well-differentiated liposarcoma – It’s the most common type, and it spread to grow slowly. It has another characteristic that usually doesn’t spread to other parts of your body. Low-grade tumor cells look much like normal fat cells under the microscope.
Myxoid liposarcoma – It is an intermediate to high-grade tumor. Its cells look less normal under the microscope and may have a high-grade component. It can grow faster than well-differentiated tumors, and it’s more likely to spread to other parts of your body. Its cells can form a unique shape or pattern.
Pleomorphic liposarcoma – It is the rarest or uncommon type and is a high-grade tumor with cells that look very different from normal cells. It is a less-common form of cancer and often spreads very quickly.
Dedifferentiated liposarcoma – It gradually occurs when a low-grade tumor changes and the newer cells in the tumor are high-grade. This type of tumor is a slow-growing tumor that starts to change to a faster-growing, more aggressive type.
Round cell – It often develops in the thigh and can involve changes in the chromosomes (proteins that carry genetic information) in cells. This type can also grow faster than well-differentiated tumors and is often found in the arms or legs.
Stages
The risk of recurrence and metastasis with liposarcoma increases with a higher grade. The following are the different stages:
Stage 1A – the tumor is 5 cm or less in size, grade 1, and cancer has not spread to lymph nodes or distant sites
Stage 1B – the tumor is larger than 5 cm, grade 1, and cancer has not spread to lymph nodes or distant sites
Stage 2A – the tumor is 5 cm or less, grade 2 or 3, and cancer has not spread to lymph nodes or distant sites
Stage 2B – the tumor is larger than 5 cm, grade 2, and cancer has not spread to lymph nodes or distant sites
Stage 3A – the tumor is larger than 5 cm, grade 3, and cancer has not spread to lymph nodes or distant sites OR the tumor is any size and cancer has spread to nearby lymph nodes but not other sites
Stage 4 – the tumor is any size and any grade, and has spread to lymph nodes and/or to other sites
Causes of Liposarcoma
The specific cause of liposarcoma is still unknown to us. But clinically, it can be first noticed particularly in the extremity in an area of recent trauma, injured soft tissue where the patient may find a large mass, however, the cause and effect are quite likely purely coincidental. Liposarcoma generally contributes to a change in some of the genes that are normally present in fat cells. A series of abnormalities in these genes (mutations or DNA alterations) can lead to malignant cancerous changes characterized by uncontrollable growth.
Some inherited or acquired DNA mutations are found, or defects can make you more prone to developing a soft tissue sarcoma
Basal cell nevus syndrome increases the risk of basal cell skin cancer, rhabdomyosarcoma, and fibrosarcoma.
Inherited retinoblastoma in the eyes causes a kind of childhood eye cancer, but it can also increase the risk of other soft tissue sarcomas.
The Li-Fraumeni syndrome increases the risk of many kinds of cancer, often from radiation exposure.
Gardner’s syndrome leads to cancers in the stomach or bowel and gastrointestinal area.
Neurofibromatosis can cause nerve sheath tumors.
Tuberous sclerosis can cause rhabdomyosarcoma.
Werner’s syndrome can cause many health problems, including an increased risk of all soft tissue sarcomas.
Exposure to certain toxins, such as dioxin, vinyl chloride, arsenic, and herbicides that contain phenoxy acetic acid at high doses may increase your risk of developing soft tissue sarcomas.
Radiation exposure, especially from radiation therapy, can be a risk factor. Radiation therapy often treats more common cancers such as breast cancer, prostate cancer, or lymphomas. However, this effective therapy can increase your risk of developing certain other forms of cancer, such as a soft tissue sarcoma.
Symptoms of Liposarcoma
Most patients with liposarcoma have no symptoms until the tumor is large and invades the neighboring organs or tissues, causing tenderness, pain, or functional problems.
As previously mentioned, most patients are diagnosed with liposarcoma do not have any early symptoms and it can go unnoticed during the initial and primary stages of the disease until the tumor has grown to a large enough size to compress neighboring tissues and cause pain or decreased function.
It can sometimes be noticed as a deep-seated mass to touch.
Liposarcoma, as with all other cancers, can present with non-specific symptoms such as fevers, chills, fatigue, night sweats, anorexia, and weight loss.
If the tumor is retroperitoneal in location, it can present with specific symptoms in the abdomen, including abdominal pain, constipation, gastritis, or flank pain, swelling, and constipation or the sensation of feeling full sooner than expected after eating.
The well-differentiated type tumor is less aggressive and tends to be a large painless mass found in deeper tissues and in the retroperitoneum.
Myxoid, round cell and pleomorphic types tend to be in the arms and legs, whereas dedifferentiated tend to be in the retroperitoneum and often associated with the well-differentiated variety.
Specifically, pleomorphic liposarcoma is the least common subtype with a high rate of recurrence and poor outcomes.
Patients usually present with progressive dysphagia with weight loss
The symptoms of liposarcoma depend on where the tumor is on your body, but they include:
A new or growing lump beneath your skin, especially around or behind your knees or on your thighs
Pain or swelling
Weakness in an arm or leg that has the lump
Feeling full soon after you start eating
A new lump anywhere on your body, or an existing lump that grows persistently
Painful swelling or numbness in the area around your lump
Blood in your stool, or black or tarry stool (an indication of blood)
Blood in your vomit
Abdominal pain or cramping
Constipation
Poop that has blood or looks black or tarry
Cramping
Bloody vomit
Your belly gets larger
Diagnosis of Liposarcoma
The most common type of esophageal liposarcoma is the well-differentiated type tumor, which is characterized histologically by mature adipocytes with a variable amount of fibrous stroma cells containing atypical nuclei are also present. It has three main subtypes, the lipoma-like subtype, the sclerosing subtype, and the inflammatory subtype. The lipoma-like subtype frequently has lip blasts and identifies atypical cells. This type closely benign lipoma. The sclerosing subtype has abundant fibrous tissue areas with scant lipogenic components also present in histology examination. The inflammatory subtype has chronic inflammatory tissue, with a presence of B cells. These cells can pose a diagnostic challenge as inflammatory infiltrate can obscure the adipocytes.
The second most common type of esophageal liposarcoma is myxoid liposarcoma (MLS), which is subdivided into low-grade and high-grade tumors. Low-grade have low cellularity with bland nuclei and a rich, prominent network of curving capillaries, blood vessels, resembling a chicken-wire pattern-like in appearance. High-grade MLS are identified hypercellular with solid sheets of round cells comprising at least 5% of the tumor.
The least common type of esophageal liposarcoma is the common type, which has marked pleomorphic cancer-causing cells occupying at least 65% of the tumor and at least focal areas of typical liposarcoma.
The most common site of esophageal liposarcoma is the upper esophagus frequently, which explains most of the symptoms associated with this neoplasm[rx]. In some cases, when the size of the tumor is large, patients also present with polyp/mass protruding from the mouth [rx][[rx].
Imaging
CT scan, magnetic resonance imaging (MRI) – and esophagogastroduodenoscopy, can be used to diagnose esophageal liposarcoma. Although none of these techniques is specific, CT scan and MRI can be more helpful in narrowing down the differential diagnosis as both modalities can detect the percentage of a lipomatous component of the tumor. Higher fat content is associated with benign lipoma, while less fat is consistent with atypical lipoma or sarcoma. A definitive diagnosis can only be achieved by tissue examination. And in the majority of the cases, complete resection of the tumor is needed for a correct diagnosis [rx].
Removing a sample of tissue for testing – During a biopsy procedure, your doctor removes a small sample of tissue to test for cancer cells. Your tumor’s location determines how the tissue sample is removed.
Using advanced lab tests to determine the kinds of cells involved in cancer – Doctors who specialize in analyzing blood and body tissue (pathologists) will study your biopsy samples using specialized laboratory tests, such as immunohistochemistry, cytogenetic analysis, fluorescence in situ hybridization, and molecular genetic testing. These tests provide information about your liposarcoma that helps your doctor determine your prognosis and your treatment options.
Treatment of Liposarcoma
Treatments for liposarcoma include
Surgery – The goal of surgery is to remove all of the cancer cells. Whenever possible, surgeons work to remove the entire liposarcoma. If a liposarcoma grows to involve nearby organs, removal of the entire liposarcoma may not be possible. In those situations, your doctor may recommend other treatments to shrink the liposarcoma to make it easier to remove during an operation.
Radiation therapy – Radiation therapy uses powerful energy beams, such as X-rays and protons, to kill cancer cells. Radiation may be used after surgery to kill any cancer cells that remain. Radiation may also be used before surgery to shrink a tumor in order to make it more likely that surgeons can remove the entire tumor.
Chemotherapy – Chemotherapy uses drugs to kill cancer cells. Not all types of liposarcoma are sensitive to chemotherapy drugs. Careful analysis of your cancer cells by an expert pathologist can determine whether chemotherapy is likely to help you. Chemotherapy may be used after surgery to kill any cancer cells that remain or before surgery to shrink a tumor. Chemotherapy is sometimes combined with radiation therapy.
Newer drugs for liposarcoma
Halaven® (eribulin) and Yondelis® (trabedectin) are approved for people who have not responded to earlier treatment, have widespread liposarcoma, or have cancers that cannot be removed via surgery.
Ongoing and Upcoming Clinical Trials of Targeted Therapy and Immunotherapy for Liposarcoma
Vertebral metastases represent the secondary involvement of the vertebral spine by hematogenously-disseminated metastatic cells. They must be included in any differential diagnosis of a spinal bone lesion in a patient older than 40 years.
Metastases to the spine can involve the bone, epidural space, leptomeninges, and spinal cord. The spine is the third most common site for metastatic disease, following the lung and the liver [rx] and the most common osseous site [rx] . These are more commonly found as bone metastasis and may present with symptoms of spinal canal invasion and cord compression
This will focus only on the metastasis involving the bony structures of the spine; please refer to the specific articles for other spinal metastatic diseases:
intradural extramedullary metastases
intramedullary metastases
Symptoms of Spine Metastases
Vertebral lesions are very frequently asymptomatic in the setting of widespread metastatic disease and are thus often found incidentally when imaging is performed for other reasons (e.g. staging).
Lesions may become symptomatic due to bone pain, pathological compression fractures, or extension into the spinal canal with cord compression and ensuing neurological deficits. The most common primary malignancies to involve the vertebrae include:
breast cancer
lung cancer
prostate cancer
lymphoma
renal cell carcinoma
gastrointestinal tract malignancies
melanoma
pancreatic cancer
thyroid carcinoma
carcinoid
Metastases are either osteoblastic or osteolytic, however, osteoid formation and mineralization is of limited help in determining the primary tumor as some metastases may secrete osteoblast- and osteoclast-stimulating factors at the same time. The new bone formation may also occur after chemotherapy or radiation therapy. Having said that some primaries more frequently result in sclerosis than others.
Primaries with predominantly osteoblastic metastases (sclerotic extradural bone lesions) include:
prostate carcinoma
osteosarcoma
medullary thyroid carcinoma
Primaries with predominantly osteolytic metastases, that may rarely become osteoblastic (mixed sclerotic and lytic extradural bone lesions) include:
breast cancer
lymphoma
urothelial carcinoma
lung cancer
gastrointestinal tract cancers
renal cell carcinoma
malignant melanoma
multiple myeloma
Diagnosis of Spine Metastases
The vertebra is the most common site affected, followed by the femur, pelvis, ribs, sternum, proximal humerus, and skull. Bone metastases may be asymptomatic or manifest in a variety of ways termed skeletal-related events (SRE) as a result of the destruction of normal bone architecture. The following further describes SRE:
Bone pain – Pain associated with bone metastasis is a frequent symptom. It is typically gradual in onset and described as a dull, boring pain that is worse at night.
Nerve root or spinal cord compression – Bone metastases causing nerve root compression can present as radicular pain different from mechanical pain.
Spinal cord compression – As the vertebra is the most common site of metastasis, a significant complication includes spinal cord compression, which is an oncologic emergency. Metastatic spinal cord compression occurs either through pathological vertebral collapse or direct epidural extension. Patients may present initially with back pain. Limb weakness is the second most common symptom of cord compression. Sensory symptoms include paresthesias and numbness at and below the level of cord compression. Autonomic dysfunction, including bowel and bladder incontinence and impotence, is typically a late presentation. Early recognition, workup, and prompt surgical consultation are pertinent to prevent permanent neurological damage with resultant paraplegia.
Hypercalcemia – In the setting of malignancy, this can be multifactorial and confers a poor prognosis overall. Osteolytic bone metastases are associated with 20% of the cases of hypercalcemia of malignancy. In osteolytic metastases, enhanced osteoclastic bone resorption occurs as a result of the release of humoral factors by tumor cells that in turn stimulate osteoclasts and lead to unchecked bone resorption and hypercalcemia. Symptoms of hypercalcemia include nausea, anorexia, abdominal pain, constipation, and mental status changes. Immediate treatment for hypercalcemia includes IV hydration
Pathological fractures – Bone metastases cause bone destruction, leading to complete or impending fracture at the site of pathology either spontaneously or with minimal trauma. The presentation is dependent on the site of the fracture; however, constant pain is a prevalent symptom. Fractures of the thoracic and lumbar spine present with pain characteristically worse with sitting or standing. Pathologic fractures result in significant morbidities resulting from pain, radiculopathy (e.g., sciatica with pelvic fracture), deformities, and immobility.
Myelophthisis – Symptomatic anemia resulting from infiltration of the bone marrow with metastatic tumor cells. Pancytopenia may also be present in late stages.
Imaging
It is pertinent to identify bone metastasis early, both types of staging and prognostication as well as the implementation of prophylactic and treatment strategies which may lead to decreased morbidity and mortality. Bone metastases can be characterized as osteolytic, sclerotic, or mixed on imaging studies.
Plain Radiograph
X-rays – or plain radiograph is the initial imaging of choice in patients presenting with bone pain. Plain films are used to assess abnormal radionuclide uptake or to detect pathological fractures. Metastatic lesions can have virtually any appearance. They can mimic a benign lesion or an aggressive primary bone tumor. It can be difficult, if not impossible, to judge the origin of the tumor from the appearance of the metastatic focus, although some appearances are fairly characteristic.
Plain radiography – best detects osteolytic lesions, but they may not be apparent until they are greater than 1 to 2 centimeters and with loss of 50% of the bone mineral content at the site of disease. Osteolytic lesions are seen as thinning of trabeculae and ill-defined margins on radiographs, while sclerotic lesions appear nodular and well-circumscribed as a result of thickened trabeculae. Plain films tend to be insensitive, especially in detecting bone metastases and with asymptomatic and subtle lesions. Progression of disease and response to therapy can be monitored with plain films and further correlation with other modalities. Sclerosis or new bone formation in osteolytic metastatic lesions is demonstrated by the sclerotic rim of reactive bone, which starts at the periphery and eventually involves the center with continued healing. Purely sclerotic lesions are more difficult to assess. Major disadvantages of plain radiographs include poor sensitivity.
Computed Tomography – Computed tomography (CT) is more sensitive (74%) than plain radiographs. It is useful in the evaluation of cortical and trabecular bone as well as in the assessment of the osteolytic and sclerotic lesions. CT scan is advantageous as it can determine staging and treatment response of other organs in addition to bone and objectively assess reactive sclerosis by calculating the change in Hounsfield units. Ribs are better evaluated with CT due to the high cortex to marrow ratio. The appearance of CT will depend on the degree of mineralization of the metastasis. The more common lytic metastases appear as regions of soft-tissue attenuation with irregular margins. The mass may breach the cortex and result in a compromise of the spinal canal. Histopathology of bone metastases is only employed for diagnostic purposes in patients of primary unknown cancers or in the presence of multiple cancers.
MRI – In the detection of bone metastasis, MRI demonstrates a sensitivity of 95% and specificity 90%. MRI is also more advantageous than a bone scan as it can detect marrow involvement before the development of osteoblastic lesions. It can be used with women who are pregnant and used to detect spinal cord compression. Bone metastases manifest as low T1 signal and high intensity on the T2 weighted sequence. Whole-body MRI requires 40 to 45 minutes to perform and involves short-tau inversion recovery (STIR) and/or T1-weighted sequences.
Nuclear Medicine – Nuclear medicine scans also are used to detect bone metastases using osteotropic radioisotopes; these include skeletal scintigraphy, SPECT, and PET scan.
Skeletal scintigraphy – or bone scan is the most commonly used radionuclide imaging which uses 99mTc-MDP employed in the detection of skeletal metastases. Radioisotopic imaging methods depict bone metastatic lesions as areas of increased tracer uptake.
Bone scan – provides the advantage of scanning the whole skeleton and has a high sensitivity (78%) therefore resulting in early diagnosis. When osteoblastic activity is prominent, the lesions are readily detected using radionuclide bone scanning. However, bone scans have low specificity for differentiating between benign and malignant bone lesions and for the detection of predominantly osteolytic lesions. Bone scans can be used to monitor the progression of disease and response to treatment.
SPECT uses 99mTc-MDP radioisotopes – uptake to detect bone lesions; however, images are acquired in cross-sectional rather than a planar fashion. SPECT has a higher specificity of 91% compared to skeletal scintigraphy.
PET – is a nuclear medicine technique that uses the radiotracers 18F FDG or 18FNaF for the detection of skeletal metastases. 18F FDG PET scan identifies bone metastases based on a high glucose metabolism exhibited by neoplastic cells. PET has a better spatial resolution compared to skeletal scintigraphy. 18F NaF-PET is proven to be substantially more sensitive and specific than bone scan and SPECT for the detection of bone metastases. Combining imaging techniques and modalities allows for improved visualization both anatomically and functionally, leading to increased diagnostic accuracy. One example of this is the 18F-Sodium fluoride (18F-NaF) PET/CT bone scanning which has significantly greater sensitivity (100%) and specificity (97%). Other hybrid imaging techniques include SPECT/CT, PET/CT, and PET/MRI.
Bone scintigraphy – is an effective means of assessing the metabolic activity of the spine, while plain radiographs can only demonstrate lesions with a loss of 30%–50% of bone mineral content[rx, rx]. Technetium‐99m (99m Tc) planar bone scintiscans detect metastatic bone deposits through increased osteoblastic activity, considered to be an indirect marker of an oncological process. For this reason, it is considered to be the most efficient modality for screening the whole body for metastasis[rx, rx]. 18F‐fluoro‐deoxy‐D‐glucose positron emission tomography (18FDG PET) offers superior spatial resolution and improved sensitivity, which is superior to bone scintigraphy in the detection of osteolytic metastases, while osteoblastic metastases show lower metabolic activity and are frequently undetectable by PET[rx, rx].
Blood tests – can aid in supporting the diagnosis of bone metastases. Complete blood count and a comprehensive metabolic panel should be obtained routinely. CBC may reveal anemia, thrombocytopenia, or pancytopenia in late stages. Serum calcium and alkaline phosphatase may be elevated due to ongoing osteolysis. Bone turnover markers are still being studied as indicators of bone resorption. Tartrate-resistant acid phosphatase has been proven to elevated in patients with breast and prostate cancer with bone metastases. Objective scoring models such as the Mirel classification system for long bones and assessment of spinal stability in addition to imaging criteria are used to determine the surgical necessity for impending pathological fractures.
Treatment of Spine Metastases
The therapeutic approach to bone metastases should be a multidisciplinary approach targeted at preserving the quality of life, including pain control, minimizing SREs, and achieving local tumor control. It is pertinent to consider a multitude of factors including the extent of disease spread, performance status, impending fracture, and side effects when creating the initial approach for the treatment of bone metastases. [rx][rx][rx][rx]
NSAIDs – A major aspect of treating bone metastases is analgesia/pain control for the debilitating pain that occurs with bone metastases. Pain control can be initiated with NSAIDs and titrated up to or in conjunction with narcotics as needed for symptom relief. Glucocorticoids may also be useful for additional pain control.
Osteoclast inhibitors – (bisphosphonates and denosumab) decrease morbidity and mortality associated with bone metastases as they reduce skeletal-related events and can be used for analgesia to some extent.
Local radiation for symptomatic bone metastases – is a significant component of the palliative approach in providing analgesia. It is also used postoperatively to consolidate continued bone healing. External beam radiation is the standard approach for painful bone metastases and is beneficial in reducing pain by up to 50% to 80%. Several studies have proven that a single 8 Gy fraction compared to more prolonged or fractionated radiation is non-inferior; however, it may carry a higher need for pretreatment (20 % vs. 8%). Stereotactic body radiation therapy spares the normal tissue while delivering highly conformal radiation to the affected area. There are no clear guidelines on stereotactic body radiotherapy (SBRT) use however it may be indicated over external beam radiation therapy (EBRT) in specific instances of bone metastases specifically vertebral from certain radio-resistant neoplasms.
Bone-targeted radiopharmaceutical therapy – (e.g., beta-emitting agents strontium-89, alpha-emitting radium-223) provides the specific advantage of treatment of diffuse pain associated with osteoblastic bone metastases. It is typically used in bowel movement associated with prostate and breast cancer or for analgesia in radiation therapy for refractory pain. Systemic chemotherapy when amenable, aimed at the primary tumor can also provide analgesia by reduction of tumor size and control of tumor spread.
Surgery is indicated – for impending or complete fracture, mechanical stability, and spinal cord compression. In cases where the spread of the primary cancer is limited to a single bone lesion, en bloc resection of the metastasis can be done by means of local tumor control.
Local ablation via radiofrequency ablation (RFA), cryoablation, and focused ultrasound (FUS) should be considered for patients with persistent pain following radiation therapy or patients with recurrent pain
National Institute for Health and Care Excellence clinical guideline for the management of spinal cord compression
In November 2018, NICE issued a clinical guideline for the diagnosis and management of adults at risk of or with MSCC.[rx]The guidelines contained treatment algorithms for patients with symptoms suggestive of spinal metastases. The guideline proposed the patient treatment pathways shown in. Treatment of patients with spinal metastases and MSCC can be broadly divided into three pathways.[rx]
Treatment of patients with spinal metastases and prevention of metastatic spinal cord compression
Patients with painful spinal metastases should be offered conventional analgesics, i.e. non-steroidal anti-inflammatory drugs. Those patients with intractable pain should be considered for specialist pain care that includes invasive procedures and neurosurgical interventions. Patients with spinal metastases from breast and prostate cancer should be offered bisphosphonates to alleviate pain and reduce the risk of pathological fracture/collapse of the spine. Those patients with non-mechanical spinal pain should be given single-fraction palliative radiotherapy. This should also be considered in those who are completely paralyzed. In asymptomatic patients, radiotherapy should not be administered.
Two vertebral augmentation techniques, vertebroplasty, and kyphoplasty should be considered in those with mechanical spinal pain resistant to conventional analgesics and no evidence of MSCC or spinal instability.
Surgery should be preferred when there is evidence of progressive disease mainly to prevent MSCC. It should also be considered in those with spinal metastases and mechanical pain resistant to conventional analgesics and in those with evidence of spinal instability.
Treatment of threatened spinal cord in patients with metastatic spinal cord compression
In patients with severe mechanical pain suggestive of spinal instability or those with neurological symptoms or signs suggestive of MSCC, the spine should be stabilized. Patients should be monitored regularly, especially during sitting from supine to 60 degrees. If patients continue to deteriorate, then they should revert back to the lying position or to the position in which there is minimal pain/neurological symptoms. In those patients not suitable for definitive treatment, the aim of the treatment should be to help the patient to achieve a comfortable position and mobilization. This is usually achieved by using orthoses.
Corticosteroids should be given to all patients with MSCC unless contraindicated. Dexamethasone at 16 mg as a loading dose should be given followed by a short course of 16 mg dexamethasone daily until definitive treatment is employed. After definitive treatment, the dose of dexamethasone should be reduced gradually over 5–7 days and then stopped. In those patients in whom symptoms have deteriorated, the dose of dexamethasone can be increased temporarily.
Definitive treatment of metastatic spinal cord compression
The definitive treatment should be given as early as possible, ideally within 24 hours of the diagnosis of MSCC. Before this, a diagnosis of the primary location of the tumor should be made. In addition, an attempt should be made to study the extent of the disease. A scoring system such as the Tokuhashi scoring system and the American Society of Anesthesiologists grading for the overall patient condition should be used to assess whether surgery is appropriate.
Radiotherapy
Patients unsuitable for surgery should receive radiotherapy within 24 hours, 7 days a week. Fractionated radiotherapy is the definitive treatment of choice for patients with epidural tumor without neurological dysfunction, mechanical pain or spinal instability. It is also an appropriate first-line treatment for patients with a good prognosis. Radiotherapy should not be given to patients with MSCC who are waiting for surgery but fractionated radiotherapy should be offered to all patients postoperatively once their wound has healed.
Supportive care and rehabilitation
Supportive care includes thromboprophylaxis, management of pressure ulcers, bladder and bowel continence, circulatory and respiratory functions, and access to specialist rehabilitation care at home.[rx]
Radiation
The aim of radiation is to alleviate pain and to prevent recurrence and tumor growth.[rx] It is indicated when the spine is stable, if the tumor is radiosensitive and the patient’s neurological condition is stable, or if the patient is in poor medical condition or has a life expectancy < 3–6 months and has had complete paraplegia for > 24 hours.[rx]
Recently, new approaches, such as intensity-modulated radiotherapy (IMRT),[rx] or stereotactic body radiotherapy, have been suggested for the treatment of vertebral metastases.[rx]
Systemic therapies
Corticosteroids
Intravenous or oral corticosteroids have been found to provide improvement or resolution of neurological symptoms and pain in patients with epidural spinal metastases.[rx] It should be noted that there is no standard dosage regimen for corticosteroids. They are often used before surgery.[rx]
In patients with MSCC undergoing surgical decompression, corticosteroids are often used in combination with radiotherapy.[rx]
Bisphosphonates and denosumab
Bisphosphonates are known to impair osteoclastic activity and so they reduce tumor-related resorption of bone.[rx,rx] Currently, bisphosphonates are used to alleviate metastatic bone pain and to reduce SREs such as pathological fractures, hypercalcemia, and MSCC. Bisphosphonates are also used to reduce the frequency of surgery and radiation therapy.[rx,rx] Bisphosphonates such as pamidronate (Aredia®; Novartis Pharmaceuticals Corporation), clodronate (Bonefos®, Clasteon®, Loron®; Bayer), ibandronate (Bondronat®; F. Hoffmann-La Roche Ltd), alendronate (Fosamax®; Merck Sharp & Dohme Corporation) and zoledronate (Aclasta®; Novartis) have all been found to be effective in the treatment of hypercalcemia.[rx] Although radiotherapy is the main treatment for reducing bone pain, bisphosphonates can be used as an alternative therapy, which in turn will considerably reduce the frequency of radiotherapy.[rx,rx] The effect of bisphosphonates on pain is not dependent on the nature or type of the tumor (i.e. sclerotic or lytic).[rx] The efficacy of these drugs has been seen in breast cancer, multiple myeloma, and other osteolytic metastases.57 Although bisphosphonates have been found to be effective in preventing skeletal-related complications, they are not so effective in reducing pain in patients with prostate cancer.[rx]
Recently, monoclonal antibody therapy with denosumab, a specific inhibitor of RANKL, has been found to be effective in delaying and preventing SREs.[rx]
Chemotherapy
The benefits of chemotherapy are limited in spinal metastases, as patients are usually at a late stage of disease.[rx] Chemotherapy can be given on its own or in combination with surgery and hormonal therapy.[rx]
Radioisotopes
Radioisotopes are administered systematically and act as local radiation therapy to the spine.[rx] Radioisotopes include strontium-89 and rhenium-186. Although radioisotopes are found to reduce pain in patients with spinal metastases, these can cause irreversible bone marrow suppression and, for this reason, they are recommended for use in those with good marrow function and in whom no other treatment is available.[rx]
Hormonal therapies
Hormonal therapies are a major treatment modality for metastatic breast and prostate cancer. As an example, a new drug, abiraterone (Zytiga®; Janssen), has recently been developed which has improved outcomes in men with metastatic castration-resistant prostate cancer.[rx,rx]
Surgery
The main aims of surgery are to remove the tumor, achieve spinal stability, and reconstruct the vertebral column.7 Surgery may also help with diagnosing the origin of the tumor and in relieving neurological symptoms.7 In those with solitary renal cell carcinoma metastases, surgery can increase disease-free survival.[rx] Current indications for surgery are (1) radioresistant tumor such as renal or colon carcinoma, (2) evidence of neurological function deterioration or tumor progression despite radiotherapy, (3) radiological images showing fragments of bone in the spinal canal, (4) spine instability due to fracture and causing pain and neurological deficit, (5) neurological deficit for > 24 hours, or significant MSCC, and (6) life expectancy of at least 3 months.[rx,rx]
Surgery should be considered only if it would increase the patient’s survival by > 3 months. The aim of this treatment is to decompress the spinal cord and stabilize the spine. Posterior decompression alone should be used only in cases of isolated epidural tumor or neural arch metastases without bony instability. In those in whom metastasis involves the vertebral body and who are therefore at increased risk of spinal instability, posterior decompression by internal fixation, with or without bone grafting, should be carried out. Reconstruction of the vertebral body should be carried out in patients with MSCC and vertebral body involvement who are expected to survive < 1 year, whereas in those expected to survive > 1 year, reconstruction of the vertebral body with anterior bone graft should be undertaken. In rare circumstances such as solitary renal or thyroid metastasis following complete staging, en bloc excisional surgery should be carried out.
Vertebral augmentation
Two techniques, percutaneous vertebroplasty and kyphoplasty, initially developed for the treatment of painful vertebral haemangiomas, are now used effectively in treating painful pathological fractures caused by metastatic spinal disease.[rx] Vertebroplasty involves an injection of polymethylmethacrylate (PMMA) into the compression fracture whereas in kyphoplasty an inflatable balloon is placed in the vertebral body and PMMA is injected.[rx,rx] Although these interventions can lead to significant pain reduction and greater mobility,[rx,rx] are contraindicated in SCC because of pathological fractures as they do not relieve cord compression.[rx] Complications of these techniques include leakage of PMMA, misplacement of PMMA, and hematogenous embolization of PMMA to the lungs.[rx]
Non‐operative Measures
Oral analgesia as titrated by the World Health Organization Analgesic Ladder is considered the first line in the treatment of bony pain. Morphine is the most commonly used opioid for moderate to severe pain and may be combined with adjuvant medications such as tricyclic antidepressants and corticosteroids. Side effects of medications can limit opioid dosage and cause significant morbidity. These include delirium, constipation, pruritus, nausea, vomiting, sedation, myoclonus, and respiratory depression[rx], [rx], [rx]. Other non‐invasive methods of pain relief include cutaneous stimulation, continuous repositioning, spine cryoablation, and regional nerve blocks[rx], [rx], [rxx].
Monthly infusions of bisphosphonates like zoledronic acid to patients with bone metastases reduce the frequency and delays the onset of skeletal‐related events[rx]. Their administration also provides significant improvements in bone pain and quality of life[rx]. Bisphosphonate use in patients with spinal metastases has increased in the past decade. Their use has decreased bone pain scores and reduced skeletal‐related events such as the need for local radiotherapy, hypocalcemia, pathologic fracture, and spinal cord compression[rx, rx]. Once injected, bisphosphonates are internalized by osteoclasts. This leads to a decrease in osteoclast activity and viability[rx]. Complications of bisphosphonate therapy include renal impairment and mandibular osteonecrosis[rx, rx]. Bisphosphonate demonstrates maximal effectiveness and safety when combined with either single or multiple fraction radiotherapy[rx, rx]. This synergism is due to the fact that radiotherapy is believed to reduce numbers of tumor‐produced osteoclast activating factors[rx]. The American Society of Clinical Oncology guidelines and the International Expert Panel guidelines recommend starting bisphosphonates when the first radiographic indication of metastatic deposits in the spine is noted[rx, rx].
Denosumab, a fully human monoclonal antibody to RANK‐L, has demonstrated in clinical trials inhibition of osteoclast‐mediated bone destruction in breast, prostate, and myeloma tumors, and is considered non‐inferior to zoledronic acid[rx], [rx]. A meta‐analysis performed by Lipton et al. compared the efficacy of denosumab to zoledronic acid in preventing skeletal‐related events in people with prostate cancer, breast cancer, solid tumors, or multiple myeloma. Denosumab increased the time to the first on‐study skeletal‐related event by 8.21 months and reduced the risk of a first skeletal‐related event by 17%[rx].
Chemotherapy is very seldom considered in the targeted treatment of metastatic spinal tumors due to its systemic nature and also owing to the fact that it requires an extended course of administration prior to pain relief[rx]. Complications are the source of morbidity and fear for the patient and include pain, gastrointestinal abnormalities, hematological disturbances, immunosuppression, and biopsychosocial sequelae (alopecia and infertility)[rx].
Pain caused by bone metastases has multiple causes, including periosteal elevation and inflammation[rx]. Radiopharmaceuticals may be used in the palliation of bony pain. Ethylenediamine tetramethylene phosphonic acid is an IV radioisotope that preferentially binds to osteoplastic metastases and osteosarcomas, and a significant analgesic effect may be achieved in 83%–93% of patients[rx]. Strontium‐89 chloride infusions have also been trialed as a similar treatment. Response rates vary in the published literature from minimal to up to 77%[rx]. Corticosteroids produce effects that include mood elevation, an anti‐inflammatory effect, and reduction of spinal cord edema in bony metastases[rx]. There is good evidence supporting the use of high-dose dexamethasone (64 mg/day) in the treatment of pain from spinal metastases, particularly if epidural compression is present. It is associated with significant pain relief and the ability to remain ambulatory in up to 81% of patients[rx].
Psychiatry
Involvement of psychiatric services may be necessary in patients with a diagnosis of metastatic spinal disease. Psychological complications in this instance usually manifest as anxiety, depression, adjustment disorder, and loss of self‐esteem[rx, rx]. Studies have shown that up to 50% of such patients may experience psychological issues following such a diagnosis[rx, rx]. Early involvement and assessment of this cohort of patients by psychiatric services is essential in the multidisciplinary management and treatment of spinal metastases.
Nutrition
Nutritional support is another consideration in the approach to metastatic spinal disease[rx]. The goals of nutrition support include preventing or reversing nutrient deficiencies, maximizing quality of life, aiding immunologic function, and preserving lean body mass.
This cohort is at risk of anorexia and cachexia[rx]. Important considerations include dysphagia after radiation, oesophagitis, and reduced motility owing to pain medications[rx]. Anorexia has been noted to be an almost universal side effect in individuals with widely metastatic disease[rx]. The multidisciplinary team must also consider decreased caloric intake as a result of diminished appetite and malaise and tumor competition for nutrients. Constipation may be secondary to opiate analgesia or spinal cord involvement by tumors causing an upper motor neuron lesion. Malnutrition may also exacerbate this. The addition of dietary calcium and vitamin D for bone health in the patients at risk of therapy‐associated fractures is warranted.
Screening and nutrition assessment should be interdisciplinary. Physicians, nurses, dietitians, and social workers (as members of the health‐care team) should all participate in nutritional management throughout the continuum of the management of the metastatic spinal disease. Such screening tests include the prognostic nutrition index[rx].
Suggestions for appetite improvement include keeping a daily menu, snacking between meals, eating small and frequent meals, and adding extra protein to meals[rx, rx]. Progestational agents such as megestrol acetate and medroxyprogesterone can lead to appetite stimulation and subsequent weight gain[rx]. The preferred method of nutritional support is via the oral route. If the GI tract is rendered dysfunctional, TPN may be indicated[rx].
Physiotherapy
Physiotherapists play a central role in the multidisciplinary approach to spinal metastasis. Their role is to maximize the quality of life by maintaining patient mobility and facilitating their capacity to perform activities of daily living[rx, rx]. Pain reduction therapies may also be employed, such as hot/cold packs, massage, and electrical stimulation. Assistive devices or orthotics, such as frames, canes, and thoracolumbosacral orthosis (TLSO), are provided by the physiotherapy department to patients with spinal metastasis when required[rx].
Vertebral metastases represent the secondary involvement of the vertebral spine by hematogenously-disseminated metastatic cells. They must be included in any differential diagnosis of a spinal bone lesion in a patient older than 40 years.
Metastases to the spine can involve the bone, epidural space, leptomeninges, and spinal cord. The spine is the third most common site for metastatic disease, following the lung and the liver [rx] and the most common osseous site [rx] . These are more commonly found as bone metastasis and may present with symptoms of spinal canal invasion and cord compression
This will focus only on the metastasis involving the bony structures of the spine; please refer to the specific articles for other spinal metastatic diseases:
intradural extramedullary metastases
intramedullary metastases
Symptoms of Vertebral Metastases
Vertebral lesions are very frequently asymptomatic in the setting of widespread metastatic disease and are thus often found incidentally when imaging is performed for other reasons (e.g. staging).
Lesions may become symptomatic due to bone pain, pathological compression fractures, or extension into the spinal canal with cord compression and ensuing neurological deficits. The most common primary malignancies to involve the vertebrae include:
breast cancer
lung cancer
prostate cancer
lymphoma
renal cell carcinoma
gastrointestinal tract malignancies
melanoma
pancreatic cancer
thyroid carcinoma
carcinoid
Metastases are either osteoblastic or osteolytic, however, osteoid formation and mineralization is of limited help in determining the primary tumor as some metastases may secrete osteoblast- and osteoclast-stimulating factors at the same time. The new bone formation may also occur after chemotherapy or radiation therapy. Having said that some primaries more frequently result in sclerosis than others.
Primaries with predominantly osteoblastic metastases (sclerotic extradural bone lesions) include:
prostate carcinoma
osteosarcoma
medullary thyroid carcinoma
Primaries with predominantly osteolytic metastases, that may rarely become osteoblastic (mixed sclerotic and lytic extradural bone lesions) include:
breast cancer
lymphoma
urothelial carcinoma
lung cancer
gastrointestinal tract cancers
renal cell carcinoma
malignant melanoma
multiple myeloma
Diagnosis of Vertebral Metastases
The vertebra is the most common site affected, followed by the femur, pelvis, ribs, sternum, proximal humerus, and skull. Bone metastases may be asymptomatic or manifest in a variety of ways termed skeletal-related events (SRE) as a result of the destruction of normal bone architecture. The following further describes SRE:
Bone pain – Pain associated with bone metastasis is a frequent symptom. It is typically gradual in onset and described as a dull, boring pain that is worse at night.
Nerve root or spinal cord compression – Bone metastases causing nerve root compression can present as radicular pain different from mechanical pain.
Spinal cord compression – As the vertebra is the most common site of metastasis, a significant complication includes spinal cord compression, which is an oncologic emergency. Metastatic spinal cord compression occurs either through pathological vertebral collapse or direct epidural extension. Patients may present initially with back pain. Limb weakness is the second most common symptom of cord compression. Sensory symptoms include paresthesias and numbness at and below the level of cord compression. Autonomic dysfunction, including bowel and bladder incontinence and impotence, is typically a late presentation. Early recognition, workup, and prompt surgical consultation are pertinent to prevent permanent neurological damage with resultant paraplegia.
Hypercalcemia – In the setting of malignancy, this can be multifactorial and confers a poor prognosis overall. Osteolytic bone metastases are associated with 20% of the cases of hypercalcemia of malignancy. In osteolytic metastases, enhanced osteoclastic bone resorption occurs as a result of the release of humoral factors by tumor cells that in turn stimulate osteoclasts and lead to unchecked bone resorption and hypercalcemia. Symptoms of hypercalcemia include nausea, anorexia, abdominal pain, constipation, and mental status changes. Immediate treatment for hypercalcemia includes IV hydration
Pathological fractures – Bone metastases cause bone destruction, leading to complete or impending fracture at the site of pathology either spontaneously or with minimal trauma. The presentation is dependent on the site of the fracture; however, constant pain is a prevalent symptom. Fractures of the thoracic and lumbar spine present with pain characteristically worse with sitting or standing. Pathologic fractures result in significant morbidities resulting from pain, radiculopathy (e.g., sciatica with pelvic fracture), deformities, and immobility.
Myelophthisis – Symptomatic anemia resulting from infiltration of the bone marrow with metastatic tumor cells. Pancytopenia may also be present in late stages.
Imaging
It is pertinent to identify bone metastasis early, both types of staging and prognostication as well as the implementation of prophylactic and treatment strategies which may lead to decreased morbidity and mortality. Bone metastases can be characterized as osteolytic, sclerotic, or mixed on imaging studies.
Plain Radiograph
X-rays – or plain radiograph is the initial imaging of choice in patients presenting with bone pain. Plain films are used to assess abnormal radionuclide uptake or to detect pathological fractures. Metastatic lesions can have virtually any appearance. They can mimic a benign lesion or an aggressive primary bone tumor. It can be difficult, if not impossible, to judge the origin of the tumor from the appearance of the metastatic focus, although some appearances are fairly characteristic.
Plain radiography – best detects osteolytic lesions, but they may not be apparent until they are greater than 1 to 2 centimeters and with loss of 50% of the bone mineral content at the site of disease. Osteolytic lesions are seen as thinning of trabeculae and ill-defined margins on radiographs, while sclerotic lesions appear nodular and well-circumscribed as a result of thickened trabeculae. Plain films tend to be insensitive, especially in detecting bone metastases and with asymptomatic and subtle lesions. Progression of disease and response to therapy can be monitored with plain films and further correlation with other modalities. Sclerosis or new bone formation in osteolytic metastatic lesions is demonstrated by the sclerotic rim of reactive bone, which starts at the periphery and eventually involves the center with continued healing. Purely sclerotic lesions are more difficult to assess. Major disadvantages of plain radiographs include poor sensitivity.
Computed Tomography – Computed tomography (CT) is more sensitive (74%) than plain radiographs. It is useful in the evaluation of cortical and trabecular bone as well as in the assessment of the osteolytic and sclerotic lesions. CT scan is advantageous as it can determine staging and treatment response of other organs in addition to bone and objectively assess reactive sclerosis by calculating the change in Hounsfield units. Ribs are better evaluated with CT due to the high cortex to marrow ratio. The appearance of CT will depend on the degree of mineralization of the metastasis. The more common lytic metastases appear as regions of soft-tissue attenuation with irregular margins. The mass may breach the cortex and result in a compromise of the spinal canal. Histopathology of bone metastases is only employed for diagnostic purposes in patients of primary unknown cancers or in the presence of multiple cancers.
MRI – In the detection of bone metastasis, MRI demonstrates a sensitivity of 95% and specificity 90%. MRI is also more advantageous than a bone scan as it can detect marrow involvement before the development of osteoblastic lesions. It can be used with women who are pregnant and used to detect spinal cord compression. Bone metastases manifest as low T1 signal and high intensity on the T2 weighted sequence. Whole-body MRI requires 40 to 45 minutes to perform and involves short-tau inversion recovery (STIR) and/or T1-weighted sequences.
Nuclear Medicine – Nuclear medicine scans also are used to detect bone metastases using osteotropic radioisotopes; these include skeletal scintigraphy, SPECT, and PET scan.
Skeletal scintigraphy – or bone scan is the most commonly used radionuclide imaging which uses 99mTc-MDP employed in the detection of skeletal metastases. Radioisotopic imaging methods depict bone metastatic lesions as areas of increased tracer uptake.
Bone scan – provides the advantage of scanning the whole skeleton and has a high sensitivity (78%) therefore resulting in early diagnosis. When osteoblastic activity is prominent, the lesions are readily detected using radionuclide bone scanning. However, bone scans have low specificity for differentiating between benign and malignant bone lesions and for the detection of predominantly osteolytic lesions. Bone scans can be used to monitor the progression of disease and response to treatment.
SPECT uses 99mTc-MDP radioisotopes – uptake to detect bone lesions; however, images are acquired in cross-sectional rather than a planar fashion. SPECT has a higher specificity of 91% compared to skeletal scintigraphy.
PET – is a nuclear medicine technique that uses the radiotracers 18F FDG or 18FNaF for the detection of skeletal metastases. 18F FDG PET scan identifies bone metastases based on a high glucose metabolism exhibited by neoplastic cells. PET has a better spatial resolution compared to skeletal scintigraphy. 18F NaF-PET is proven to be substantially more sensitive and specific than bone scan and SPECT for the detection of bone metastases. Combining imaging techniques and modalities allows for improved visualization both anatomically and functionally, leading to increased diagnostic accuracy. One example of this is the 18F-Sodium fluoride (18F-NaF) PET/CT bone scanning which has significantly greater sensitivity (100%) and specificity (97%). Other hybrid imaging techniques include SPECT/CT, PET/CT, and PET/MRI.
Bone scintigraphy – is an effective means of assessing the metabolic activity of the spine, while plain radiographs can only demonstrate lesions with a loss of 30%–50% of bone mineral content[rx, rx]. Technetium‐99m (99m Tc) planar bone scintiscans detect metastatic bone deposits through increased osteoblastic activity, considered to be an indirect marker of an oncological process. For this reason, it is considered to be the most efficient modality for screening the whole body for metastasis[rx, rx]. 18F‐fluoro‐deoxy‐D‐glucose positron emission tomography (18FDG PET) offers superior spatial resolution and improved sensitivity, which is superior to bone scintigraphy in the detection of osteolytic metastases, while osteoblastic metastases show lower metabolic activity and are frequently undetectable by PET[rx, rx].
Blood tests – can aid in supporting the diagnosis of bone metastases. Complete blood count and a comprehensive metabolic panel should be obtained routinely. CBC may reveal anemia, thrombocytopenia, or pancytopenia in late stages. Serum calcium and alkaline phosphatase may be elevated due to ongoing osteolysis. Bone turnover markers are still being studied as indicators of bone resorption. Tartrate-resistant acid phosphatase has been proven to elevated in patients with breast and prostate cancer with bone metastases. Objective scoring models such as the Mirel classification system for long bones and assessment of spinal stability in addition to imaging criteria are used to determine the surgical necessity for impending pathological fractures.
Treatment of Vertebral Metastases
The therapeutic approach to bone metastases should be a multidisciplinary approach targeted at preserving the quality of life, including pain control, minimizing SREs, and achieving local tumor control. It is pertinent to consider a multitude of factors including the extent of disease spread, performance status, impending fracture, and side effects when creating the initial approach for the treatment of bone metastases. [rx][rx][rx][rx]
NSAIDs – A major aspect of treating bone metastases is analgesia/pain control for the debilitating pain that occurs with bone metastases. Pain control can be initiated with NSAIDs and titrated up to or in conjunction with narcotics as needed for symptom relief. Glucocorticoids may also be useful for additional pain control.
Osteoclast inhibitors – (bisphosphonates and denosumab) decrease morbidity and mortality associated with bone metastases as they reduce skeletal-related events and can be used for analgesia to some extent.
Local radiation for symptomatic bone metastases – is a significant component of the palliative approach in providing analgesia. It is also used postoperatively to consolidate continued bone healing. External beam radiation is the standard approach for painful bone metastases and is beneficial in reducing pain by up to 50% to 80%. Several studies have proven that a single 8 Gy fraction compared to more prolonged or fractionated radiation is non-inferior; however, it may carry a higher need for pretreatment (20 % vs. 8%). Stereotactic body radiation therapy spares the normal tissue while delivering highly conformal radiation to the affected area. There are no clear guidelines on stereotactic body radiotherapy (SBRT) use however it may be indicated over external beam radiation therapy (EBRT) in specific instances of bone metastases specifically vertebral from certain radio-resistant neoplasms.
Bone-targeted radiopharmaceutical therapy – (e.g., beta-emitting agents strontium-89, alpha-emitting radium-223) provides the specific advantage of treatment of diffuse pain associated with osteoblastic bone metastases. It is typically used in bowel movement associated with prostate and breast cancer or for analgesia in radiation therapy for refractory pain. Systemic chemotherapy when amenable, aimed at the primary tumor can also provide analgesia by reduction of tumor size and control of tumor spread.
Surgery is indicated – for impending or complete fracture, mechanical stability, and spinal cord compression. In cases where the spread of the primary cancer is limited to a single bone lesion, en bloc resection of the metastasis can be done by means of local tumor control.
Local ablation via radiofrequency ablation (RFA), cryoablation, and focused ultrasound (FUS) should be considered for patients with persistent pain following radiation therapy or patients with recurrent pain
National Institute for Health and Care Excellence clinical guideline for the management of spinal cord compression
In November 2018, NICE issued a clinical guideline for the diagnosis and management of adults at risk of or with MSCC.[rx]The guidelines contained treatment algorithms for patients with symptoms suggestive of spinal metastases. The guideline proposed the patient treatment pathways shown in. Treatment of patients with spinal metastases and MSCC can be broadly divided into three pathways.[rx]
Treatment of patients with spinal metastases and prevention of metastatic spinal cord compression
Patients with painful spinal metastases should be offered conventional analgesics, i.e. non-steroidal anti-inflammatory drugs. Those patients with intractable pain should be considered for specialist pain care that includes invasive procedures and neurosurgical interventions. Patients with spinal metastases from breast and prostate cancer should be offered bisphosphonates to alleviate pain and reduce the risk of pathological fracture/collapse of the spine. Those patients with non-mechanical spinal pain should be given single-fraction palliative radiotherapy. This should also be considered in those who are completely paralyzed. In asymptomatic patients, radiotherapy should not be administered.
Two vertebral augmentation techniques, vertebroplasty, and kyphoplasty should be considered in those with mechanical spinal pain resistant to conventional analgesics and no evidence of MSCC or spinal instability.
Surgery should be preferred when there is evidence of progressive disease mainly to prevent MSCC. It should also be considered in those with spinal metastases and mechanical pain resistant to conventional analgesics and in those with evidence of spinal instability.
Treatment of threatened spinal cord in patients with metastatic spinal cord compression
In patients with severe mechanical pain suggestive of spinal instability or those with neurological symptoms or signs suggestive of MSCC, the spine should be stabilized. Patients should be monitored regularly, especially during sitting from supine to 60 degrees. If patients continue to deteriorate, then they should revert back to the lying position or to the position in which there is minimal pain/neurological symptoms. In those patients not suitable for definitive treatment, the aim of the treatment should be to help the patient to achieve a comfortable position and mobilization. This is usually achieved by using orthoses.
Corticosteroids should be given to all patients with MSCC unless contraindicated. Dexamethasone at 16 mg as a loading dose should be given followed by a short course of 16 mg dexamethasone daily until definitive treatment is employed. After definitive treatment, the dose of dexamethasone should be reduced gradually over 5–7 days and then stopped. In those patients in whom symptoms have deteriorated, the dose of dexamethasone can be increased temporarily.
Definitive treatment of metastatic spinal cord compression
The definitive treatment should be given as early as possible, ideally within 24 hours of the diagnosis of MSCC. Before this, a diagnosis of the primary location of the tumor should be made. In addition, an attempt should be made to study the extent of the disease. A scoring system such as the Tokuhashi scoring system and the American Society of Anesthesiologists grading for the overall patient condition should be used to assess whether surgery is appropriate.
Radiotherapy
Patients unsuitable for surgery should receive radiotherapy within 24 hours, 7 days a week. Fractionated radiotherapy is the definitive treatment of choice for patients with epidural tumor without neurological dysfunction, mechanical pain or spinal instability. It is also an appropriate first-line treatment for patients with a good prognosis. Radiotherapy should not be given to patients with MSCC who are waiting for surgery but fractionated radiotherapy should be offered to all patients postoperatively once their wound has healed.
Supportive care and rehabilitation
Supportive care includes thromboprophylaxis, management of pressure ulcers, bladder and bowel continence, circulatory and respiratory functions, and access to specialist rehabilitation care at home.[rx]
Radiation
The aim of radiation is to alleviate pain and to prevent recurrence and tumor growth.[rx] It is indicated when the spine is stable, if the tumor is radiosensitive and the patient’s neurological condition is stable, or if the patient is in poor medical condition or has a life expectancy < 3–6 months and has had complete paraplegia for > 24 hours.[rx]
Recently, new approaches, such as intensity-modulated radiotherapy (IMRT),[rx] or stereotactic body radiotherapy, have been suggested for the treatment of vertebral metastases.[rx]
Systemic therapies
Corticosteroids
Intravenous or oral corticosteroids have been found to provide improvement or resolution of neurological symptoms and pain in patients with epidural spinal metastases.[rx] It should be noted that there is no standard dosage regimen for corticosteroids. They are often used before surgery.[rx]
In patients with MSCC undergoing surgical decompression, corticosteroids are often used in combination with radiotherapy.[rx]
Bisphosphonates and denosumab
Bisphosphonates are known to impair osteoclastic activity and so they reduce tumor-related resorption of bone.[rx,rx] Currently, bisphosphonates are used to alleviate metastatic bone pain and to reduce SREs such as pathological fractures, hypercalcemia, and MSCC. Bisphosphonates are also used to reduce the frequency of surgery and radiation therapy.[rx,rx] Bisphosphonates such as pamidronate (Aredia®; Novartis Pharmaceuticals Corporation), clodronate (Bonefos®, Clasteon®, Loron®; Bayer), ibandronate (Bondronat®; F. Hoffmann-La Roche Ltd), alendronate (Fosamax®; Merck Sharp & Dohme Corporation) and zoledronate (Aclasta®; Novartis) have all been found to be effective in the treatment of hypercalcemia.[rx] Although radiotherapy is the main treatment for reducing bone pain, bisphosphonates can be used as an alternative therapy, which in turn will considerably reduce the frequency of radiotherapy.[rx,rx] The effect of bisphosphonates on pain is not dependent on the nature or type of the tumor (i.e. sclerotic or lytic).[rx] The efficacy of these drugs has been seen in breast cancer, multiple myeloma, and other osteolytic metastases.57 Although bisphosphonates have been found to be effective in preventing skeletal-related complications, they are not so effective in reducing pain in patients with prostate cancer.[rx]
Recently, monoclonal antibody therapy with denosumab, a specific inhibitor of RANKL, has been found to be effective in delaying and preventing SREs.[rx]
Chemotherapy
The benefits of chemotherapy are limited in spinal metastases, as patients are usually at a late stage of disease.[rx] Chemotherapy can be given on its own or in combination with surgery and hormonal therapy.[rx]
Radioisotopes
Radioisotopes are administered systematically and act as local radiation therapy to the spine.[rx] Radioisotopes include strontium-89 and rhenium-186. Although radioisotopes are found to reduce pain in patients with spinal metastases, these can cause irreversible bone marrow suppression and, for this reason, they are recommended for use in those with good marrow function and in whom no other treatment is available.[rx]
Hormonal therapies
Hormonal therapies are a major treatment modality for metastatic breast and prostate cancer. As an example, a new drug, abiraterone (Zytiga®; Janssen), has recently been developed which has improved outcomes in men with metastatic castration-resistant prostate cancer.[rx,rx]
Surgery
The main aims of surgery are to remove the tumor, achieve spinal stability, and reconstruct the vertebral column.7 Surgery may also help with diagnosing the origin of the tumor and in relieving neurological symptoms.7 In those with solitary renal cell carcinoma metastases, surgery can increase disease-free survival.[rx] Current indications for surgery are (1) radioresistant tumor such as renal or colon carcinoma, (2) evidence of neurological function deterioration or tumor progression despite radiotherapy, (3) radiological images showing fragments of bone in the spinal canal, (4) spine instability due to fracture and causing pain and neurological deficit, (5) neurological deficit for > 24 hours, or significant MSCC, and (6) life expectancy of at least 3 months.[rx,rx]
Surgery should be considered only if it would increase the patient’s survival by > 3 months. The aim of this treatment is to decompress the spinal cord and stabilize the spine. Posterior decompression alone should be used only in cases of isolated epidural tumor or neural arch metastases without bony instability. In those in whom metastasis involves the vertebral body and who are therefore at increased risk of spinal instability, posterior decompression by internal fixation, with or without bone grafting, should be carried out. Reconstruction of the vertebral body should be carried out in patients with MSCC and vertebral body involvement who are expected to survive < 1 year, whereas in those expected to survive > 1 year, reconstruction of the vertebral body with anterior bone graft should be undertaken. In rare circumstances such as solitary renal or thyroid metastasis following complete staging, en bloc excisional surgery should be carried out.
Vertebral augmentation
Two techniques, percutaneous vertebroplasty and kyphoplasty, initially developed for the treatment of painful vertebral haemangiomas, are now used effectively in treating painful pathological fractures caused by metastatic spinal disease.[rx] Vertebroplasty involves an injection of polymethylmethacrylate (PMMA) into the compression fracture whereas in kyphoplasty an inflatable balloon is placed in the vertebral body and PMMA is injected.[rx,rx] Although these interventions can lead to significant pain reduction and greater mobility,[rx,rx] are contraindicated in SCC because of pathological fractures as they do not relieve cord compression.[rx] Complications of these techniques include leakage of PMMA, misplacement of PMMA, and hematogenous embolization of PMMA to the lungs.[rx]
Non‐operative Measures
Oral analgesia as titrated by the World Health Organization Analgesic Ladder is considered the first line in the treatment of bony pain. Morphine is the most commonly used opioid for moderate to severe pain and may be combined with adjuvant medications such as tricyclic antidepressants and corticosteroids. Side effects of medications can limit opioid dosage and cause significant morbidity. These include delirium, constipation, pruritus, nausea, vomiting, sedation, myoclonus, and respiratory depression[rx], [rx], [rx]. Other non‐invasive methods of pain relief include cutaneous stimulation, continuous repositioning, spine cryoablation, and regional nerve blocks[rx], [rx], [rxx].
Monthly infusions of bisphosphonates like zoledronic acid to patients with bone metastases reduce the frequency and delays the onset of skeletal‐related events[rx]. Their administration also provides significant improvements in bone pain and quality of life[rx]. Bisphosphonate use in patients with spinal metastases has increased in the past decade. Their use has decreased bone pain scores and reduced skeletal‐related events such as the need for local radiotherapy, hypocalcemia, pathologic fracture, and spinal cord compression[rx, rx]. Once injected, bisphosphonates are internalized by osteoclasts. This leads to a decrease in osteoclast activity and viability[rx]. Complications of bisphosphonate therapy include renal impairment and mandibular osteonecrosis[rx, rx]. Bisphosphonate demonstrates maximal effectiveness and safety when combined with either single or multiple fraction radiotherapy[rx, rx]. This synergism is due to the fact that radiotherapy is believed to reduce numbers of tumor‐produced osteoclast activating factors[rx]. The American Society of Clinical Oncology guidelines and the International Expert Panel guidelines recommend starting bisphosphonates when the first radiographic indication of metastatic deposits in the spine is noted[rx, rx].
Denosumab, a fully human monoclonal antibody to RANK‐L, has demonstrated in clinical trials inhibition of osteoclast‐mediated bone destruction in breast, prostate, and myeloma tumors, and is considered non‐inferior to zoledronic acid[rx], [rx]. A meta‐analysis performed by Lipton et al. compared the efficacy of denosumab to zoledronic acid in preventing skeletal‐related events in people with prostate cancer, breast cancer, solid tumors, or multiple myeloma. Denosumab increased the time to the first on‐study skeletal‐related event by 8.21 months and reduced the risk of a first skeletal‐related event by 17%[rx].
Chemotherapy is very seldom considered in the targeted treatment of metastatic spinal tumors due to its systemic nature and also owing to the fact that it requires an extended course of administration prior to pain relief[rx]. Complications are the source of morbidity and fear for the patient and include pain, gastrointestinal abnormalities, hematological disturbances, immunosuppression, and biopsychosocial sequelae (alopecia and infertility)[rx].
Pain caused by bone metastases has multiple causes, including periosteal elevation and inflammation[rx]. Radiopharmaceuticals may be used in the palliation of bony pain. Ethylenediamine tetramethylene phosphonic acid is an IV radioisotope that preferentially binds to osteoplastic metastases and osteosarcomas, and a significant analgesic effect may be achieved in 83%–93% of patients[rx]. Strontium‐89 chloride infusions have also been trialed as a similar treatment. Response rates vary in the published literature from minimal to up to 77%[rx]. Corticosteroids produce effects that include mood elevation, an anti‐inflammatory effect, and reduction of spinal cord edema in bony metastases[rx]. There is good evidence supporting the use of high-dose dexamethasone (64 mg/day) in the treatment of pain from spinal metastases, particularly if epidural compression is present. It is associated with significant pain relief and the ability to remain ambulatory in up to 81% of patients[rx].
Psychiatry
Involvement of psychiatric services may be necessary in patients with a diagnosis of metastatic spinal disease. Psychological complications in this instance usually manifest as anxiety, depression, adjustment disorder, and loss of self‐esteem[rx, rx]. Studies have shown that up to 50% of such patients may experience psychological issues following such a diagnosis[rx, rx]. Early involvement and assessment of this cohort of patients by psychiatric services is essential in the multidisciplinary management and treatment of spinal metastases.
Nutrition
Nutritional support is another consideration in the approach to metastatic spinal disease[rx]. The goals of nutrition support include preventing or reversing nutrient deficiencies, maximizing quality of life, aiding immunologic function, and preserving lean body mass.
This cohort is at risk of anorexia and cachexia[rx]. Important considerations include dysphagia after radiation, oesophagitis, and reduced motility owing to pain medications[rx]. Anorexia has been noted to be an almost universal side effect in individuals with widely metastatic disease[rx]. The multidisciplinary team must also consider decreased caloric intake as a result of diminished appetite and malaise and tumor competition for nutrients. Constipation may be secondary to opiate analgesia or spinal cord involvement by tumors causing an upper motor neuron lesion. Malnutrition may also exacerbate this. The addition of dietary calcium and vitamin D for bone health in the patients at risk of therapy‐associated fractures is warranted.
Screening and nutrition assessment should be interdisciplinary. Physicians, nurses, dietitians, and social workers (as members of the health‐care team) should all participate in nutritional management throughout the continuum of the management of the metastatic spinal disease. Such screening tests include the prognostic nutrition index[rx].
Suggestions for appetite improvement include keeping a daily menu, snacking between meals, eating small and frequent meals, and adding extra protein to meals[rx, rx]. Progestational agents such as megestrol acetate and medroxyprogesterone can lead to appetite stimulation and subsequent weight gain[rx]. The preferred method of nutritional support is via the oral route. If the GI tract is rendered dysfunctional, TPN may be indicated[rx].
Physiotherapy
Physiotherapists play a central role in the multidisciplinary approach to spinal metastasis. Their role is to maximize the quality of life by maintaining patient mobility and facilitating their capacity to perform activities of daily living[rx, rx]. Pain reduction therapies may also be employed, such as hot/cold packs, massage, and electrical stimulation. Assistive devices or orthotics, such as frames, canes, and thoracolumbosacral orthosis (TLSO), are provided by the physiotherapy department to patients with spinal metastasis when required[rx].
Giant cell tumor is a relatively common, locally aggressive, potential behavior, capacity to metastasize and benign neoplasm that is associated with a large biological spectrum ranging from latent benign to highly recurrent and occasionally metastatic malignant potential [rx]. It is one of the most common benign bone tumors, occurring in young adults ages 20–40 years with a high recurrence rate and a potential for aggressive behavior. It is most commonly located in the metaphysis or at the epiphysis of the femur or tibia. This activity reviews the evaluation and management of giant cell tumors and highlights the role of the interprofessional team in improving care for the patients with this condition.
A giant cell tumor is a rare, aggressive non-cancerous tumor. It usually develops near a joint at the end of the bone. Most occur in the long bones of the legs and arms. Giant cell tumors most often occur in young adults when skeletal bone growth is complete.
Giant cell tumor (GCT) is one of the most common benign bone tumors, which occurs in young adults 20-40 years old with a high recurrence rate and a potential for aggressive behavior.[rx] It is most commonly located at the metaphyseal or epiphyseal portion of the tibia or femur. While overall GCT has a benign characteristic, the disease behavior spectrum is extremely unpredictable. Local aggressiveness can range from focal symptoms due to bony or cortical destruction, up to surrounding soft tissue expansion and metastasis. An occurrence within the axial skeleton can lead to severe local complications and is usually unresectable.[rx]
The biopsied tissue have multinucleated giant cells under the microscope. They consist of three different cell types:
giant cell tumor stromal cells of osteoblastic origin
mononuclear histiocytic cells
the multinucleated giant cell of an osteoclast-monocyte lineage.[rx]
The tumor bone resorption is primarily done by the giant cells. The spindle-like stromal cells recruit monocytes and promote their fusion into giant cells. The stromal cells also improve the resorptive ability of the giant cells.
Pathophysiology
The receptor activator of nuclear factor kappa B [NF-kB] ligand (RANKL) appears to be critical to the pathogenesis of GCT. Under normal physiologic conditions, osteoclast formation requires interaction with cells of the osteoblastic lineage, which may depend upon cell-cell contact, and the interaction of RANKL with its receptor RANK.[14] This receptor is highly expressed on monocytes, while RANKL is expressed by a variety of cell types, including stromal cells and lymphocytes. A variety of co-regulatory molecules also take part in osteoclast formation, including monocyte-colony-stimulating factor, vitamin D, parathyroid hormone and parathyroid hormone-related protein, and prostaglandins.[rx]
Several studies identified RANKL as highly expressed by the stromal cells within a GCT.[rx][rx] The stromal cells also secrete factors that can regulate or prevent osteoclastogenesis, including osteoprotegerin, which blocks osteoclast/osteoblast interactions and functions as a natural negative regulator of RANKL.[rx] The expression of RANKL by the osteoblast-like mononuclear stromal cells stimulates the recruitment of the osteoclastic cells from a normal monocytic pre-osteoclast cell. The osteoclastic giant cells then actively absorb host bone via a cathepsin K and matrix metalloproteinase 13-mediated process, which would account for the osteolysis associated with these tumors.[rx]
Mutations in the H3F3A gene, present in over 90% of GCT, may drive tumorigenesis. These mutations are restricted to the stromal cell population and are not detected in the osteoclasts or their precursors.[rx] The neoplastic stromal cell likely owns an immature osteoblast phenotype, part of whose transcriptional repertoire includes RANKL, besides other markers of the early osteoblast lineage.[rx] It is also postulated that the stromal cells have become activated not because of some inherent genetic change but instead from a local hemorrhage-induced release of red cells and plasma proteins into the matrix. Unknown reciprocal giant cell signals may be involved in maintaining the stromal cells’ immature state. RANKL has been identified as a primary molecular target for therapy.[rx]
Causes of Giant Cell Tumor
The exact etiology of GCT is not fully understood yet. It remains uncertain whether it is a true neoplasm or just a reactive condition. A 20q11 amplification is seen in 54% of GCTs, over-expression of p53 in 20% of them. Centrosome amplification, and boosted telomerase activity with the prevention of telomeres shortening support a neoplastic etiology.[rx][rx][rx]
Most often, the tumors occur close to the knee joint—either in the lower end of the thighbone (femur) or the upper end of the shinbone (tibia).
Other common locations include the:
Wrist (lower end of the lower arm bone)
Hip (upper end of the thighbone)
Shoulder (upper end of the upper arm bone)
Lower back (connection of the spine and pelvis)
Most giant cell tumors occur in patients between 20 and 40 years of age. Only rarely do they occur in children or in adults older than 65 years of age. They occur slightly more often in females.
Symptoms of Giant Cell Tumor
The following are the most common symptoms of a giant cell tumor. However, each person may experience symptoms differently. Symptoms may include:
A visible mass
Bone fracture
Fluid buildup in the joint nearest the affected bone
Limited movement in the nearest joint
Swelling
Pain at the nearest joint
The symptoms of a giant cell tumor may look like other medical problems. Always talk with your healthcare provider for a diagnosis.
Diagnosis of Giant Cell Tumor
On gross inspection, these lesions are characteristically chocolate brown, soft, spongy, and fragile.[rx] Yellow-to-orange discoloration from the hemosiderin could also be present. Cystic blood-filled cavities within the tumor are common.[rx] Examination reveals a variable degree of cortical expansion and disruption with an intact periosteum.[rx]
Histologically, the lesions are cellular. Although the multinucleated giant cell is the characteristic cell type, these lesions have a background network of mononuclear stromal cells.[rx] The mononuclear cells could be plump, oval, or spindle-shaped. They could have prominent mitotic activity, but cellular atypia is rare. Multinucleate giant cells have numerous centrally located nuclei in opposition to the peripherally located nuclei of Langerhans-type giant cells seen in atypical infections. The nuclei are compact and oval and contain prominent nucleoli. Giant cells are distributed throughout the lesion. The concentration of multinucleated giant cells differs from tumor to tumor. Some tumors have numerous multinucleated giant cells, whereas others have a small number of giant cells settled in whirls of spindle-shaped stromal cells. In approximately 5% of the cases, giant cells invade the small perforating vessels.
Three types of cells are found in benign bone GCT:[rx]
Type I cells look like interstitial fibroblasts, make collagen, and can proliferate. This cell is probably the tumor component of GCT. Type I cells share features of mesenchymal stem cells. They have characteristics that suggest they could represent an early differentiation into osteoblasts.[rx]
Type II cells are also interstitial but resemble the monocyte/macrophage family and could be recruited from the peripheral bloodstream.[rx] These cells are precursors of the multinucleated giant cells.
Type III cells are the multinucleated giant cells. They share many characteristics of osteoclasts and have similar morphologies.[rx] They own enzymes for bone resorption, including tartrate-resistant acid phosphatase and type II carbonic anhydrase.[rx][rx]
Significant level activity for insulin-like growth factors I and II are found in type II and type III cells but absent in type I cells, which suggests that these factors are essential in the development and regulation of GCT.[rx]
Genetically, 80% of individuals with GCT of the bone exhibit the cytogenetic abnormality of telomeric associations (tas), whereas half of the cells in the tumor show the tas abnormality.[rx] The RANK pathway is often reported to be involved in the pathogenesis of GCT. This pathway is a crucial signaling pathway of bone remodeling that plays a critical role in the differentiation of precursors into multinucleated osteoclasts and activation of osteoclasts leading to bone resorption.[rx]
The most common history and physical findings include
Pain is the most common presenting symptom secondary to the mechanical insufficiency resulting from bone destruction.
Swelling and deformity – are associated with more extensive lesions.
A soft tissue mass or bump – can occasionally be seen and results from the cortical destruction and tumor progression outside the bone. It is often found close to the joint. Thus, a limited range of motion at the joint area is common.
Joint effusion and synovitis – are also possible. At the time of diagnosis, approximately 12% of patients present with pathological fractures.[rx][rx] The pathologic fracture incidence at presentation is 11-37%. Presentation with a pathologic fracture is thought to show a more aggressive disease with a higher risk of local recurrence and metastatic spread.[rx][rx]
The typical epiphyseal location – is found in 90% of the tumors. The tumor often extends to the articular subchondral bone or even abuts the cartilage. It rarely invades the joint and or its capsule. In those rare instances in which GCT occurs in a skeletally immature patient, the lesion is likely to be found in the metaphysis.[rx][rx] Only 1.2% of GCT involved metaphysis or diaphysis without epiphyseal involvement.
The most common locations – for the tumor in descending order are the distal femur, the proximal tibia, the distal radius, and the sacrum.[rx] Fifty percent of GCT arise around the knee region. Other sites include the proximal femur, the fibular head, and the proximal humerus. A pelvic bone tumor is somewhat rare.[rx]
Multicentricity – or the simultaneous occurrence of GCT in different sites occurs but is exceedingly rare.[rx][rx] Most commonly, GCT is a solitary lesion. Multicentric involvement (less than 1%) is much more clinically aggressive and has a propensity for the small bones of the hands and feet, which is totally different from the solitary lesions. Patients with multicentric lesions are generally younger than those with lesions elsewhere.
Imaging
The workup includes:
X-ray – reveals a characteristic radiolucent, geographic appearance with a narrow transition zone found at the lesion margin. This margin, as opposed to that of many other benign lesions, lacks a prominent sclerotic rim. Calcification of matrix, periosteal reaction, and new bone formation are typically absent.[rx] It is an eccentric lesion in the epiphyseal portion with a tendency to extend up to a centimeter of the subchondral bone.
Imaging modalities such as computed tomographic (CT) – scan and magnetic resonance imaging (MRI) may be helpful to confirm the typical subchondral location of these lesions within the bone and the extent of a soft tissue mass, either beyond the bone cortex or through the adjacent joint.[rx][rx]
Positron emission tomography – Modern imaging modalities to determine the extent of disease involvement include MRI, CT scan, functional positron emission tomography (PET), and bone scans.
CT scans – give a more accurate assessment of cortical thinning, penetration, and bone mineralization than the plain radiographs. The presence of a primary bone formation within the tumor suggests primary osteosarcoma. CT scan chest may be indicated to look for pulmonary metastasis. The metastatic spread is most common in the setting of a local recurrence; therefore, a chest CT is recommended in patients with locally recurrent disease.
MRI scan – helps assess the surrounding soft tissue integrity, such as the neurovascular structures or the extent of subchondral extension into adjacent joints. In the typical GCT, there is homogeneous signal intensity, and the lesion is well-circumscribed. They present with low signal intensity on T1-weighted images and intermediate signal intensity on T2-weighted images. Expansile hypervascular mass with cystic changes and heterogeneous low to intermediate signal intensity on T1-weighted images and intermediate to high intensity on T2-weighted images are its characteristic findings on MRI.[rx][rx] Huge amounts of hemosiderin account for the areas of low signal intensity on both T1 and T2-weighted images.[rx]
A bone scan – can help stage multicentric disease, but findings of the bone scan, typically a decrease in the uptake of radiotracer in the tumor’s center, are not specific for GCT. Aneurysmal bone cysts have a similar appearance. There are limited data about the use of fluorine-18 fluorodeoxyglucose (FDG)-PET for newly diagnosed GCT. GCT accumulates the FDG tracer, unlike many benign bone tumors, presumably because of the active metabolism of osteoclast-like giant cells.[rx][rx] However, the advantages of evaluation with FDG PET as compared to conventional imaging with CT, MRI, or a bone scan are still unclear. Changes in FDG uptake over time correlate with the metabolism of the tumor and its angiogenic activity.[rx]
Treatment of Giant Cell Tumor
Nonsurgical Treatment
Nonsurgical treatment may include:
Radiation. Radiation therapy may sometimes be used to shrink giant cell tumors in areas where surgery may be difficult to perform without damaging sensitive tissues—such as the spine. However, radiation therapy can result in the formation of cancer in some patients, so it is used only in the most difficult cases.
Tumor embolization. During this procedure, specific arteries that supply blood to the tumor are blocked off. Without their supply of oxygen and nutrients, the tumor cells begin to die. Most often, embolization is performed prior to surgery, but it may also be used on its own in cases where surgery cannot be performed.
Medication. The FDA has recently approved the use of an injectable medication for the treatment of giant cell tumors. The medication works by targeting a special receptor on the tumor cells. This decreases activity and slows down the breakdown of bone. The medication is sometimes used in cases where surgery cannot be performed or for recurrent tumors.
Surgical Resection
It is the standard of care for the treatment of GCT. As most GCTs are benign and near a joint in young adults, several authors favor an intra-lesional approach that preserves the anatomy of bone instead of resection.[rx][rx][rx]
Wide resection is correlated with a decreased local recurrence risk when compared with an intra-lesional curettage and could increase the recurrence-free survival rate from about 84% to 100%.[rx][rx][rx] However, wide resection is associated with higher rates of surgical complications and leads to functional impairment requiring reconstruction.[rx][rx][rx]
Resection may be the preferred option even in benign tumors when the bone salvageability by intralesional methods would cause a severe compromise in mechanical characteristics. In the so-called “expendable bones,” such as the lower ulnar end, upper fibular end, excision may be the treatment of choice.
Either in primary or recurrent cases, as the tumor involves the end of a long bone and causes significant dysfunction of the joint surface, reconstruction of the joint surface is necessary. A mega prosthetic joint replacement, a biologic reconstruction with an autograft arthrodesis with internal/external fixation, a microvascular fibula reconstruction, an Ilizarov method of bone regeneration, and an osteoarticular allograft are the options for those cases.[rx][rx]
In the past GCT – was treated with amputations, wide resections, or reconstructions. But having in mind that GCT is a benign yet locally-aggressive tumor, a local intralesional surgical approach is appropriate in most cases. Curettage, curettage and bone grafting, curettage and insertion of polymethylmethacrylate (PMMA), and primary resection are among the recommended treatment options. Radiation therapy and embolization of the feeding vessels are used for pelvic and sacral tumors, which are not amenable to surgery.[rx]
Radiotherapy – is also recommended for the spinal, sacral, or aggressive tumors when complete excision or curettage is impractical for any functional or medical reasons. Intralesional curettage and bone grafting are the limb-sparing treatment of choice, which is associated with acceptable functional and oncologic outcomes. However, a simple curettage with or without a bone graft has a recurrence rate between 27 to 55%. Many surgeons choose to replace bone graft packing of the lesion with PMMA packing due to the high recurrence rate.
Wide en-bloc resection is another option that offers the lowest recurrence rate and can be used in an expendable bone. In the proximal fibula, a wide resection without reconstruction is often performed. GCT of the distal radius is usually resected and reconstructed with an allograft or an autograft.
Adjuvant treatments (liquid nitrogen, phenol, or HO and argon beam coagulation) offers an excellent recurrence-free survival, especially when paired with an intralesional curettage. A successful treatment for GCT heavily relies on the aggressiveness of the intralesional curettage than on the specific adjuvant used. The adequacy of tumor removal is influenced by the tumor location, associated fractures, extensions to the soft tissue, and an understanding of the functional consequences of the resection.
Topical or systemic bisphosphonates like zoledronate or pamidronate can be used as a novel adjuvant therapy for GCT. Bisphosphonates induce apoptosis and limit the tumor progression by targeting the osteoclast-like giant cells.[50][51]
Denosumab, a monoclonal antibody, is widely used to treat unresectable GCTs of bone in adults and skeletally-matured adolescents, and acts by specifically binding to RANKL.
There is no recognized effective chemotherapeutic agent available for the management of these tumors yet.
Key points about giant cell tumors
A giant cell tumor is a rare, aggressive non-cancerous tumor. It usually develops near a joint at the end of the bone. Most occur in the long bones of the legs and arms.
Giant cell tumors most often occur in young adults when skeletal bone growth is complete.
The exact cause of giant cell tumors remains unknown.
Symptoms may include joint pain, swelling, and limited movement.
Diagnostic tests may include X-rays, biopsy, and bone scans.
The goal for treatment of a giant cell tumor is to remove the tumor and prevent damage to the affected bone.
Tumors that can’t be removed surgically can often be controlled and sometimes destroyed with radiation therapy.
Giant cell tumors can come back.
Next steps
Tips to help you get the most from a visit to your healthcare provider:
Know the reason for your visit and what you want to happen.
Before your visit, write down questions you want answered.
Bring someone with you to help you ask questions and remember what your healthcare provider tells you.
At the visit, write down the name of a new diagnosis, and any new medicines, treatments, or tests. Also write down any new instructions your provider gives you.
Know why a new medicine or treatment is prescribed, and how it will help you. Also know what the side effects are.
Ask if your condition can be treated in other ways.
Know why a test or procedure is recommended and what the results could mean.
Know what to expect if you do not take the medicine or have the test or procedure.
If you have a follow-up appointment, write down the date, time, and purpose for that visit.
Know how you can contact your healthcare provider if you have questions.
A spinal tumor is an abnormal mass of tissue within or surrounding the spinal cord and/or spinal column. These cells grow and multiply uncontrollably, seemingly unchecked by the mechanisms that control normal cells. Spinal tumors can be benign (non-cancerous) or malignant (cancerous). Primary tumors originate in the spine or spinal cord, and metastatic or secondary tumors result from cancer spreading from another site to the spine.
Spine tumors may arise from any of the structures of the spine or the spinal column. They may arise in the cervical (neck), thoracic (mid-back), or lumbosacral (low back) regions. They may originate in the spinal cord itself, the spinal roots, the dural sac which surrounds the spinal cord, or the vertebrae (bones). They may be primary—originating from the spine or spinal cord—or metastatic, originating elsewhere (ie, lung, breast, etc).
Tumors of the brain and spinal cord are abnormal growths of tissue found inside the skull or the bony spinal column. The brain and spinal cord are the primary components of the central nervous system (CNS). Benign tumors are noncancerous, and malignant tumors are cancerous. The CNS is housed within rigid, bony quarters (i.e., the skull and spinal column), so any abnormal growth, whether benign or malignant, can place pressure on sensitive tissues and impair function. Tumors that originate in the brain or spinal cord are called primary tumors. Most primary tumors are caused by out-of-control growth among cells that surround and support neurons, specific genetic diseases (such as neurofibromatosis type 1 and tuberous sclerosis), or exposure to radiation or cancer-causing chemicals. Metastatic, or secondary, tumors in the CNS are caused by cancer cells that break away from a primary tumor located in another region of the body. Tumors can place pressure on sensitive tissues and impair function. Symptoms of brain tumors include headaches, seizures, nausea and vomiting, poor vision or hearing, changes in behavior, unclear thinking, and unsteadiness. Spinal cord tumor symptoms include pain, numbness, and paralysis. Diagnosis is made after a neurological examination, special imaging techniques (computed tomography, and magnetic resonance imaging, positron emission tomography), laboratory tests, and a biopsy (in which a sample of tissue is taken from a suspected tumor and examined).
Types of Spinal Tumor
By the region of the spine in which they occur. These basic areas are cervical, thoracic, lumbar, and sacrum. By their location within the spine.
Intradural-extramedullary – The tumor is located inside the thin covering of the spinal cord (the dura), but outside the actual spinal cord. The frequency of occurrence in this location is 40%. The most common of these types of tumors develop in the spinal cord’s arachnoid membrane (meningiomas), in the nerve roots that extend out from the spinal cord (schwannomas and neurofibromas), or at the spinal cord base (filum terminale ependymomas). Although meningiomas are often benign, they can be difficult to remove and may recur. Nerve root tumors are also generally benign, although neurofibromas may become malignant over time. Ependymomas at the end of the spinal cord can be large, and the delicate nature of fine neural structures in that area may make removal difficult.
Intramedullary – These tumors grow inside the spinal cord. They typically derive from glial or ependymal cells (a type of glial cell) that are found throughout the interstitium of the spinal cord. The frequency of occurrence in this location is approximately 5%. Astrocytomas and ependymomas are the two most common types. Astrocytomas are more common in the thoracic region followed by the cervical. Ependymomas are most common in the filum (bottom region of the spinal cord), followed by the cervical region. They are often benign (compared to intracranial), but can be difficult to remove. Intramedullary lipomas are rare congenital tumors most commonly located in the cervicothoracic spinal cord.
Extradural – The tumor is located outside the dura, which is the thin covering surrounding the spinal cord. The frequency of occurrence in this location vs the ones above is approximately 55%. These lesions are typically attributed to metastatic cancer or less commonly schwannomas derived from the cells covering the nerve roots. Occasionally, an extradural tumor extends through the intervertebral foramina, lying partially within and partially outside of the spinal canal.
The bony spinal column is the most common site for bone metastasis. Estimates indicate that at least 30% and as high as 70% of patients with cancer will experience the spread of cancer to their spine. The most common primary spine tumor (originated in the bony spine) is vertebral hemangiomas. These are benign lesions and rarely cause symptoms such as pain.
Common primary cancers that spread to the spine are lung, breast, and prostate. Lung cancer is the most common cancer to metastasize to the bone in men, and breast cancer is the most common in women. Other cancers that spread to the spine include multiple myeloma, lymphoma, melanoma, and sarcoma, as well as cancers of the gastrointestinal tract, kidney, and thyroid. Prompt diagnosis and identification of the primary malignancy are crucial to overall treatment. Numerous factors can affect outcomes, including the nature of primary cancer, the number of lesions, the presence of distant non-skeletal metastases, and the presence and/or severity of spinal cord compression.
Symptoms
Non-mechanical back pain, especially in the middle or lower back, is the most frequent symptom of both benign and malignant spinal tumors. This back pain is not specifically attributed to injury, stress or physical activity. However, the pain may increase with activity and can be worse at night when lying down. Pain may spread beyond the back to the hips, legs, feet or arms and may worsen over time — even when treated by conservative, nonsurgical methods that can often help alleviate back pain attributed to mechanical causes. Depending on the location and type of tumor, other signs and symptoms can develop, especially as a tumor grows and compresses on the spinal cord, the nerve roots, blood vessels or bones of the spine.
Additional symptoms can include the following:
Loss of sensation or muscle weakness in the legs, arms, or chest
Stiff neck or back
Pain and/or neurologic symptoms (such as tingling) increase with the Valsalva maneuver
Difficulty walking, which may cause falls
Decreased sensitivity to pain, heat, and cold
Loss of bowel or bladder function
Paralysis may occur in varying degrees and in different parts of the body, depending on which nerves are compressed
Scoliosis or other spinal deformity resulting from a large and/or destructive tumor
Testing & Diagnosis
A thorough medical examination with emphasis on back pain and neurological deficits is the first step to diagnosing a spinal tumor. Radiological tests are required for an accurate and positive diagnosis.
X-ray – Application of radiation to produce a film or picture of a part of the body can show the structure of the vertebrae and the outline of the joints. X-rays of the spine are obtained to search for other potential causes of pain, i.e. tumors, infections, fractures, etc. X-rays, however, are not very reliable in diagnosing tumors.
Computed tomography scan (CT or CAT scan) – A diagnostic image created after a computer reads X-rays, a CT/CAT scan can show the shape and size of the spinal canal, its contents, and the structures around it. It also is very good at visualizing bony structures.
Magnetic resonance imaging (MRI) – A diagnostic test that produces three-dimensional images of body structures using powerful magnets and computer technology. An MRI can show the spinal cord, nerve roots, and surrounding areas, as well as enlargement, degeneration, and tumors.
Bone Scan – A diagnostic test using Technetium-99. Helpful as an adjunct for identification of bone tumors (such as primary bone tumors of the spine), infection, and diseases involving abnormal bone metabolism.
Radiology studies noted above provide imaging findings that suggest the most likely tumor type. In some cases, however, a biopsy may be needed if the diagnosis is unclear or if concern for malignancy vs benign tumor type. If the tumor is malignant, a biopsy also helps determine cancer’s type, which subsequently determines treatment options.
Staging classifies neoplasms (abnormal tissue) according to the extent of the tumor, assessing bony, soft tissue, and spinal canal involvement. A doctor may order a whole-body scan utilizing nuclear technology, as well as a CT scan of the lungs and abdomen for staging purposes. To confirm the diagnosis, a doctor compares laboratory test results and findings from the aforementioned scans to the patient’s symptoms.
Treatment
Treatment decision-making is often multidisciplinary, incorporating the expertise of spinal surgeons, medical oncologists, radiation oncologists, and other medical specialists. The selection of treatments including both surgical and non-surgical is therefore made keeping in mind the various aspects of the patient’s overall health and goals of care.
Non-Surgical Treatments
Nonsurgical treatment options include observation, chemotherapy, and radiation therapy. Tumors that are asymptomatic or mildly symptomatic and do not appear to be changing or progressing may be observed and monitored with regular MRIs. Some tumors respond well to chemotherapy and others to radiation therapy. However, there are specific types of metastatic tumors that are inherently radioresistant (i.e. gastrointestinal tract and kidney): in those cases, surgery may be the only viable treatment option.
Surgery
Indications for surgery vary depending on the type of tumor. Primary (non-metastatic) spinal tumors may be removed through complete en bloc resection for a possible cure. In patients with metastatic tumors, treatment is primarily palliative, with the goal of restoring or preserving neurological function, stabilizing the spine, and alleviating pain. Generally, surgery is only considered as an option for patients with metastases when they are expected to live 3 – 4 months or longer, and the tumor is resistant to radiation or chemotherapy. Indications for surgery include intractable pain, spinal cord compression, and the need for stabilization of pathological fractures.
For cases in which surgical resection is possible, preoperative embolization may be used to enable an easier resection. This procedure involves the insertion of a catheter or tube through an artery in the groin. The catheter is guided up through the blood vessels to the site of the tumor, where it delivers a glue-like liquid embolic agent that blocks the vessels that feed the tumor. When the blood vessels that feed the tumor are blocked off, bleeding can often be controlled better during surgery, helping to decrease surgical risks.
If surgery is considered, the approach to the tumor is determined by the tumor’s location within the spinal canal. The posterior (back) approach allows for the identification of the dura and exposure of the nerve roots. This approach is commonly used for tumors in the posterior aspect of the spinal column or to expose tumors inside the dura. Multiple levels can be decompressed, and multilevel segmental fixation can be performed if necessary. The anterior (front) approach is excellent for tumors in the front of the spine. This approach also allows for the reconstruction of defects caused by the removal of the vertebral bodies. This approach also allows the placement of short-segment fixation devices. Thoracic and lumbar spinal tumors that affect both the anterior and posterior vertebral columns can be a challenge to resect completely. Not infrequently, a posterior (back) approach followed by a separately staged anterior (front) approach has been utilized surgically to treat these complex lesions.
Cervical Cancer Surgery is a disease in which cells in the body grow out of control. Cancer is always named for the part of the body where it starts, even if it spreads to other body parts later. When cancer starts in the cervix, it is called cervical cancer. The cervix connects the vagina (birth canal) to the upper part of the uterus. The uterus (or womb) is where a baby grows when a woman is pregnant.
Cervical cancer is cancer arising from the cervix. It is due to the abnormal growth of cells that have the ability to invade or spread to other parts of the body.[rx] Early on, typically no symptoms are seen.[rx] Later symptoms may include abnormal vaginal bleeding, pelvic pain or pain during sexual intercourse.[rx] While bleeding after sex may not be serious, it may also indicate the presence of cervical cancer.[rx]
Cervical cancer usually develops slowly over time. Before cancer appears in the cervix, the cells of the cervix go through changes known as dysplasia, in which abnormal cells begin to appear in the cervical tissue. Over time, the abnormal cells may become cancer cells and start to grow and spread more deeply into the cervix and to surrounding areas.
Cervical Cancer Surgery
Large loop excision of the transformation zone (LLETZ)
This is where the cancerous cells are removed using a fine wire and an electrical current.
It’s usually done under local anaesthetic (while you’re awake but the area is numbed) and can be done at the same time as a colposcopy.
Cone biopsy
A cone-shaped area of abnormal tissue is removed during surgery. This is usually done under general anaesthetic (while you’re asleep).
Surgery
There are 3 main types of surgery for cervical cancer:
trachelectomy – the cervix, surrounding tissue and upper part of the vagina are removed, but the womb is left in place
hysterectomy – the cervix and womb are removed and, depending on the stage of the cancer, it may be necessary to remove the ovaries and fallopian tubes
pelvic exenteration – a major operation in which the cervix, vagina, womb, ovaries, fallopian tubes, bladder and rectum may all be removed
Pelvic exenteration is only offered when cervical cancer has come back.
Trachelectomy
A trachelectomy is usually only suitable if cervical cancer is diagnosed at a very early stage. It’s usually offered to women who want to have children in the future.
During the procedure, the cervix and upper section of the vagina are removed, leaving the womb in place. Your womb will then be reattached to the lower section of your vagina.
Lymph nodes (part of the lymphatic system, the body’s waste-removal system) from your pelvis may also be removed.
Compared with a hysterectomy or pelvic exenteration, the advantage of this type of surgery is that your womb remains in place. This means you may still be able to have children.
However, it’s important to be aware that the surgeons carrying out this operation cannot guarantee you will still be able to have children.
A stitch will be put in the bottom of your womb during the surgery. This is to help support and keep a baby in your womb in future pregnancies. If you do get pregnant after the operation, your baby will have to be delivered by caesarean section.
It’s also usually recommended you wait 6 to 12 months after surgery before trying for a baby so your womb and vagina have time to heal.
Trachelectomy is a highly skilled procedure. It’s only available at certain specialist centres in the UK, so it may not be offered in your area and you may need to travel to another city for treatment.
Hysterectomy
A hysterectomy is usually recommended for early cervical cancer. This may be followed by a course of radiotherapy to help prevent the cancer coming back.
Two types of hysterectomies are used to treat cervical cancer:
simple hysterectomy – the cervix and womb are removed and, in some cases, the ovaries and fallopian tubes are too; only appropriate for very early-stage cervical cancers
radical hysterectomy – preferred option in advanced stage 1 and some early stage 2 cervical cancers; the cervix, womb, top of the vagina, surrounding tissue, lymph nodes, fallopian tubes and, sometimes, ovaries are all removed
Short-term complications of a hysterectomy include infection, bleeding, blood clots and accidental injury to your ureter, bladder or rectum.
Although the risk of them is small, long-term complications can be troublesome. They include:
your vagina becoming shorter and drier, which can make sex painful
swelling of your arms and legs, caused by a build-up of fluid (lymphoedema)
your bowel becoming blocked by a build-up of scar tissue – this may require further surgery
Because your womb is removed during a hysterectomy, you will not be able to have children.
If your ovaries are removed, it will also trigger the menopause if you have not already experienced it.
Pelvic exenteration
A pelvic exenteration is a major operation that’s usually only recommended when cervical cancer comes back. It’s offered if the cancer returns to the pelvis but has not spread beyond this area.
A pelvic exenteration involves 2 phases:
the cancer and the vagina are removed – it may also involve removing the bladder, rectum or lower section of the bowel, or all 3
1 or 2 holes, called stomas, are created in your tummy – the holes are used to pass pee and poo out of your body into pouches called colostomy bags
Following pelvic exenteration, it may be possible to reconstruct your vagina using skin and tissue taken from other parts of your body. This would mean you could still have sex after the procedure, although it may be several months until you feel well enough to do so.
Radiotherapy
Radiotherapy may be used on its own or in combination with surgery for early-stage cervical cancer. It may be combined with chemotherapy for advanced cervical cancer, where it can be used to control bleeding and pain.
Radiotherapy can be delivered either:
externally – a machine beams high-energy waves into your pelvis to destroy cancerous cells
internally (brachytherapy) – a radioactive implant is placed next to the tumour inside your vagina
In most cases, a combination of internal and external radiotherapy will be used. A course of radiotherapy usually lasts about 5 to 8 weeks.
As well as destroying cancerous cells, radiotherapy can sometimes also harm healthy tissue. This means it can cause significant side effects many months, or even years, after treatment.
Brachytherapy aims to reduce harm to surrounding tissue by delivering the radiation as close as possible to the tumour, but it can still cause side effects.
However, the benefits of radiotherapy often tend to outweigh the risks. For some people, radiotherapy offers the only hope of getting rid of the cancer.
damage to the ovaries, which will usually trigger an early menopause if you have not already gone through it
bladder and bowel damage, which could lead to incontinence
Most of these side effects will resolve within about 8 weeks of finishing treatment, although in some cases they can be permanent. It’s also possible to develop side effects several months, or even years, after treatment has finished.
If infertility is a concern for you, it may be possible to surgically remove eggs from your ovaries before you have radiotherapy so they can be implanted in your womb at a later date. However, you may have to pay for this.
It may also be possible to prevent an early menopause by surgically removing your ovaries and replanting them outside the area of your pelvis that will be affected by radiation. This is called an ovarian transposition.
Your doctors can provide more information about the possible options for treating infertility and whether you’re suitable for an ovarian transposition.
Chemotherapy
Chemotherapy can be combined with radiotherapy to try to cure cervical cancer, or it can be used as a sole treatment for advanced cancer to slow its progression and relieve symptoms (palliative chemotherapy).
Chemotherapy for cervical cancer usually involves using either a single chemotherapy drug, called cisplatin, or a combination of different chemotherapy drugs to kill the cancerous cells.
Chemotherapy is usually given straight into your vein using a drip. You will probably be seen as an outpatient so will be able to go home once you’ve received your dose.
As with radiotherapy, these medications can also damage healthy tissue. Side effects are therefore common and can include:
feeling and being sick
diarrhoea
feeling tired all the time
reduced production of blood cells, which can make you tired, breathless and vulnerable to infection
mouth ulcers
loss of appetite
hair loss – cisplatin does not usually cause you to lose your hair, but other chemotherapy drugs may
If you do lose your hair, it usually should grow back within 6 months of the completion of your course of chemotherapy.
Some types of chemotherapy medication can damage your kidneys so you may need to have regular blood tests to assess the health of your kidneys.
Follow-up
After you finish your treatment and the cancer has been removed, you’ll need to attend regular appointments for testing. This will usually involve a physical examination of your vagina and cervix (if it hasn’t been removed).
Because cervical cancer can return, these examinations will be used to look for signs of this happening. If the examination finds anything suspicious, a further biopsy can be done.
Follow-up appointments are usually recommended every 3 to 6 months for the first 2 years, and then every 6 to 12 months for a further 3 years.
Your multidisciplinary team (MDT)
Members of your MDT may include:
a surgeon
a clinical oncologist (a specialist in chemotherapy and radiotherapy)
Cervical Cancer Treatment is a disease in which cells in the body grow out of control. Cancer is always named for the part of the body where it starts, even if it spreads to other body parts later. When cancer starts in the cervix, it is called cervical cancer. The cervix connects the vagina (birth canal) to the upper part of the uterus. The uterus (or womb) is where a baby grows when a woman is pregnant.
Cervical cancer is cancer arising from the cervix. It is due to the abnormal growth of cells that have the ability to invade or spread to other parts of the body.[rx] Early on, typically no symptoms are seen.[rx] Later symptoms may include abnormal vaginal bleeding, pelvic pain or pain during sexual intercourse.[rx] While bleeding after sex may not be serious, it may also indicate the presence of cervical cancer.[rx]
Cervical cancer usually develops slowly over time. Before cancer appears in the cervix, the cells of the cervix go through changes known as dysplasia, in which abnormal cells begin to appear in the cervical tissue. Over time, the abnormal cells may become cancer cells and start to grow and spread more deeply into the cervix and to surrounding areas.
Cervical cancer in children is rare.
See the following PDQ summaries for more information about cervical cancer:
Cervical Cancer Prevention
Cervical Cancer Screening
Childhood Cervical and Vaginal Cancer Treatment
Causes of Cervical Cancer
Human papillomavirus (HPV) infection is the major risk factor for cervical cancer.
Anything that increases your chance 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 to your doctor if you think you may be at risk for cervical cancer.
Risk factors for cervical cancer include the following:
Being infected with human papillomavirus (HPV). This is the most important risk factor for cervical cancer.
Being exposed to the drug DES (diethylstilbestrol) while in the mother’s womb.
In women who are infected with HPV, the following risk factors add to the increased risk of cervical cancer:
Giving birth to many children.
Smoking cigarettes.
Using oral contraceptives (“the Pill”) for a long time.
There are also risk factors that increase the risk of HPV infection:
Having a weakened immune system caused by immunosuppression. Immunosuppression weakens the body’s ability to fight infections and other diseases. The body’s ability to fight HPV infection may be lowered by long-term immunosuppression from:
being infected with human immunodeficiency virus (HIV).
taking medicine to help prevent organ rejection after a transplant.
Being sexually active at a young age.
Having many sexual partners.
Older age is a main risk factor for most cancers. The chance of getting cancer increases as you get older.
There are usually no signs or symptoms of early cervical cancer but it can be detected early with regular check-ups.
Early cervical cancer may not cause signs or symptoms. Women should have regular check-ups, including tests to check for human papillomavirus (HPV) or abnormal cells in the cervix. The prognosis (chance of recovery) is better when the cancer is found early.
Symptoms of Cervical Cancer
These and other signs and symptoms may be caused by cervical cancer or by other conditions. Check with your doctor if you have any of the following:
Vaginal bleeding (including bleeding after sexual intercourse).
Unusual vaginal discharge.
Pelvic pain.
Pain during sexual intercourse.
The early stages of cervical cancer may be completely free of symptoms.[rx][rx]
Vaginal bleeding, contact bleeding (one most common form being bleeding after sexual intercourse), or (rarely) a vaginal mass may indicate the presence of malignancy.
Also, moderate pain during sexual intercourse and vaginal discharge are symptoms of cervical cancer.[rx] In advanced disease, metastases may be present in the abdomen, lungs, or elsewhere.[rx]
Symptoms of advanced cervical cancer may include loss of appetite, weight loss, fatigue, pelvic pain, back pain, leg pain, swollen legs, heavy vaginal bleeding, bone fractures, and (rarely) leakage of urine or feces from the vagina.[rx] Bleeding after douching or after a pelvic exam is a common symptom of cervical cancer.[rx]
Diagnosis of Cervical Cancer
Tests that examine the cervix are used to detect (find) and diagnose cervical cancer.
The following 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.
Pelvic exam: An exam of the vagina, cervix, uterus, fallopian tubes, ovaries, and rectum. A speculum is inserted into the vagina and the doctor or nurse looks at the vagina and cervix for signs of disease. A Pap test of the cervix is usually done. The doctor or nurse also inserts one or two lubricated, gloved fingers of one hand into the vagina and places the other hand over the lower abdomen to feel the size, shape, and position of the uterus and ovaries. The doctor or nurse also inserts a lubricated, gloved finger into the rectum to feel for lumps or abnormal areas.
Pap test: A procedure to collect cells from the surface of the cervix and vagina. A piece of cotton, a brush, or a small wooden stick is used to gently scrape cells from the cervix and vagina. The cells are viewed under a microscope to find out if they are abnormal. This procedure is also called a Pap smear.
Human papillomavirus (HPV) test: A laboratory test used to check DNA or RNA for certain types of HPV infection. Cells are collected from the cervix and DNA or RNA from the cells is checked to find out if an infection is caused by a type of HPV that is linked to cervical cancer. This test may be done using the sample of cells removed during a Pap test. This test may also be done if the results of a Pap test show certain abnormal cervical cells.
Endocervical curettage: A procedure to collect cells or tissue from the cervical canal using a curette (spoon-shaped instrument). Tissue samples are taken and checked under a microscope for signs of cancer. This procedure is sometimes done at the same time as a colposcopy.
Colposcopy: A procedure in which a colposcope (a lighted, magnifying instrument) is used to check the vagina and cervix for abnormal areas. Tissue samples may be taken using a curette (spoon-shaped instrument) or a brush and checked under a microscope for signs of disease.
Biopsy: If abnormal cells are found in a Pap test, the doctor may do a biopsy. A sample of tissue is cut from the cervix and viewed under a microscope by a pathologist to check for signs of cancer. A biopsy that removes only a small amount of tissue is usually done in the doctor’s office. A woman may need to go to a hospital for a cervical cone biopsy (removal of a larger, cone-shaped sample of cervical tissue).
After cervical cancer has been diagnosed, tests are done to find out if cancer cells have spread within the cervix or to other parts of the body.
The process used to find out if cancer has spread within the cervix or to other parts of the body is called staging. The information gathered from the staging process determines the stage of the disease. It is important to know the stage in order to plan treatment.
The following tests and procedures may be used in the staging process:
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.
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.
MRI (magnetic resonance imaging): A procedure that uses a magnet, radio waves, and a computer to make a series of detailed pictures of areas inside the body. This procedure is also called nuclear magnetic resonance imaging (NMRI).
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. This picture can be printed to be looked at later.
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.
Lymph node biopsy: The removal of all or part of a lymph node. A pathologist views the lymph node tissue under a microscope to check for cancer cells.
Cystoscopy: A procedure to look inside the bladder and urethra to check for abnormal areas. A cystoscope is inserted through the urethra into the bladder. A cystoscope is a thin, tube-like instrument with a light and a lens for viewing. It may also have a tool to remove tissue samples, which are checked under a microscope for signs of cancer.
Laparoscopy: A surgical procedure to look at the organs inside the abdomen to check for signs of disease. Small incisions (cuts) are made in the wall of the abdomen and a laparoscope (a thin, lighted tube) is inserted into one of the incisions. Other instruments may be inserted through the same or other incisions to perform procedures such as removing organs or taking tissue samples to be checked under a microscope for signs of disease.
Pretreatment surgical staging: Surgery (an operation) is done to find out if the cancer has spread within the cervix or to other parts of the body. In some cases, the cervical cancer can be removed at the same time. Pretreatment surgical staging is usually done only as part of a clinical trial.
The results of these tests are viewed together with the results of the original tumor biopsy to determine the cervical cancer stage.
There are three ways that cancer spreads in the body.
Cancer can spread through tissue, the lymph system, and the blood:
Tissue. Cancer spreads from where it began by growing into nearby areas.
Lymph system. Cancer spreads from where it began by getting into the lymph system. Cancer travels through the lymph vessels to other parts of the body.
Blood. Cancer spreads from where it began by getting into the blood. 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. Cancer gets into the lymph system, travels through the lymph vessels, and forms a tumor (metastatic tumor) in another part of the body.
Blood. 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 cervical cancer spreads to the lung, the cancer cells in the lung are actually cervical cancer cells. The disease is metastatic cervical cancer, not lung cancer.
Abnormal cells may form in the lining of the cervix (carcinoma in situ).
In carcinoma in situ, abnormal cells are found in the innermost lining of the cervix. These abnormal cells may become cancer and spread into nearby normal tissue.
The following stages are used for cervical cancer:
Stage I
In stage I, cancer has formed and is found in the cervix only.
Stage I is divided into stages IA and IB, based on the size of the tumor and the deepest point of tumor invasion.
Stage IA: Stage IA is divided into stages IA1 and IA2, based on the deepest point of tumor invasion.
In stage IA1, a very small amount of cancer that can only be seen with a microscope is found in the tissues of the cervix. The deepest point of tumor invasion is 3 millimeters or less.
In stage IA2, a very small amount of cancer that can only be seen with a microscope is found in the tissues of the cervix. The deepest point of tumor invasion is more than 3 millimeters but not more than 5 millimeters.
Stage IB: Stage IB is divided into stages IB1, IB2, and IB3, based on the size of the tumor and the deepest point of tumor invasion.
In stage IB1, the tumor is 2 centimeters or smaller and the deepest point of tumor invasion is more than 5 millimeters.
In stage IB2, the tumor is larger than 2 centimeters but not larger than 4 centimeters.
In stage IB3, the tumor is larger than 4 centimeters.
Stage II
In stage II, cancer has spread to the upper two-thirds of the vagina or to the tissue around the uterus.
Stage II is divided into stages IIA and IIB, based on how far the cancer has spread.
Stage IIA: Cancer has spread from the cervix to the upper two-thirds of the vagina but has not spread to the tissue around the uterus. Stage IIA is divided into stages IIA1 and IIA2, based on the size of the tumor.
In stage IIA1, the tumor is 4 centimeters or smaller.
In stage IIA2, the tumor is larger than 4 centimeters.
Stage IIB: Cancer has spread from the cervix to the tissue around the uterus.
Stage III
In stage III, cancer has spread to the lower third of the vagina and/or to the pelvic wall, and/or has caused kidney problems, and/or involves lymph nodes.
Stage III is divided into stages IIIA, IIIB, and IIIC, based on how far the cancer has spread.
Stage IIIA: Cancer has spread to the lower third of the vagina but has not spread to the pelvic wall.
Stage IIIB: Cancer has spread to the pelvic wall; and/or the tumor has become large enough to block one or both ureters or has caused one or both kidneys to get bigger or stop working.
Stage IIIC: Stage IIIC is divided into stages IIIC1 and IIIC2, based on the spread of cancer to the lymph nodes.
In stage IIIC1, cancer has spread to lymph nodes in the pelvis.
In stage IIIC2, cancer has spread to lymph nodes in the abdomen near the aorta.
Stage IV
In stage IV, cancer has spread beyond the pelvis, or has spread to the lining of the bladder or rectum, or has spread to other parts of the body.
Stage IV is divided into stages IVA and IVB, based on where the cancer has spread.
Stage IVA: Cancer has spread to nearby organs, such as the bladder or rectum.
Stage IVB: Cancer has spread to other parts of the body, such as the liver, lungs, bones, or distant lymph nodes.
Treatment of Cervical Cancer
KEY POINTS
There are different types of treatment for patients with cervical cancer.
Five types of standard treatment are used:
Surgery
Radiation therapy
Chemotherapy
Targeted therapy
Immunotherapy
New types of treatment are being tested in clinical trials.
Treatment for cervical cancer may cause side effects.
Patients may want to think about taking part in a clinical trial.
Patients can enter clinical trials before, during, or after starting their cancer treatment.
Follow-up tests may be needed.
There are different types of treatment for patients with cervical cancer.
Different types of treatment are available for patients with cervical cancer. Some treatments are standard (the currently used treatment), and some are being tested in clinical trials. A treatment clinical trial is a research study meant to help improve current treatments or obtain information on new treatments for patients with cancer. When clinical trials show that a new treatment is better than the standard treatment, the new treatment may become the standard treatment. Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
Five types of standard treatment are used:
Surgery
Surgery (removing the cancer in an operation) is sometimes used to treat cervical cancer. The following surgical procedures may be used:
Conization: A procedure to remove a cone-shaped piece of tissue from the cervix and cervical canal. A pathologist views the tissue under a microscope to look for cancer cells. Conization may be used to diagnose or treat a cervical condition. This procedure is also called a cone biopsy.
Conization may be done using one of the following procedures:
Cold-knife conization: A surgical procedure that uses a scalpel (sharp knife) to remove abnormal tissue or cancer.
Loop electrosurgical excision procedure (LEEP): A surgical procedure that uses electrical current passed through a thin wire loop as a knife to remove abnormal tissue or cancer.
Laser surgery: A surgical procedure that uses a laser beam (a narrow beam of intense light) as a knife to make bloodless cuts in tissue or to remove a surface lesion such as a tumor.
The type of conization procedure used depends on where the cancer cells are in the cervix and the type of cervical cancer.
Total hysterectomy: Surgery to remove the uterus, including the cervix. If the uterus and cervix are taken out through the vagina, the operation is called a vaginal hysterectomy. If the uterus and cervix are taken out through a large incision (cut) in the abdomen, the operation is called a total abdominal hysterectomy. If the uterus and cervix are taken out through a small incision in the abdomen using a laparoscope, the operation is called a total laparoscopic hysterectomy.
Radical hysterectomy: Surgery to remove the uterus, cervix, part of the vagina, and a wide area of ligaments and tissues around these organs. The ovaries, fallopian tubes, or nearby lymph nodes may also be removed.
Modified radical hysterectomy: Surgery to remove the uterus, cervix, upper part of the vagina, and ligaments and tissues that closely surround these organs. Nearby lymph nodes may also be removed. In this type of surgery, not as many tissues and/or organs are removed as in a radical hysterectomy.
Radical trachelectomy: Surgery to remove the cervix, nearby tissue and lymph nodes, and the upper part of the vagina. The uterus and ovaries are not removed.
Bilateral salpingo-oophorectomy: Surgery to remove both ovaries and both fallopian tubes.
Pelvic exenteration: Surgery to remove the lower colon, rectum, and bladder. The cervix, vagina, ovaries, and nearby lymph nodes are also removed. Artificial openings (stoma) are made for urine and stool to flow from the body to a collection bag. Plastic surgery may be needed to make an artificial vagina after this operation.
Radiation therapy
Radiation therapy is a cancer treatment that uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. There are two types of radiation therapy:
External radiation therapy uses a machine outside the body to send radiation toward the cancer. Certain ways of giving radiation therapy can help keep radiation from damaging nearby healthy tissue. This type of radiation therapy includes the following:
Intensity-modulated radiation therapy (IMRT): IMRT is a type of 3-dimensional (3-D) radiation therapy that uses a computer to make pictures of the size and shape of the tumor. Thin beams of radiation of different intensities (strengths) are aimed at the tumor from many angles.
Internal radiation therapy uses a radioactive substance sealed in needles, seeds, wires, or catheters that are placed directly into or near the cancer.
The way the radiation therapy is given depends on the type and stage of the cancer being treated. External and internal radiation therapy are used to treat cervical cancer, and may also be used as palliative therapy to relieve symptoms and improve quality of life.
Chemotherapy
Chemotherapy is a cancer treatment that uses drugs to stop the growth of cancer cells, either by killing the cells or by stopping them from dividing. When chemotherapy is taken by mouth or injected into a vein or muscle, the drugs enter the bloodstream and can reach cancer cells throughout the body (systemic chemotherapy). When chemotherapy is placed directly into the cerebrospinal fluid, an organ, or a body cavity such as the abdomen, the drugs mainly affect cancer cells in those areas (regional chemotherapy). The way the chemotherapy is given depends on the type and stage of the cancer being treated.
Targeted therapy
Targeted therapy is a type of treatment that uses drugs or other substances to identify and attack specific cancer cells without harming normal cells.
Monoclonal antibody therapy is a type of targeted therapy that uses antibodies made in the laboratory from a single type of immune system cell. These antibodies can identify substances on cancer cells or normal substances that may help cancer cells grow. The antibodies attach to the substances and 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.
Bevacizumab is a monoclonal antibody that binds to a protein called vascular endothelial growth factor (VEGF) and may prevent the growth of new blood vessels that tumors need to grow. Bevacizumab is used to treat cervical cancer that has metastasized (spread to other parts of the body) and recurrent cervical cancer.
Immunotherapy
Immunotherapy is a treatment that uses the patient’s immune system to fight cancer. Substances made by the body or made in a laboratory are used to boost, direct, or restore the body’s natural defenses against cancer. This type of cancer treatment is also called biotherapy or biologic therapy.
Immune checkpoint inhibitor therapy is a type of immunotherapy.
Immune checkpoint inhibitor therapy: PD-1 is a protein on the surface of T cells that helps keep the body’s immune responses in check. When PD-1 attaches to another protein called PDL-1 on a cancer cell, it stops the T cell from killing the cancer cell. PD-1 inhibitors attach to PDL-1 and allow the T cells to kill cancer cells. Pembrolizumab is a type of immune checkpoint inhibitor used to treat recurrent cervical cancer.
New types of treatment are being tested in clinical trials.
Information about clinical trials is available from the NCI website.
Treatment for cervical cancer may cause side effects.
Patients may want to think about taking part in a clinical trial.
For some patients, taking part in a clinical trial may be the best treatment choice. Clinical trials are part of the cancer research process. Clinical trials are done to find out if new cancer treatments are safe and effective or better than the standard treatment.
Many of today’s standard treatments for cancer are based on earlier clinical trials. Patients who take part in a clinical trial may receive the standard treatment or be among the first to receive a new treatment.
Patients who take part in clinical trials also help improve the way cancer will be treated in the future. Even when clinical trials do not lead to effective new treatments, they often answer important questions and help move research forward.
Patients can enter clinical trials before, during, or after starting their cancer treatment.
Some clinical trials only include patients who have not yet received treatment. Other trials test treatments for patients whose cancer has not gotten better. There are also clinical trials that test new ways to stop cancer from recurring (coming back) or reduce the side effects of cancer treatment.
Clinical trials are taking place in many parts of the country. Information about clinical trials supported by NCI can be found on NCI’s clinical trials search webpage. Clinical trials supported by other organizations can be found on the ClinicalTrials.gov website.
Follow-up tests may be needed.
Some of the tests that were done to diagnose the cancer or to find out the stage of the cancer may be repeated. Some tests will be repeated in order to see how well the treatment is working. Decisions about whether to continue, change, or stop treatment may be based on the results of these tests.
Some of the tests will continue to be done from time to time after treatment has ended. The results of these tests can show if your condition has changed or if the cancer has recurred (come back). These tests are sometimes called follow-up tests or check-ups.
Your doctor will ask if you have any of the following signs or symptoms, which may mean the cancer has come back:
Pain in the abdomen, back, or leg.
Swelling in the leg.
Trouble urinating.
Cough.
Feeling tired.
For cervical cancer, follow-up tests are usually done every 3 to 4 months for the first 2 years, followed by check-ups every 6 months. The check-up includes a current health history and exam of the body to check for signs and symptoms of recurrent cervical cancer and for late effects of treatment.
Treatment Options by Stage
Carcinoma in Situ
Treatment of carcinoma in situ may include the following:
Conization, such as cold-knife conization, loop electrosurgical excision procedure (LEEP), or laser surgery.
Hysterectomy for women who cannot or no longer want to have children. This is done only if the tumor cannot be completely removed by conization.
Internal radiation therapy for women who cannot have surgery.
Stage IA Cervical Cancer
Stage IA cervical cancer is separated into stage IA1 and IA2.
Treatment for stage IA1 may include the following:
Conization.
Total hysterectomy with or without bilateral salpingo-oophorectomy.
Treatment for stage IA2 may include the following:
Modified radical hysterectomy and removal of lymph nodes.
Radical trachelectomy.
Internal radiation therapy for women who cannot have surgery.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Stages IB and IIA Cervical Cancer
Treatment of stage IB and stage IIA cervical cancer may include the following:
Radiation therapy with chemotherapy given at the same time.
Radical hysterectomy and removal of pelvic lymph nodes with or without radiation therapy to the pelvis, plus chemotherapy.
Radical trachelectomy.
Chemotherapy followed by surgery.
Radiation therapy alone.
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.
Stages IIB, III, and IVA Cervical Cancer
Treatment of stage IIB, stage III, and stage IVA cervical cancer may include the following:
Radiation therapy with chemotherapy given at the same time.
Surgery to remove pelvic lymph nodes followed by radiation therapy with or without chemotherapy.
Internal radiation therapy.
A clinical trial of chemotherapy to shrink the tumor followed by surgery.
A clinical trial of chemotherapy and radiation therapy given at the same time, followed by chemotherapy.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Stage IVB Cervical Cancer
Treatment of stage IVB cervical cancer may include the following:
Radiation therapy as palliative therapy to relieve symptoms caused by cancer and improve quality of life.
Chemotherapy and targeted therapy.
Chemotherapy as palliative therapy to relieve symptoms caused by cancer and improve quality of life.
Clinical trials of new anticancer drugs or drug combinations.
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. Treatment Options for Recurrent Cervical Cancer
Treatment of recurrent cervical cancer may include the following:
Immunotherapy.
Radiation therapy and chemotherapy.
Chemotherapy and targeted therapy.
Chemotherapy as palliative therapy to relieve symptoms caused by cancer and improve quality of life.
Pelvic exenteration.
Clinical trials of new anticancer drugs or drug combinations.
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.
Cervical Cancer During Pregnancy
General Information About Cervical Cancer During Pregnancy
Treatment of cervical cancer during pregnancy depends on the stage of the cancer and how long the patient has been pregnant. A biopsy and imaging tests may be done to determine the stage of the disease. To avoid exposing the fetus to radiation, MRI (magnetic resonance imaging) is used.
Treatment Options for Cervical Cancer During Pregnancy
Carcinoma in Situ During Pregnancy
Usually, no treatment is needed for carcinoma in situ during pregnancy. A colposcopy may be done to check for invasive cancer.
Stage I Cervical Cancer During Pregnancy
Pregnant women with slow-growing stage I cervical cancer may be able to delay treatment until the second trimester of pregnancy or after delivery.
Pregnant women with fast-growing stage I cervical cancer may need immediate treatment. Treatment may include:
Conization.
Radical trachelectomy.
Women should be tested to find out if the cancer has spread to the lymph nodes. If cancer has spread to the lymph nodes, immediate treatment may be needed.
Stage II, III, and IV Cervical Cancer During Pregnancy
Chemotherapy to shrink the tumor in the second or third trimester of pregnancy. Surgery or radiation therapy may be done after delivery.
Radiation therapy plus chemotherapy. Talk with your doctor about the effects of radiation on the fetus. It may be necessary to end the pregnancy before treatment begins.
Cervical cancer is the fourth most common cancer in women worldwide, and it has the fourth highest mortality rate among cancers in women.[rx] Most cases of cervical cancer are preventable by routine screening and by treatment of precancerous lesions. As a result, most of the cervical cancer cases are diagnosed in women who live in regions with inadequate screening protocols.
Incidence and Mortality
Estimated new cases and deaths from cervical (uterine cervix) cancer in the United States in 2020:[rx]
New cases: 13,800.
Deaths: 4,290.
Anatomy of Cervical Cancer
The uterine cervix is contiguous with the uterine body, and it acts as the opening to the body of the uterus. The uterine cervix is a cylindrical, fibrous organ that is an average of 3 to 4 cm in length. The portio of the cervix is the part of the cervix that is visible on vaginal inspection. The opening of the cervix is termed the external os. The os is the beginning of the endocervical canal, which forms the inner aspect of the cervix. At the upper aspect of the endocervical canal is the internal os, a narrowing of the endocervical canal. The narrowing marks the transition from the cervix to the uterine body. The endocervical canal beyond the internal os is termed the endometrial canal.
The cervix is lined by two types of epithelial cells: squamous cells at the outer aspect, and columnar, glandular cells along the inner canal. The transition between squamous cells and columnar cells is an area termed the squamocolumnar junction. Most the precancerous and cancerous changes arise in this zone.
Pathogenesis
Cervical carcinoma has its origins at the squamous-columnar junction; it can involve the outer squamous cells, the inner glandular cells, or both. The precursor lesion is dysplasia: cervical intraepithelial neoplasia (CIN) or adenocarcinoma in situ, which can subsequently become invasive cancer. This process can be quite slow. Longitudinal studies have shown that in patients with untreated in situ cervical cancer, 30% to 70% will develop invasive carcinoma over a period of 10 to 12 years. However, in about 10% of patients, lesions can progress from in situ to invasive in a period of less than 1 year. As it becomes invasive, the tumor breaks through the basement membrane and invades the cervical stroma. Extension of the tumor in the cervix may ultimately manifest as ulceration, exophytic tumor, or extensive infiltration of underlying tissue, including the bladder or rectum.
Risk Factors
Increasing age is the most important risk factor for most cancers. The primary risk factor for cervical cancer is human papillomavirus (HPV) infection.[rx–rx]
Other risk factors for cervical cancer include the following:
Exposure to diethylstilbestrol (DES) in utero.[rx]
Human papillomavirus (HPV) infection
HPV infection is a necessary step in the development of virtually all precancerous and cancerous lesions. Epidemiologic studies convincingly demonstrate that the major risk factor for the development of preinvasive or invasive carcinoma of the cervix is HPV infection, far outweighing other known risk factors.
More than 6 million women in the United States are estimated to be infected with HPV. Transient HPV infection is common, particularly in young women,[rx] while cervical cancer is rare. The persistence of an HPV infection leads to an increased risk of developing precancerous and cancerous lesions.[rx,rx]
The strain of HPV infection is also important in conferring risk. There are multiple subtypes of HPV that infect humans; of these, subtypes 16 and 18 have been most closely associated with high-grade dysplasia and cancer. Studies suggest that acute infection with HPV types 16 and 18 conferred an 11-fold to 16.9-fold risk of rapid development of high-grade CIN.[rx–rx] Further studies have shown that infection with either HPV 16 or 18 is more predictive than cytologic screening of high-grade CIN or greater disease, and that the predictive ability is seen for up to 18 years after the initial test.[rx–rx]
There are two commercially available vaccines that target anogenital-related strains of HPV. The vaccines are directed towards HPV-naïve girls and young women, and although penetration of the vaccine has been moderate, significant decreases in HPV-related diseases have been documented.[rx]
Symptoms of Cervical Cancer
Early cervical cancer may not cause noticeable signs or symptoms.
Possible signs and symptoms of cervical cancer include the following:
Vaginal bleeding.
Unusual vaginal discharge.
Pelvic pain.
Dyspareunia.
Postcoital bleeding.
Diagnosis of Cervical Cancer
The following procedures may be used to diagnose cervical cancer:
History and physical exam.
Pelvic exam.
Cervical cytology (Pap smear).
HPV test.
Endocervical curettage.
Colposcopy.
Biopsy.
HPV testing
Cervical cytology (Pap smear) has been the mainstay of cervical cancer screening since its introduction. However, molecular techniques for the identification of HPV DNA are highly sensitive and specific. Current screening options include the following:
Cytology alone.
Cytology and HPV testing.
HPV testing is suggested when it is likely to successfully triage patients into low- and high-risk groups for a high-grade dysplasia or greater lesion.
HPV DNA tests are unlikely to separate patients with low-grade squamous intraepithelial lesions into those who do and those who do not need further evaluation. A study of 642 women found that 83% had one or more tumorigenic HPV types when cervical cytologic specimens were assayed by a sensitive (hybrid capture) technique.[rx] The authors of the study and of an accompanying editorial concluded that using HPV DNA testing in this setting does not add sufficient information to justify its cost.[rx]
HPV DNA testing has proven useful in triaging patients with atypical squamous cells of undetermined significance to colposcopy and has been integrated into current screening guidelines.[rx–rx]
Other studies show that patients with low-risk cytology and high-risk HPV infection with types 16, 18, and 31 are more likely to have CIN or micro-invasive histopathology on biopsy.[rx,rx–rx] One method has also shown that integration of HPV types 16 and 18 into the genome, leading to transcription of viral and cellular messages, may predict patients who are at greater risk for high-grade dysplasia and invasive cancer.[rx]
For women older than 30 years who are more likely to have persistent HPV infection, HPV typing can successfully triage women into high- and low-risk groups for CIN 3 or worse disease. In this age group, HPV DNA testing is more effective than cytology alone in predicting the risk of developing CIN 3 or worse.[rx] Other studies have shown the effectiveness of a primary HPV DNA–screening strategy with cytology triage over the previously used cytology-based screening algorithms.[rx,rx]
Prognostic Factors
The prognosis for patients with cervical cancer is markedly affected by the extent of disease at the time of diagnosis. More than 90% of cervical cancer cases can be detected early through the use of the Pap test and HPV testing.[35] Pap and HPV testing are not performed on approximately 33% of eligible women, which results in a higher-than-expected death rate.
Clinical stage
Clinical stage as a prognostic factor is supplemented by several gross and microscopic pathologic findings in surgically treated patients.
Evidence (clinical stage and other findings):
In a large, surgicopathologic staging study of patients with clinical stage IB disease reported by the Gynecologic Oncology Group (GOG) (GOG-49), the factors that most prominently predicted for lymph node metastases and a decrease in disease-free survival were capillary-lymphatic space involvement by tumor, increasing tumor size, and increasing depth of stromal invasion, with the latter being the most important and reproducible.[rx,rx]
In a study of 1,028 patients treated with radical surgery, survival rates correlated more consistently with tumor volume (as determined by precise volumetry of the tumor) than with clinical or histologic stage.[rx]
A multivariate analysis of prognostic variables in 626 patients with locally advanced disease (primarily stages II, III, and IV) studied by the GOG identified the following variables that were significant for progression-free interval and survival:[rx]
Periaortic and pelvic lymph node status.
Tumor size.
Patient age.
Performance status.
Bilateral disease.
Clinical stage.
The study confirmed the overriding importance of positive periaortic nodes and suggested further evaluation of these nodes in locally advanced cervical cancer. The status of the pelvic nodes was important only if the periaortic nodes were negative. This was also true for tumor size.
It is controversial whether adenocarcinoma of the cervix carries a significantly worse prognosis than squamous cell carcinoma of the cervix.[rx] Several population-based and retrospective studies show a worse outcome for patients with adenocarcinoma, with an increase in distant metastasis noted, when compared with those with squamous histology.[rx–rx] Reports conflict about the effect of adenosquamous cell type on the outcome.[rx,rx] One report showed that approximately 25% of apparent squamous tumors have demonstrable mucin production and behave more aggressively than their pure squamous counterparts, suggesting that any adenomatous differentiation may confer a negative prognosis.[rx]
In a large series of cervical cancer patients treated by radiation therapy, the incidence of distant metastases (most frequently to the lung, abdominal cavity, liver, and gastrointestinal tract) was shown to increase as the stage of disease increased, from 3% in stage IA to 75% in stage IVA.[rx] A multivariate analysis of factors influencing the incidence of distant metastases showed stage, an endometrial extension of tumor, and pelvic tumor control to be significant indicators of distant dissemination.[rx]
GOG studies have indicated that prognostic factors vary depending on whether clinical or surgical staging are utilized and with different treatments. Delay in radiation delivery completion is associated with poorer progression-free survival when clinical staging is used. To-date, stage, tumor grade, race, and age are uncertain prognostic factors in studies utilizing chemoradiation.[rx]
Other prognostic factors
Other prognostic factors that may affect outcome include the following:
HIV status: Women with HIV have more aggressive and advanced disease and a poorer prognosis.[rx]
C-myc overexpression: A study of patients with known invasive squamous carcinoma of the cervix found that overexpression of the C-myc oncogene was associated with a poorer prognosis.[rx]
Number of cells in S phase: The number of cells in S phase may also have prognostic significance in early cervical carcinoma.[rx]
HPV-18 DNA: HPV-18 DNA has been found to be an independent adverse molecular prognostic factor. Two studies have shown a worse outcome when HPV-18 was identified in cervical cancers of patients undergoing radical hysterectomy and pelvic lymphadenectomy.[rx,rx]
A polymorphism in the Gamma-glutamyl hydrolase enzyme, which is related to folate metabolism, has been shown to decrease response to cisplatin, and as a result is associated with poorer outcomes.[rx]
Follow-up After Treatment
High-quality studies are lacking, and the optimal treatment follow-up for patients after treatment for cervical cancer is unknown. Retrospective studies have shown that patients who recur are most likely to do so within the first 2 years.[rx] As a result, most guidelines suggest routine follow-up every 3 to 4 months for the first 2 years, followed by evaluations every 6 months. Most recurrences are diagnosed secondary to new patient symptoms and signs,[57,58] and the usefulness of routine testing including a Pap smear and chest x-ray is unclear.
Follow-up should be centered around a thorough history and physical examination with a careful review of symptoms; imaging should be reserved for evaluation of a positive finding. Patients should be asked about possible warning signs, including the following:
Abdominal pain.
Back pain.
Painful or swollen leg.
Problems with urination.
Cough.
Fatigue.
The follow-up examination should also screen for possible complications of previous treatment because of the multiple modalities (surgery, chemotherapy, and radiation) that patients often undergo during their treatment.
Cellular Classification of Cervical Cancer
Squamous cell (epidermoid) carcinoma comprises approximately 90% of cervical cancers, and adenocarcinoma comprises approximately 10% of cervical cancers. Adenosquamous and small cell carcinomas are relatively rare. Primary sarcomas of the cervix and primary and secondary malignant lymphomas of the cervix have also been reported.
Stage Information for Cervical Cancer
Carcinoma of the cervix can spread via local invasion, the regional lymphatics, or bloodstream. Tumor dissemination is generally a function of the extent and invasiveness of the local lesion. While cancer of the cervix generally progresses in an orderly manner, occasionally a small tumor with distant metastasis is seen. For this reason, patients must be carefully evaluated for metastatic disease.
Pretreatment surgical staging is the most accurate method to determine the extent of disease,[1] but there is little evidence to demonstrate overall improved survival with routine surgical staging; the staging is usually performed only as part of a clinical trial. Pretreatment surgical staging in bulky but locally curable disease may be indicated in select cases when a nonsurgical search for metastatic disease is negative. If abnormal nodes are detected by computed tomography (CT) scan or lymphangiography, fine-needle aspiration should be negative before a surgical staging procedure is performed.
Tests and procedures to evaluate the extent of the disease include the following:
The Fédération Internationale de Gynécologie et d’Obstétrique (FIGO) and the American Joint Committee on Cancer have designated staging to define cervical cancer; the FIGO system is most commonly used.[rx,rx]
Table 1. Definitions of FIGO Stage Ia
Stage
Description
Illustration
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
aAdapted from FIGO Committee for Gynecologic Oncology.[rx]
bImaging and pathology can be used, when available, to supplement clinical findings with respect to tumor size and extent, in all stages. Pathological findings supersede imaging and clinical findings.
cThe involvement of vascular/lymphatic spaces should not change the staging. The lateral extent of the lesion is no longer considered.
I
The carcinoma is strictly confined to the cervix (extension to the corpus should be disregarded).
IA
Invasive carcinoma that can be diagnosed only by microscopy, with maximum depth of invasion ≤5 mm.b
–IA1
–Measured stromal invasion ≤3 mm in depth.
–IA2
–Measured stromal invasion >3 mm and ≤5 mm in depth.
IB
Invasive carcinoma with measured deepest invasion >5 mm (greater than stage IA); lesion limited to the cervix uteri with size measured by maximum tumor diameter.c
–IB1
–Invasive carcinoma >5 mm depth of stromal invasion and ≤2 cm in greatest dimension.
–IB2
–Invasive carcinoma >2 cm and ≤4 cm in greatest dimension.
–IB3
–Invasive carcinoma >4 cm in greatest dimension.
Table 2. Definitions of FIGO Stage IIa
Stage
Description
Illustration
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
aAdapted from FIGO Committee for Gynecologic Oncology.[rx]
II
The cervical carcinoma invades beyond the uterus, but has not extended onto the lower third of the vagina or to the pelvic wall.
IIA
Involvement limited to the upper two-thirds of the vagina without parametrial involvement.
–IIA1
–Invasive carcinoma ≤4 cm in greatest dimension.
–IIA2
–Invasive carcinoma >4 cm in greatest dimension.
IIB
With parametrial involvement but not up to the pelvic wall.
Table 3. Definitions of FIGO Stage IIIa
Stage
Description
Illustration
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
aAdapted from FIGO Committee for Gynecologic Oncology.[3]
bIsolated tumor cells do not change the stage but their presence should be recorded.
cAdding notation of r (imaging) and p (pathology) to indicate the findings that are used to allocate the case to stage IIIC. For example, if imaging indicates pelvic lymph node metastasis, the stage allocation would be stage IIIC1r; if confirmed by pathological findings, it would be stage IIIC1p. The type of imaging modality or pathology technique used should always be documented. When in doubt, the lower staging should be assigned.
III
The carcinoma involves the lower third of the vagina and/or extends to the pelvic wall and/or causes hydronephrosis or nonfunctioning kidney and/or involves pelvic and/or para-aortic lymph nodes.
IIIA
Carcinoma involves the lower third of the vagina, with no extension to the pelvic wall.
IIIB
Extension to the pelvic wall and/or hydronephrosis or nonfunctioning kidney (unless known to be due to another cause).
IIIC
Involvement of pelvic and/or para-aortic lymph nodes (including micrometastases)b, irrespective of tumor size and extent (with r and p notations).c
–IIIC1
–Pelvic lymph node metastasis only.
–IIIC2
–Para-aortic lymph node metastasis.
Table 4. Definitions of FIGO Stage IVa
Stage
Description
Illustration
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
aAdapted from FIGO Committee for Gynecologic Oncology.[rx]
IV
The carcinoma has extended beyond the true pelvis or has involved (biopsy proven) the mucosa of the bladder or rectum. A bullous edema, as such, does not permit a case to be allotted to stage IV.
IVA
Spread of the growth to adjacent organs.
IVB
Spread to distant organs.
Treatment Option Overview for Cervical Cancer
Patterns-of-care studies clearly demonstrate the negative prognostic effect of increasing tumor volume and spread pattern.[1] Treatment, therefore, may vary within each stage as the individual stages are currently defined by Fédération Internationale de Gynécologie et d’Obstétrique (FIGO).
Table 5. Standard Treatment Options for Cervical Cancer
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
In situ carcinoma of the cervix (this stage is not recognized by FIGO)
Conization
Hysterectomy for postreproductive patients
Internal radiation therapy for medically inoperable patients
Stage IA cervical cancer
Conization
Total hysterectomy
Modified radical hysterectomy with lymphadenectomy
Radical trachelectomy
Intracavitary radiation therapy
Stages IB, IIA cervical cancer
Radiation therapy with concomitant chemotherapy
Radical hysterectomy and bilateral pelvic lymphadenectomy with or without total pelvic radiation therapy plus chemotherapy
Radical trachelectomy
Neoadjuvant chemotherapy
Radiation therapy alone
Intensity Modulated Radiation Therapy (IMRT)
Stages IIB, III, and IVA cervical cancer
Radiation therapy with concomitant chemotherapy
Interstitial brachytherapy
Neoadjuvant chemotherapy
Stage IVB cervical cancer
Palliative radiation therapy
Palliative chemotherapy and other systemic therapy
Recurrent cervical cancer
Radiation therapy and chemotherapy
Palliative chemotherapy and other systemic therapy
Pelvic exenteration
Chemoradiation Therapy
Five randomized, phase III trials (GOG-85, RTOG-9001, GOG-120, GOG-123, and SWOG-8797) have shown an overall survival advantage for cisplatin-based therapy given concurrently with radiation therapy,[rx–rx] while one trial examining this regimen demonstrated no benefit.[rx] The patient populations in these studies included women with FIGO stages IB2 to IVA cervical cancer treated with primary radiation therapy and women with FIGO stages I to IIA disease who were found to have poor prognostic factors (a metastatic disease in pelvic lymph nodes, parametrial disease, or positive surgical margins) at the time of primary surgery.
Although the positive trials vary in terms of the stage of the disease, dose of radiation, and schedule of cisplatin and radiation, the trials demonstrate a significant survival benefit for this combined approach. The risk of death from cervical cancer was decreased by 30% to 50% with the use of concurrent chemoradiation therapy.
Based on these results, strong consideration should be given to the incorporation of concurrent cisplatin-based chemotherapy with radiation therapy in women who require radiation therapy for treatment of cervical cancer.[rx–rx]
Surgery and Radiation Therapy
Surgery and radiation therapy are equally effective for early stage, small-volume disease.[11] Younger patients may benefit from surgery to preserve the ovaries and avoid vaginal atrophy and stenosis.
Therapy for patients with cancer of the cervical stump is effective and yields results that are comparable with those seen in patients with an intact uterus.[rx]
In Situ Cervical Cancer Treatment
Consensus guidelines have been issued for managing women with cervical intraepithelial neoplasia or adenocarcinoma in situ.[rx] Properly treated, tumor control of in situ cervical carcinoma should be nearly 100%. Either expert colposcopic-directed biopsy or cone biopsy is required to exclude invasive disease before therapy is undertaken. A correlation between cytology and colposcopic-directed biopsy is also necessary before local ablative therapy is done. Unrecognized invasive disease treated with inadequate ablative therapy may be the most common cause of failure.[rx] Failure to identify the disease, lack of correlation between the Pap smear and colposcopic findings, adenocarcinoma in situ, or extension of disease into the endocervical canal makes a laser, loop, or cold-knife conization mandatory.
The choice of treatment depends on the extent of disease and several patient factors, including age, cell type, desire to preserve fertility, and medical condition.
Standard Treatment Options for In Situ Cervical Cancer
Standard treatment options for in situ cervical cancer include the following:
Internal radiation therapy for medically inoperable patients.
Hysterectomy is the standard treatment for patients with adenocarcinoma in situ. The disease, which originates in the endocervical canal, may be more difficult to completely excise with a conization procedure. Conization may be offered to select patients with adenocarcinoma in situ who desire future fertility.
Conization
When the endocervical canal is involved, laser or cold-knife conization may be used for selected patients to preserve the uterus, avoid radiation therapy, and more extensive surgery.[rx]
In selected cases, the outpatient LEEP may be an acceptable alternative to cold-knife conization. This procedure requires only local anesthesia and obviates the risks associated with general anesthesia for cold-knife conization.[rx–rx] However, controversy exists about the adequacy of LEEP as a replacement for conization; LEEP is unlikely to be sufficient for patients with adenocarcinoma in situ.[rx]
Evidence (conization using LEEP):
A trial comparing LEEP with cold-knife cone biopsy showed no difference in the likelihood of complete excision of dysplasia.[rx]
Two case reports suggested that the use of LEEP in patients with occult invasive cancer led to an inability to accurately determine depth of invasion when a focus of the cancer was transected.[rx]
Hysterectomy for postreproductive patients
Hysterectomy is standard therapy for women with cervical adenocarcinoma in situ, because of the location of the disease in the endocervical canal and the possibility for skip lesions in this region, making margin status a less reliable prognostic factor. However, the effect of hysterectomy compared with conservative surgical measures on mortality has not been studied. Hysterectomy may be performed for squamous cell carcinoma in situ if conization is not possible because of previous surgery, or if positive margins are noted after conization therapy. Hysterectomy is not an acceptable front-line therapy for squamous carcinoma in situ.[rx]
Internal radiation therapy for medically inoperable patients
For medically inoperable patients, a single intracavitary insertion with tandem and ovoids for 5,000 mg hours (80 Gy vaginal surface dose) may be used.[rx]
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.
Stage IA Cervical Cancer Treatment
Standard Treatment Options for Stage IA1 Cervical Cancer
Standard treatment options for stage IA1 cervical cancer include the following:
Conization.
Total hysterectomy.
Conization
If the depth of invasion is less than 3 mm, no vascular or lymphatic channel invasion is noted, and the margins of the cone are negative, conization alone may be appropriate in patients who wish to preserve fertility.[rx]
Total hysterectomy
If the depth of invasion is less than 3 mm, which is proven by cone biopsy with clear margins,[rx] no vascular or lymphatic channel invasion is noted, and the frequency of lymph-node involvement is sufficiently low, lymph-node dissection at the time of hysterectomy is not required. Oophorectomy is optional and should be deferred for younger women.
Standard Treatment Options for Stage IA2 Cervical Cancer
Standard treatment options for stage IA2 cervical cancer include the following:
Modified radical hysterectomy with lymphadenectomy.
Modified radical hysterectomy with lymphadenectomy
For patients with tumor invasion between 3 mm and 5 mm, modified radical hysterectomy with pelvic-node dissection has been recommended because of a reported risk of lymph-node metastasis of as much as 10%.[rx] Radical hysterectomy with node dissection may also be considered for patients for whom the depth of tumor invasion was uncertain because of invasive tumor at the cone margins.
Evidence (open abdominal surgery [open] versus minimally invasive surgery [MIS]):
A multicenter, international, randomized trial, the Laparoscopic Approach to Cervical Cancer (LACC [NCT00614211]) trial explored the efficacy of radical hysterectomy and staging via open abdominal surgery versus MIS for patients with early-stage cervical cancer.[rx] Patients with stages IA1 (with lymphovascular space invasion), IA2, and IB1 disease and histologic subtypes of squamous cell, adenocarcinoma or adenosquamous carcinoma were eligible for inclusion. The primary endpoint was noninferiority of MIS compared with open surgery; the metric utilized was the percent of disease-free patients at 4.5 years postsurgery. The secondary endpoints were a comparison of the recurrence and survival rates between the two groups.
Of the planned 740 patients, 632 were accrued when the study was stopped early because of an imbalance in deaths between the two groups. Of 631 eligible patients, 319 were assigned to MIS and 312 to open surgery.
The disease-free survival (DFS) at 4.5 years was 86% for the MIS group and 96.5% for the open group, a difference of 10.6 percentage points (95% confidence interval [CI], -16.4 to -4.7). At 3 years, the MIS group had a DFS of 91.2% versus 97.1% for the open surgery group (hazard ratio [HR] for disease recurrence or death, 3.74; 95% CI, 1.63–8.58).
The MIS group also had a lower overall survival (OS) rate at 3 years (OS, 93.8% vs. 99.0% for the open surgery group; HR for death from any cause, 6.0; 95% CI, 1.77–20.30).[rx][Level of evidence: 1iiA]
The study concludes that MIS is not noninferior to an open abdominal approach and should not replace open surgery as the standard for cervical cancer patients.
An epidemiologic study utilized two large U.S. databases, the National Cancer Database (NCDB) and the Surveillance, Epidemiology, and End Results Database (SEER), and confirmed a reduction in OS in patients undergoing MIS radical hysterectomy for stage IA2 and stage IB1 cervical cancer from 2010 to 2013. Additionally, among women who underwent radical hysterectomy in the years 2000 to 2010, there was a decrease in OS after 2006, coincident with the widespread adoption of MIS for cervical cancer.[rx][Level of evidence: 3iA]
Although questions remain regarding the use of MIS radical hysterectomy for some subpopulations of good-risk patients, the data from this trial suggest that open abdominal surgery should be considered the standard of care for patients with early-stage cervical cancer who are candidates for radical hysterectomy.
Other Treatment Options
Radical trachelectomy.
Intracavitary radiation therapy.
Radical trachelectomy
Patients with stages IA2 to IB disease who desire future fertility may be candidates for radical trachelectomy. In this procedure, the cervix and lateral parametrial tissues are removed, and the uterine body and ovaries are maintained. Most centers utilize the following criteria for patient selection:
Desire for future pregnancy.
Age younger than 40 years.
Presumed stage IA2 to IB1 disease and a lesion size no greater than 2 cm.
Preoperative magnetic resonance imaging that shows a margin from the most distal edge of the tumor to the lower uterine segment.
Squamous, adenosquamous, or adenocarcinoma cell types.
Intraoperatively, the patient is assessed in a manner similar to a radical hysterectomy; the procedure is aborted if more advanced disease than expected is encountered. The margins of the specimen are also assessed at the time of surgery, and a radical hysterectomy is performed if inadequate margins are obtained.[rx–rx]
Intracavitary radiation therapy
Intracavitary radiation therapy is a treatment option when palliative treatment is appropriate because of other medical conditions and for women who are not surgical candidates.
If the depth of invasion is less than 3 mm and no capillary lymphatic space invasion is noted, and the frequency of lymph-node involvement is sufficiently low, external-beam radiation therapy is not required. One or two insertions with tandem and ovoids for 6,500 mg to 8,000 mg hours (100–125 Gy vaginal surface dose) are recommended.[rx]
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.
Stages IB and IIA Cervical Cancer Treatment
Standard Treatment Options for Stages IB and IIA Cervical Cancer
Standard treatment options for stage IB and stage IIA cervical cancer include the following:
Radiation therapy with concomitant chemotherapy.
Radical hysterectomy and bilateral pelvic lymphadenectomy with or without total pelvic radiation therapy plus chemotherapy.
The size of the tumor is an important prognostic factor and should be carefully evaluated in choosing optimal therapy.[1]
Either radiation therapy or radical hysterectomy and bilateral lymph–node dissection results in cure rates of 85% to 90% for women with Fédération Internationale de Gynécologie et d’Obstétrique (FIGO) stages IA2 and IB1 small-volume disease. The choice of either treatment depends on patient factors and available local expertise. A randomized trial reported identical 5-year overall survival (OS) and disease-free survival (DFS) rates when comparing radiation therapy with radical hysterectomy.[rx]
In stage IB2, for tumors that expand the cervix more than 4 cm, the primary treatment should be concomitant chemotherapy and radiation therapy.[rx]
Radiation therapy with concomitant chemotherapy
Concurrent, cisplatin-based chemotherapy with radiation therapy is the standard of care for women who require radiation therapy for the treatment of cervical cancer.[rx–rx] Radiation therapy protocols for patients with cervical cancer have historically used dosing at two anatomical points, termed point A and point B, to standardize the doses received. Point A is defined as 2 cm from the external os, and 2 cm lateral, relative to the endocervical canal. Point B is also 2 cm from the external os, and 5 cm lateral from the patient midline, relative to the bony pelvis. In general, for smaller tumors, the curative-intent dose for point A is around 70 Gy, whereas for larger tumors, the point A dose may approach 90 Gy.
Evidence (radiation with concomitant chemotherapy):
Three randomized, phase III trials have shown an OS advantage for cisplatin-based therapy given concurrently with radiation therapy,[rx–rx] while one trial that examined this regimen demonstrated no benefit.[rx] The patient populations in these studies included women with FIGO stages IB2 to IVA cervical cancer treated with primary radiation therapy, and women with FIGO stages I to IIA disease who, at the time of primary surgery, were found to have poor prognostic factors, including metastatic disease in pelvic lymph nodes, parametrial disease, and positive surgical margins.
Although the positive trials vary somewhat in terms of the stage of disease, dose of radiation, and schedule of cisplatin and radiation, the trials demonstrate significant survival benefit for this combined approach.
The risk of death from cervical cancer was decreased by 30% to 50% with the use of concurrent chemoradiation therapy.
Other trials have confirmed these findings.[rx,rx]
Brachytherapy
Standard radiation therapy for cervical cancer includes brachytherapy after external-beam radiation therapy (EBRT). Although low-dose rate (LDR) brachytherapy, typically with cesium Cs 137 (137Cs), has been the traditional approach, the use of high-dose rate (HDR) therapy, typically with iridium Ir 192, is rapidly increasing. HDR brachytherapy provides the advantage of eliminating radiation exposure to medical personnel, a shorter treatment time, patient convenience, and improved outpatient management. The American Brachytherapy Society has published guidelines for the use of LDR and HDR brachytherapy as components of cervical cancer treatment.[rx,rx]
Evidence (brachytherapy):
In three randomized trials, HDR brachytherapy was comparable with LDR brachytherapy in terms of local-regional control and complication rates.[rx–rx][Level of evidence: 1iiDii]
Surgery after radiation therapy may be indicated for some patients with tumors confined to the cervix that respond incompletely to radiation therapy or for patients whose vaginal anatomy precludes optimal brachytherapy.[rx]
Pelvic node disease
The resection of macroscopically involved pelvic nodes may improve rates of local control with postoperative radiation therapy.[rx] Patients who underwent extraperitoneal lymph–node sampling had fewer bowel complications than those who had transperitoneal lymph–node sampling.[rx–rx] Patients with close vaginal margins (<0.5 cm) may also benefit from pelvic radiation therapy.[rx]
Radical hysterectomy and bilateral pelvic lymphadenectomy with or without total pelvic radiation therapy plus chemotherapy
Radical hysterectomy and bilateral pelvic lymphadenectomy may be considered for women with stages IB to IIA disease.
Evidence (radical hysterectomy and bilateral pelvic lymphadenectomy with or without total pelvic radiation therapy plus chemotherapy):
An Italian group randomly assigned 343 women with stage IB and IIA cervical cancer to surgery or radiation therapy. The radiation therapy included EBRT and one 137Cs LDR insertion, with a total dose to point A from 70 to 90 Gy (median 76 Gy). Patients in the surgery arm underwent a class III radical hysterectomy, pelvic lymphadenectomy, and selective, para-aortic lymph–node dissection. Adjuvant radiation therapy was given to patients with high-risk pathologic features in the uterine specimen or positive lymph nodes. Adjuvant radiation therapy was EBRT to a total dose of 50.4 Gy over 5 to 6 weeks.[rx][Level of evidence: 1iiA]
The primary outcome was OS at 5 years, with secondary measures of rate of recurrence and complications. With a median follow-up of 87 months, OS was the same in both groups at 83% (hazard ratio [HR], 1.2; confidence interval [CI], 0.7–2.3; P = .8).
Complications were highest among the patients who received adjuvant radiation after surgery.
In general, radical hysterectomy should be avoided in patients who are likely to require adjuvant therapy.
Evidence (open abdominal surgery [open] versus minimally invasive surgery [MIS]):
A multicenter, international, randomized trial, the Laparoscopic Approach to Cervical Cancer (LACC [NCT00614211]) trial explored the efficacy of radical hysterectomy and staging via open abdominal surgery (open) versus MIS for patients with early-stage cervical cancer.[rx] Patients with stages IA1 (with lymphovascular space invasion), IA2, and IB1 disease and histologic subtypes of squamous cell, adenocarcinoma or adenosquamous carcinoma were eligible for inclusion. The primary endpoint was noninferiority of MIS compared with open surgery; the metric utilized was the percent of disease-free patients at 4.5 years postsurgery. The secondary endpoints were a comparison of the recurrence and survival rates between the two groups.
Of the planned 740 patients, 632 were accrued when the study was stopped early because of an imbalance in deaths between the two groups. Of 631 eligible patients, 319 were assigned to MIS and 312 to open surgery.
The DFS at 4.5 years was 86% for the MIS group and 96.5% for the open group, a difference of 10.6 percentage points (95% confidence interval [CI], -16.4 to -4.7). At 3 years, the MIS group had a DFS of 91.2% versus 97.1% for the open surgery group (hazard ratio [HR] for disease recurrence or death, 3.74; 95% CI, 1.63–8.58).
The MIS group also had a lower overall survival (OS) rate at 3 years (OS, 93.8% vs. 99.0% for the open surgery group; HR for death from any cause, 6.0; 95% CI, 1.77– 20.30).[22][Level of evidence: 1iiA]
The study concludes that MIS is not inferior to an open abdominal approach and should not replace open surgery as the standard for cervical cancer patients.
An epidemiologic study utilized two large U.S. databases (National Cancer Database [NCDB] and Surveillance, Epidemiology, and End Results [SEER] database) and confirmed a reduction in OS in patients undergoing MIS radical hysterectomy for stage IA2 and stage IB1 cervical cancer from 2010 to 2013. Additionally, among women who underwent radical hysterectomy in the years 2000 to 2010, there was a decrease in OS after 2006, coincident with the widespread adoption of MIS for cervical cancer.[rx][Levels of evidence: 3iA and 3iiiA]
Although questions remain regarding the use of MIS radical hysterectomy for some subpopulations of good-risk patients, the data from this trial suggest that open abdominal surgery should be considered the standard of care for patients with early-stage cervical cancer who are candidates for radical hysterectomy.
Adjuvant radiation therapy postsurgery
Based on recurrence rates in previous clinical trials, two classes of recurrence risk have been defined. Patients with a combination of large tumor size, lymph vascular space invasion, and deep stromal invasion in the hysterectomy specimen are deemed to have intermediate-risk disease. These patients are candidates for adjuvant EBRT.[rx] Patients whose pathology shows positive margins, positive parametria, or positive lymph nodes are high-risk candidates for recurrence.
The Gynecologic Oncology Group (GOG) compared adjuvant radiation therapy alone with radiation therapy plus cisplatin plus fluorouracil (5-FU) after radical hysterectomy for patients in the high-risk group. Postoperative patients were eligible if their pathology showed any one of the following: positive parametria, positive margins, or positive lymph nodes. Patients in both arms received 49 Gy to the pelvis. Patients in the experimental arm also received cisplatin (70 mg/m2) and a 96-hour infusion of 5-FU (1000 mg/m2/d every 3 weeks for four cycles); the first two cycles were concurrent with the radiation therapy.[rx][Level of evidence: 1iiA]
There were 268 patients evaluated with a primary endpoint of OS. The study results were reported early because of the positive results in other trials of concomitant cisplatin and radiation therapy.
Estimated 4-year survival was 81% for chemotherapy plus radiation therapy and 71% for radiation therapy alone (HR, 1.96; P = .007).
As expected, grade 4 toxicity was more common in the chemotherapy plus radiation therapy group, with hematologic toxicity predominating.
Radical surgery has been performed for small lesions, but the high incidence of pathologic factors leading to postoperative radiation with or without chemotherapy make primary concomitant chemotherapy and radiation a more common approach in patients with larger tumors. Radiation in the range of 50 Gy administered for 5 weeks plus chemotherapy with cisplatin with or without 5-FU should be considered in patients with a high risk of recurrence.
Para-aortic nodal disease
After surgical staging, patients found to have small-volume para-aortic nodal disease and controllable pelvic disease may be cured with pelvic and para-aortic radiation therapy.[rx] Treatment of patients with unresected para-aortic nodes with extended-field radiation therapy and chemotherapy leads to long-term disease control in patients with low-volume (<2 cm) nodal disease below L3.[rx] A single study (RTOG-7920) showed a survival advantage in patients with tumors larger than 4 cm who received radiation therapy to para-aortic nodes without histologic evidence of disease.[rx] Toxic effects were greater with para-aortic radiation therapy than with pelvic radiation therapy alone but were mostly confined to patients with previous abdominopelvic surgery.[rx] The use of intensity-modulated radiation therapy (IMRT) may minimize the effects to the small bowel usually associated with this treatment.[rx]
Other Treatment Options
Radical trachelectomy.
Neoadjuvant chemotherapy.
Radiation therapy alone.
IMRT.
Radical trachelectomy
Patients with presumed early-stage disease who desire future fertility may be candidates for radical trachelectomy. In this procedure, the cervix and lateral parametrial tissues are removed, and the uterine body and ovaries are maintained. The patient selection differs somewhat between groups; however, general criteria include the following:
Desire for future pregnancy.
Age younger than 40 years.
Presumed stage IA2 to IB1 disease and a lesion size no greater than 2 cm.
Preoperative magnetic resonance imaging that shows a margin from the most distal edge of the tumor to the lower uterine segment.
Squamous, adenosquamous, or adenocarcinoma cell types.
Intraoperatively, the patient is assessed in a manner similar to a radical hysterectomy; the procedure is aborted if more advanced disease than expected is encountered. The margins of the specimen are also assessed at the time of surgery, and a radical hysterectomy is performed if inadequate margins are obtained.[rx–rx]
Neoadjuvant chemotherapy
Several groups have investigated the role of neoadjuvant chemotherapy to convert patients who are conventional candidates for chemoradiation into candidates for radical surgery.[rx–rxrx] Multiple regimens have been used; however, almost all utilize a platinum backbone. The largest randomized trial to date was reported in 2001, and its accrual was completed before the standard of care included the addition of cisplatin to radiation therapy.[rx] As a result, the control arm utilized radiation therapy alone. Although there was an improvement in OS for the experimental arm, the results are not reflective of current practice. This study accrued patients with stages IB through IVA disease, but improvement in the experimental arm was only noted for participants with early stage disease (stages IB, IIA, or IIB).
EORTC-55994 (NCT00039338) randomly assigned patients with stages IB2, IIA2, and IIB cervical cancer to standard chemoradiation or neoadjuvant chemotherapy (with a cisplatin backbone for three cycles) followed by evaluation for surgery. With OS as the primary endpoint, this trial may delineate whether there is a role for neoadjuvant chemotherapy for this patient population.
Radiation therapy alone
External-beam pelvic radiation therapy combined with two or more intracavitary brachytherapy applications is appropriate therapy for patients with stage IA2 and IB1 lesions. For patients with stage IB2 and larger lesions, radiosensitizing chemotherapy is indicated. The role of radiosensitizing chemotherapy in patients with stage IA2 and IB1 lesions is untested. However, it may prove beneficial in certain cases.
IMRT
IMRT is a radiation therapy technique that allows for conformal dosing of target anatomy while sparing neighboring tissue. Theoretically, this technique should decrease radiation therapy–related toxicity, but this could come at the cost of decreased efficacy if tissue is inappropriately excluded from the treatment field. Several institutions have reported their experience with IMRT for postoperative adjuvant therapy in patients with intermediate-risk and high-risk disease after radical surgery.[rx–rx] The Radiation Therapy Oncology Group (RTOG) has closed accrual for a phase II trial (RTOG-0418 [NCT00331760]) that is evaluating the use of IMRT in patients with both cervical and endometrial cancers who require adjuvant radiation 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.
Stages IIB, III, and IVA Cervical Cancer Treatment
Standard Treatment Options for Stages IIB, III, and IVA Cervical Cancer
The size of the primary tumor is an important prognostic factor and should be carefully evaluated in choosing optimal therapy.[rx] Survival and local control are better with unilateral rather than bilateral parametrial involvement.[rx] Patterns-of-care studies in stages IIIA and IIIB patients indicate that survival is dependent on the extent of the disease, with unilateral pelvic wall involvement predicting a better outcome than bilateral involvement, which in turn predicts a better outcome than involvement of the lower third of the vaginal wall.[rx] These studies also reveal a progressive increase in local control and survival paralleling a progressive increase in paracentral (point A) dose and use of intracavitary treatment. The highest rate of central control was seen with paracentral (point A) doses of more than 85 Gy.[rx]
Strong consideration should be given to the use of intracavitary radiation therapy and external-beam radiation therapy (EBRT) to the pelvis combined with cisplatin or cisplatin/fluorouracil (5-FU).[rx–rx]
Evidence (radiation therapy with concomitant chemotherapy):
Five randomized, phase III trials have shown an overall survival (OS) advantage for cisplatin-based therapy given concurrently with radiation therapy,[rx–rx] but one trial that examined this regimen demonstrated no benefit.[rx] The patient populations in these studies included women with Fédération Internationale de Gynécologie et d’Obstétrique (FIGO) stages IB2 to IVA cervical cancer treated with primary radiation therapy, and women with FIGO stages I to IIA disease who, at the time of primary surgery, were found to have poor prognostic factors, including metastatic disease in pelvic lymph nodes, parametrial disease, and positive surgical margins.
Although the positive trials vary somewhat in terms of the stage of disease, dose of radiation, and schedule of cisplatin and radiation, the trials demonstrate significant survival benefit for this combined approach.
The risk of death from cervical cancer was decreased by 30% to 50% with the use of concurrent chemoradiation therapy.
Evidence (low-dose rate vs. high-dose rate intracavitary radiation therapy):
Although low-dose rate (LDR) brachytherapy, typically with cesium Cs 137, has been the traditional approach, the use of high-dose rate (HDR) therapy, typically with iridium Ir 192, is rapidly increasing. HDR brachytherapy provides the advantage of eliminating radiation exposure to medical personnel, a shorter treatment time, patient convenience, and improved outpatient management. The American Brachytherapy Society has published guidelines for the use of LDR and HDR brachytherapy as a component of cervical cancer treatment.[rx,rx]
In three randomized trials, HDR brachytherapy was comparable with LDR brachytherapy in terms of local-regional control and complication rates.[rx–rx][Level of evidence: 1iiDii]
In an attempt to improve upon standard chemoradiation, a phase III randomized trial compared concurrent gemcitabine plus cisplatin and radiation therapy followed by adjuvant gemcitabine and cisplatin (experimental arm) with concurrent cisplatin plus radiation (standard chemoradiation) in patients with stages IIB to IVA cervical cancer.[rx][Level of evidence: 1iiA] A total of 515 patients from nine countries were enrolled. The schedule for the experimental arm was cisplatin (40 mg/m2) and gemcitabine (125 mg/m2) weekly for 6 weeks with concurrent EBRT (50.4 Gy in 28 fractions) followed by brachytherapy (30–35 Gy in 96 hours) and then two adjuvant 21-day cycles of cisplatin (50 mg/m2) on day 1 plus gemcitabine (1,000 mg/m2) on days 1 and 8. The standard arm was cisplatin (40 mg/m2) weekly for 6 weeks with concurrent EBRT and brachytherapy as described for the experimental arm.
The primary endpoint was progression-free survival (PFS) at 3 years; however, the study found improvement in the experimental arm for PFS at 3 years (74.4%; 95% confidence interval [CI], 68%–79.8% vs. 65.0%; 95% CI, 58.5%–70.7%); overall PFS (hazard ratio [HR], 0.68; 95% CI, 0.49–0.95); and OS (HR, 0.68; 95% CI, 0.49–0.95). Patients in the experimental arm had increased hematologic and nonhematologic grade 3 or 4 toxic effects, and two deaths in the experimental arm were possibly related to treatment.
A subgroup analysis showed an increased benefit in patients with a higher stage of disease (stages III–IVA vs. stage IIB), which suggested that the increased toxic effects of the experimental protocol may be justified for these patients.[rx] Additional investigation is needed to determine which aspect of the experimental arm led to improved survival (i.e., the addition of the weekly gemcitabine, the adjuvant chemotherapy, or both) and to determine quality of life during and after treatment, a condition that was omitted from the protocol.
The addition of adjuvant chemotherapy following chemoradiation therapy is currently being evaluated as part of a large multinational clinical trial. The OUTBACK trial (NCT01414608) is randomly assigning women to receive cisplatin (40 mg/m2 weekly for 5 doses) with whole-pelvic radiation therapy (standard chemoradiation therapy) with or without standard chemoradiation therapy plus adjuvant carboplatin (AUC 5 + paclitaxel 155 mg/m2).
Lymph Node Management
Patients who are surgically staged as part of a clinical trial and are found to have small-volume para-aortic nodal disease and controllable pelvic disease may be cured with pelvic and para-aortic radiation therapy.[rx] Treatment of patients with unresected periaortic nodes with extended-field radiation therapy leads to long-term disease control in patients with low-volume (<2 cm) nodal disease below L3.[rx] A single study (RTOG-7920) showed a survival advantage in patients who received radiation therapy to para-aortic nodes without histologic evidence of disease.[rx] Toxic effects are greater with para-aortic radiation than with pelvic radiation alone but were mostly confined to patients with previous abdominopelvic surgery.[rx]
If postoperative EBRT is planned following surgery, extraperitoneal lymph–node sampling is associated with fewer radiation-induced complications than a transperitoneal approach.[rx] Patients who underwent extraperitoneal lymph–node sampling had fewer bowel complications than those who had transperitoneal lymph–node sampling.[rx,rx,rx]
The resection of macroscopically involved pelvic nodes may improve rates of local control with postoperative radiation therapy.[rx] In addition, prospective data points to improvement in outcomes for patients who undergo resection of positive para-aortic lymph nodes before curative intent chemoradiation therapy; however, only patients with minimal nodal involvement (<5mm) benefited.[rx]
For patients who complete EBRT and have bulky cervical disease such that standard brachytherapy cannot be placed anatomically, interstitial brachytherapy has been used to deliver adequate tumoricidal doses with an acceptable toxicity profile.[28]
Neoadjuvant chemotherapy
Several groups have investigated the role of neoadjuvant chemotherapy to convert patients who are conventional candidates for chemoradiation into candidates for radical surgery.[rx–rx] Multiple regimens have been used; however, almost all utilize a platinum backbone. The largest randomized trial to date was reported in 2001, and its accrual was completed before the standard of care included the addition of cisplatin to radiation therapy.[rx] As a result, although there was an improvement in OS for the experimental arm, the results are not reflective of current practice. This study accrued patients with stages IB through IVA disease, but improvement in the experimental arm was only noted for participants with early stage disease (stages IB, IIA, or IIB).
EORTC-55994 (NCT00039338) randomly assigned patients with stages IB2, IIA2, and IIB cervical cancer to standard chemoradiation or neoadjuvant chemotherapy (with a cisplatin backbone for three cycles) followed by evaluation for surgery. With OS as the primary endpoint, this trial may delineate whether there is a role for neoadjuvant chemotherapy for this patient population.
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. Stage IVB Cervical Cancer Treatment
Standard Treatment Options for Stage IVB Cervical Cancer
Standard treatment options for stage IVB cervical cancer include the following:
Single-agent cisplatin administered intravenously at 50 mg/m² every 3 weeks has been the regimen most often used to treat recurrent cervical cancer since the drug was initially introduced in the 1970’s.[rx] More recently, the GOG has reported on sequential randomized trials dealing with combination chemotherapy for stages IVB, recurrent, or persistent cervical cancer.[rx,rx–rx]
Evidence (cisplatin in combination with other drugs):
GOG-110: The ifosfamide + cisplatin combination was superior to cisplatin alone in the secondary endpoint of response rates, but at the cost of increased toxicity.
GOG-0179: The cisplatin + topotecan (CT) doublet combination had a significant advantage in overall survival (OS) compared with cisplatin alone, leading to approval of this indication for topotecan by the U.S. Food and Drug Administration. However, cisplatin alone underperformed in this trial because as many as 40% of the patients had already received cisplatin up front as a radiosensitizer.[rx]
GOG-0169: The paclitaxel + cisplatin (PC) combination, similarly, was superior in response rates and progression-free survival (PFS), and its toxicity was similar to that of the single agent except in patients with GOG performance status 2 (scale: 0, asymptomatic–4, totally bedridden). Therefore, PC was chosen as the reference arm in GOG-0204 (NCT00064077).
GOG-0204 enrolled 513 patients and compared four cisplatin-based doublet regimens. The trial was closed early because no one experimental arm was likely to significantly lower the hazard ratio of death relative to PC:[rx]
1.32 (95% CI, 0.91–1.92) for gemcitabine plus cisplatin.
1.27 (95% CI, 0.90–1.78) for CT. Trend in response rates, PFS, and OS favored CT.
The patients in the various arms of the study differed in the extent of neutropenia, infection, and alopecia that they experienced,[rx] but none of the patients in the study arms differed in health-related quality of life during treatment.[rx] However, there were more neurologic side effects for PC.
GOG-0240 (NCT00803062) was designed to answer the following two questions:[rx]
Can a nonplatinum combination show improvement over the standard of cisplatin-paclitaxel in this population that was previously treated with cisplatin during radiation therapy?
Can the addition of bevacizumab improve combination chemotherapy in patients with stages IVB, persistent, or recurrent cervical cancer?
Patients were randomly assigned to the following four treatment arms:
Cisplatin (50 mg/m2) + paclitaxel (135 mg/m2 or 175 mg/m2) on day 1 (PC).
Additional study methods and results included the following:
The primary endpoint was OS, and 452 patients were evaluable.
The combination PT was not superior to PC and had a hazard ratio (HR) for death of 1.2 (99% CI, 0.82–1.76). Previous exposure to platinum did not affect this result.
The addition of bevacizumab to combination chemotherapy led to an improvement in OS: 17 months for chemotherapy plus bevacizumab versus 13.3 months for chemotherapy alone (HR, 0.71; 98% CI, 0.54–0.95), and extended PFS: 8.2 months for chemotherapy plus bevacizumab versus 5.9 months for chemotherapy alone, (HR, 0.67; 95% CI, 0.54–0.82).
The addition of bevacizumab was well tolerated and showed no difference in quality of life between the two groups.
Patients on bevacizumab were more likely to have grade 3 or higher fistulae (6% vs. 0%), and grade 3 or higher thromboembolic events (8% vs. 1%) compared with patients on chemotherapy alone.
As a result, the addition of bevacizumab may be considered for this patient population.
Treatment Options Under Clinical Evaluation for Stage IVB Cervical Cancer
New anticancer drugs in phase I and phase II clinical trials.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
Recurrent Cervical Cancer Treatment
Treatment Options for Recurrent Cervical Cancer
With the exception of immunotherapy, which has provided prolonged disease-free survival, other options are unlikely to result in curative outcomes and are mostly applied for palliative purposes.
Treatment options for recurrent cervical cancer include the following:
Immunotherapy.
Radiation therapy and chemotherapy.
Palliative chemotherapy and other systemic therapy.
Pelvic exenteration.
Immunotherapy
Pembrolizumab
Favorable experience with the anti–programmed cell death-1 (PD-1) immune checkpoint inhibitor, pembrolizumab, has led to U.S. Food and Drug Administration (FDA) approval based on the phase II KEYNOTE-158 (NCT02628067) trial for women with recurrent or metastatic cervical cancer whose tumors express greater than or equal to 1 programmed death-ligand (PD-L1) (combined positive score [CPS], >1).
KEYNOTE-028 (NCT02054806) was an earlier phase 1b expansion cohort of the anti-PD-1 immune checkpoint inhibitor, pembrolizumab, that included 24 patients, all but 1 with squamous cell carcinoma.[1]
The overall response rate was 17% (95% CI, 5%–37%), with 4 patients achieving a confirmed partial response.
Treatment-related adverse events that were considered to be related to the study drug were observed in 18 patients, with only rash (n = 5; 21%) and pyrexia (n = 4; 17%) occurring in 10% or more of the patients.
Grade 3 treatment-related adverse events were experienced by 5 patients and included neutropenia, rash, colitis, Guillain-Barre syndrome, and proteinuria.
The phase II findings in the preliminary analysis of 47 patients that led to the FDA approval of pembrolizumab were presented in abstract form;[2] these have been updated in Keytruda’s package insertExit Disclaimer
KEYNOTE-158 (NCT02628067) was a multicenter, nonrandomized trial that entered 98 patients with recurrent or metastatic cervical cancer who received 200 mg every 3 weeks intravenously until there was unacceptable toxicity or disease progression.[3] A separate analysis was performed in 77 patients whose tumors expressed PD-L1 (CPS >1); 92% had squamous histology.
The overall response rate among PD-L1–positive patients was 16% (95% CI, 8.8%–25.9%) with 3 complete responses and 10 partial responses; 17 patients were stable.
Median progression-free survival was 2.1 months and overall survival was 9.4 months in these marker-positive patients.
Treatment-related adverse events were noted in 65% of patients; the most common were hypothyroidism (10.2%), decreased appetite (9.2%), fatigue (9.2%), and diarrhea 8.2%.[3][Level of evidence: 2Diii]
The experience in a case series of 11 patients that showed 2 patients with partial responses and 2 patients with disease stabilization associated with pembrolizumab treatment has been published.[4]
In the CheckMate 358Exit Disclaimer (NCT02488759) trial, presented in abstract form, nivolumab (240 mg IV q 2 weeks) was tested in 19 cervical cancer patients and 5 patients with other virus-associated tumors of vaginal and vulvar origins.[5]
One complete and four partial responses were noted among cervical cancer patients; the median PFS was 5.5 months.
Radiation therapy and chemotherapy
For recurrence in the pelvis after initial radical surgery, radiation therapy and chemotherapy (fluorouracil with or without mitomycin) may cure 40% to 50% of patients.[rx]
Palliative chemotherapy and other systemic therapy
Chemotherapy can be used for palliation. Drugs used for palliative chemotherapy are shown in Table 7.
Table 7. Drugs Used to Treat Recurrent Cervical Cancer
Single-agent cisplatin administered intravenously at 50 mg/m² every 3 weeks has been the regimen most often used to treat recurrent cervical cancer since the drug was initially introduced in the 1970’s.[7] More recently, the GOG has reported on sequential randomized trials dealing with combination chemotherapy for stage IVB, recurrent, or persistent cervical cancer.[rx,rx,rx–rx]
Evidence (cisplatin in combination with other drugs):
GOG-110: The ifosfamide + cisplatin combination was superior to cisplatin alone in the secondary endpoint of response rates, but at the cost of increased toxicity.
GOG-0179: The cisplatin + topotecan (CT) doublet combination had a significant advantage in OS compared with cisplatin alone, leading to approval of this indication for topotecan by the U.S. Food and Drug Administration. However, cisplatin alone underperformed in this trial because as many as 40% of the patients had already received cisplatin up front as a radiosensitizer.[rx]
GOG-0169: The paclitaxel + cisplatin (PC) combination, similarly, was superior in response rates and PFS, and its toxicity was similar to that of the single agent except in patients with GOG performance status 2 (scale: 0, asymptomatic–4, totally bedridden). Therefore, paclitaxel plus cisplatin (PC) was chosen as the reference arm in GOG-0204 (NCT00064077).
GOG-0204 enrolled 513 patients and compared four cisplatin-based doublet regimens. The trial was closed early because no one experimental arm was likely to significantly lower the hazard ratio of death relative to PC:[rx]
1.32 (95% CI, 0.91–1.92) for gemcitabine plus cisplatin.
1.27 (95% CI, 0.90–1.78) for CT. Trend in RR, PFS, and OS favored CT.
The patients in the various arms of the study differed in the extent of neutropenia, infection, and alopecia that they experienced,[rx] but none of the patients in the study arms differed in health-related quality of life during treatment.[rx] However, there were more neurologic side effects for PC.
GOG-0240 (NCT00803062) was designed to answer the following two questions:[rx]
Can a nonplatinum combination show improvement over the standard of cisplatin-paclitaxel in this population previously treated with cisplatin during radiation therapy?
Can the addition of bevacizumab improve upon combination chemotherapy in patients with stage IVB, persistent or recurrent cervical cancer?
Patients were randomly assigned to the following four treatment arms:
Cisplatin (50 mg/m2) + paclitaxel (135 mg/m2 or 175 mg/m2) on day 1 (PC).
PC + bevacizumab (15mg/kg) on day 1.
Topotecan (0.75 mg/m2) d1–d3 + paclitaxel (175 mg/m2) on day 1 (PT).
PT + bevacizumab (15mg/kg) on day 1.
Additional study methods and results included the following:
The primary endpoint was OS, and 452 patients were evaluable.
The combination PT was not superior to PC and had a hazard ratio (HR) for death of 1.2 (99% CI, 0.82–1.76). Previous exposure to platinum did not affect this result.
The addition of bevacizumab to combination chemotherapy led to an improvement in OS: 17 months for chemotherapy plus bevacizumab versus 13.3 months for chemotherapy alone (HR, 0.71; 98% CI, 0.54–0.95), and extended PFS: 8.2 months for chemotherapy plus bevacizumab versus 5.9 months for chemotherapy alone, HR, 0.67; (95% CI, 0.54–0.82).
The addition of bevacizumab was well tolerated and showed no difference in quality of life between the two groups.
Patients on bevacizumab were more likely to have grade 3 or higher fistulae (6% vs. 0%), and grade 3 or higher thromboembolic events (8% vs. 1%) compared with patients on chemotherapy alone.
As a result, the addition of bevacizumab may be considered for this patient population.
Pelvic exenteration
No standard treatment is available for patients with recurrent cervical cancer that has spread beyond the confines of a radiation or surgical field. For locally recurrent disease, pelvic exenteration can lead to a 5-year survival rate of 32% to 62% in selected patients.[rx,rx] These patients are appropriate candidates for clinical trials testing drug combinations or new anticancer agents.
Treatment Options Under Clinical Evaluation for Recurrent Cervical Cancer
Treatment options under clinical evaluation for recurrent cervical cancer include the following:
New anticancer drugs in phase I and phase II clinical trials.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria.
Cervical Cancer During Pregnancy
During pregnancy, no therapy is warranted for preinvasive lesions of the cervix, including carcinoma in situ, although expert colposcopy is recommended to exclude invasive cancer.
Diagnosis
Treatment of cervical cancer in pregnancy is predicated on the extent of disease and the gestational age at diagnosis. Patients should undergo biopsy as needed and imaging to establish the extent of disease to make the most informed choices. The most appropriate imaging modality in pregnancy is magnetic resonance imaging, when indicated.
Treatment for Stage I Disease
Pregnancy does not alter the course of cervical cancer. As a result, in certain cases, patients may elect to postpone treatment until its effects on the pregnancy are minimized. This may be considered for patients with the more common, and less aggressive histologic subtypes: squamous, adenocarcinoma, and adenosquamous. Patients with high-risk subtypes, such as small cell or neuroendocrine tumors, should be counseled toward immediate treatment despite the effects on the fetus, given their risk of progression.
Patients with early stage (IA) disease may safely undergo fertility-sparing treatments including cervical conization or radical trachelectomy, as indicated. The optimal timing for this procedure is in the second trimester, before viability. Some authors have suggested waiting until the completion of a pregnancy to initiate treatment.[rx] For patients with IA2 and IB disease such a delay may also be safe, but because of a risk of lymphatic spread, assessment of lymph-node status should first be ascertained. The status is best determined surgically via a laparoscopic or open lymph-node dissection, which can be safely performed up to approximately 20 weeks of pregnancy.[rx,rx] In patients without lymphatic spread, waiting for fetal viability to initiate treatment is an option. Patients with positive lymph nodes should be counseled toward immediate treatment.
Treatment for Stages II, III, and IV Disease
For patients with stage II or greater disease, waiting for viability is generally not acceptable.[rx] The standard of care is curative intent chemotherapy and radiation therapy. This treatment is toxic to the fetus and without ovarian transposition will render the ovaries nonfunctional after treatment. Evacuation of the fetus should be performed before the initiation of radiation. When this is not possible, the radiation will generally cause a spontaneous abortion 3 to 5 weeks after initiating treatment.
Neoadjuvant Chemotherapy
Neoadjuvant chemotherapy has been offered to patients with locally advanced disease as a way to initiate treatment while maintaining the pregnancy.[rx] Most chemotherapy agents can be initiated safely in the second trimester of pregnancy and beyond; mild growth restriction of the fetus is the most common side effect. Restriction of growth has been reported in a relatively small number of patients, and data is lacking on long-term outcomes for these women; as a result, this strategy should be considered with caution. Most of the patients in the reports underwent standard treatment (either surgery or radiation) after the completion of the pregnancy.
Adenomyosis is a uterine condition that is histologically characterized by the presence of ectopic endometrial glands and stroma within the myometrium, surrounded by hypertrophic and hyperplastic myometrial changes [rx]. For several decades, the diagnosis of adenomyosis was made in hysterectomy specimens either coincidentally, or in women treated surgically for chronic pelvic pain and/or abnormal uterine bleeding[rx].
Adenomyosis is a medical condition characterized by the growth of cells that build up the inside of the uterus (endometrium) atypically located within the cells that put up the uterine wall (myometrium),[rx] as a result, thickening of the uterus occurs. As well as being misplaced in patients with this condition, endometrial tissue is completely functional. The tissue thickens, sheds, and bleeds during every menstrual cycle.[rx]
Causes of Adenomyosis
Adenomyosis and endometriosis are usually regarded as closely related, but
Microscopic appearance, and probably their pathogenesis, are somewhat different
They may occur independently of each other
Adenomyosis mostly is made up of nonfunctional (basal) endometrium and is frequently connected with the mucosa (vs. endometriosis, composed of functional layers)
Adenomyosis may represent a unique form of endometrial diverticulosis
In situ metaplasia of pluripotent stem cells retained in myometrium or
Improper partitioning of the endometrium from the myometrium
Of note, del(7) (q21.2q31.2), a deletion found in typical leiomyoma, has been found in three cases of adenomyosis, suggesting some pathobiology overlap between leiomyomata and adenomyosis (Cancer Genet Cytogenet 1995;80:118)
Invasive tissue growth – Some experts believe that endometrial cells from the lining of the uterus invade the muscle that forms the uterine walls. Uterine incisions made during an operation such as a cesarean section (C-section) might promote the direct invasion of the endometrial cells into the wall of the uterus.
Developmental origins – Other experts suspect that endometrial tissue is deposited in the uterine muscle when the uterus is first formed in the fetus.
Uterine inflammation related to childbirth – Another theory suggests a link between adenomyosis and childbirth. Inflammation of the uterine lining during the postpartum period might cause a break in the normal boundary of cells that line the uterus.
Stem cell origins – A recent theory proposes that bone marrow stem cells might invade the uterine muscle, causing adenomyosis.
Symptoms of Adenomyosis
Adenomyosis can vary widely in the type and severity of symptoms that it causes, ranging from being entirely asymptomatic 33% of the time to being a severe and debilitating condition in some cases. Women with adenomyosis typically first report symptoms when they are between 40 and 50, but symptoms can occur in younger women.[rx][rx]
Symptoms and the estimated percent affected may include:[rx]
Chronic pelvic pain (77%)
Heavy menstrual bleeding (40-60%), which is more common in women with deeper adenomyosis. Blood loss may be significant enough to cause anemia, with associated symptoms of fatigue, dizziness, and moodiness.
Abnormal uterine bleeding
Painful cramping menstruation (15-30%)
Painful vaginal intercourse (7%)
A ‘bearing’ down feeling
Pressure on bladder
Dragging sensation down thighs and legs
Uterine enlargement (30%), which in turn can lead to symptoms of pelvic fullness.
Tender uterus
Infertility or sub-fertility (11-12%) – In addition, adenomyosis is associated with an increased incidence of preterm labor and premature rupture of membranes.[rx][rx]
Women with adenomyosis are also more likely to have other uterine conditions, including:
Uterine fibroids (50%)
Endometriosis (11%)
Endometrial polyp (7%)
Nonneoplastic condition presenting with palpably enlarged uterus
When extensive, it confers a potential risk of infarction and thrombosis and exacerbates menorrhagia via activation of coagulation and fibrinolysis during menstruation (Eur J Obstet Gynecol Reprod Biol 2016;204:99)
Painful menstrual cramps (dysmenorrhea).
Heavy menstrual bleeding (menorrhagia).
Abnormal menstruation.
Pelvic pain.
Painful intercourse (dyspareunia).
Infertility.
Enlarged uterus.
Diagnosis of Adenomyosis
Histopathology
The diagnosis of adenomyosis is through a pathologist microscopically examining small tissue samples of the uterus.[rx] These tissue samples can come from a uterine biopsy or directly following a hysterectomy. Uterine biopsies can be obtained by either a laparoscopic procedure through the abdomen or hysteroscopy through the vagina and cervix.[rx]
The diagnosis is established when the pathologist finds invading clusters of endometrial tissue within the myometrium. Several diagnostic criteria can be used, but typically they require either the endometrial tissue to have invaded greater than 2% of the myometrium, or a minimum invasion depth between 2.5 to 8mm.[rx] Histopathological image of uterine adenomyosis observed in the hysterectomy specimen. Hematoxylin & eosin stain.
Gross Findings
Enlarged uterus
Thickened uterine wall with a trabeculated appearance
Hemorrhagic pinpoint or cystic spaces throughout wall[rx]
Microscopic Findings
Endometrial glands and stroma haphazardly distributed throughout the myometrium
Concentric myometrial hyperplasia frequent around adenomyotic foci
Laboratory testing is useful to rule out other disease entities included in the differential diagnosis, in addition to identifying certain complicating features such as anemia due to heavy menstruation. While some biomarkers due exist, none are specific for adenomyosis.[rx]
Imaging
Adenomyosis can vary widely in the extent and location of its invasion within the uterus. As a result, there are no established pathognomonic features to allow for a definitive diagnosis of adenomyosis through non-invasive imaging. Nevertheless, non-invasive imaging techniques such as transvaginal ultrasonography (TVUS) and magnetic resonance imaging (MRI) can both be used to strongly suggest the diagnosis of adenomyosis, guide treatment options, and monitor response to treatment.[rx] Indeed, TVUS and MRI are the only two practical means available to establish a pre-surgical diagnosis.[rx]
Ultrasound
Transvaginal ultrasound is the preferred diagnostic imaging modality for adenomyosis. The characteristic findings reflect the histopathologic changes of the disease process and can be broken down into three categories:
Endometrial infiltration – echogenic striations and nodules, myometrial cysts, and “lollipop” diverticula (cystic striations)
Smooth muscle proliferation – focal or diffuse myometrial thickening with indistinct borders more commonly involving the posterior fundus and heterogenous echotexture manifesting as “Venetian blind” appearance of thin linear shadows
Vascularity – color Doppler demonstrating an increased number of tortuous vessels throughout the involved myometrium as opposed to leiomyomas which displace vessels
A number of mimics can have similar findings on the ultrasound exam, including tamoxifen use, prior endometrial ablation, endometriosis, uterine contractions, vascular malformations, leiomyomas, and cancer. Certain techniques such as low-frequency, coronal reconstructions, 3-D ultrasound, cine-clips, color Doppler, and saline infusion sonohysterography (SIS) can be used to differentiate between these entities.
MRI
Characteristic findings on MRI parallel the same features seen on ultrasound[rx][rx]:
On T2-weighted imaging, uterine enlargement characterized by ill-defined, low-signal-intensity regions within the junctional zone is reflective of smooth muscle hyperplasia (junctional zone thicker than 12mm is generally accepted as diagnostic)
T2 hyperintense myometrial cysts reflecting regions of ectopic endometrial tissue (can also have increased intrinsic T1 signal or increased susceptibility in hemorrhagic foci)
Contrast enhancement is generally not reliable for assessment of vascularity as compared to a color Doppler ultrasound
Similar to ultrasound, a variety of mimics ranging from co-existing gynecologic pathologies to physiologic variants exist. Susceptibility weighted imaging, diffusion-weighted imaging, MR spectroscopy, cine MR imaging, and increased 3T field strength are all problem-solving strategies.[rx]
It is important to obtain the MRI in the late proliferative or secretory phase (days 7 to 28) due to the decreased signal of normal myometrium during the early proliferative phase (days 1 to 6).[rx]
Transvaginal ultrasonography
Transvaginal ultrasound of the uterus, showing the endometrium as a hyperechoic (brighter) area in the middle, with linear striations extending upwards from it.
Transvaginal ultrasonography is a cheap and readily available imaging test that is typically used early during the evaluation of gynecologic symptoms.[rx] Ultrasound imaging, like MRI, does not use radiation and is safe for the examination of the pelvis and female reproductive organs.[rx] Overall, it is estimated that transvaginal ultrasonography has a sensitivity of 79% and specificity of 85% for the detection of adenomyosis.[rx]
Common transvaginal ultrasound findings in patients with adenomyosis include the following:[rx][rx][rx]
globular, enlarged, and/or asymmetric uterus
abnormally dense or especially varied density within the myometrium
myometrial cysts – pockets of fluid within the smooth muscle of the uterus
linear, acoustic shadowing without the presence of a uterine fibroid
echogenic linear striations – bright lines or stripes
anterior/posterior wall asymmetry
the diffuse spread of small vessels within the myometrium
Lack of contour abnormality
Absence of mass effect
Ill-defined margins between a normal and abnormal myometrium
Others Study
The power Doppler or Doppler ultrasonography function – can be used during transvaginal ultrasonography to help differentiate adenomyomas from uterine fibroids.[rx][rx][rx] This is because uterine fibroids typically have blood vessels circling the fibroid’s capsule. In contrast, adenomyomas are characterized by widespread blood vessels within the lesion.[rx] Doppler ultrasonography also serves to differentiate the static fluid within myometrial cysts from flowing blood within vessels.[rx]
The junction zone (JZ) – or a small distinct hormone-dependent region at the endometrial-myometrial interface, may be assessed by three-dimensional transvaginal ultrasound (3D TVUS) and MRI. Features of adenomyosis are disruption, thickening, enlargement or invasion of the junctional zone.[rx]
Sagittal MRI of a woman’s pelvis – showing a uterus with adenomyosis in the posterior wall. Gross enlargement of the posterior wall is noted, with many foci of hyperintensity.
Shear Wave Elastography – A recent study also showed that using Aixplorer (Supersonic Imagine, France) scanner with the application of shear wave elastography during transvaginal scanning may improve the diagnostic accuracy of adenomyosis [rx]. This study found that adenomyosis was associated with a significant increase of the myometrial stiffness estimated with shear wave elastography. Further studies are required to verify the clinical usefulness of such an approach.
Hysterosalpingography – Hysterosalpingography is seldom used to diagnose adenomyosis. However, in patients undergoing infertility assessment, the occasional finding of speculations measuring 1–4 mm in length, arising from the endometrium towards the myometrium, or a uterus with the “tuba erecta” finding may be suggestive of adenomyosis [rx].
Hysteroscopy – Several hysteroscopic appearances have been found to be associated with adenomyosis, including irregular endometrium with endometrial defects or superficial openings, hypervascularization, strawberry pattern, or cystic hemorrhagic lesions [rx]. Nevertheless, there is limited data available on the diagnostic accuracy of these various features.
Hysteroscopic and Laparoscopic Myometrial Biopsy – The study found that the depth of adenomyosis was correlated with the severity of menorrhagia. Of the 90 patients studied, 50 patients had normal hysteroscopy in which 55% of them had significant adenomyosis (greater than 1 mm) when compared to controls (0.8 mm).
Laparoscopic Myometrial Biopsy – In a prospective, nonrandomized study conducted by Jeng et al. [rx] evaluating 100 patients with clinical signs and symptoms strongly suggestive of adenomyosis, the sensitivity of myometrial biopsy were 98% and the specificity 100%; the positive predictive value was 100% and the negative predictive value 80%, which were superior to those of transvaginal sonography, serum CA-125 determination, or the combination of both. The group suggested that a laparoscopy-guided myometrial biopsy is a valuable tool in the diagnosis of diffuse adenomyosis in women presenting with infertility, dysmenorrhea, or chronic pelvic pain.
Important features in the diagnosis of myoma and adenomyosis FIGO (International Federation of Gynaecology and Obstetrics).
Feature
Typical myoma
Adenomyosis
The serosal contour of the uterus
Lobulated or regular
Globally enlarged uterus
Definition of lesion
Well-defined
Ill-defined in diffuse adenomyosis
(Maybe well-defined in adenomyoma)
The symmetry of uterine wall
Asymmetrical in presence of a well-defined lesion
Myometrial anteroposterior asymmetry
Lesion
Well-defined outline Round/oval/lobulated
Smooth contour
Hypo/hyperechogenic rim
Edge/internal shadow
Uniform (hypo or hyperechogenic)
Non-uniform (mixed echogenicity)
Circumferential flow
Ill-defined outline
Ill-defined shape
Irregular/ill-defined contour
No rim
No edge shadow, fan-shaped shadowing
Non-uniform (mixed echogenicity)
Cysts, hyperechogenic islands
Subendometrial lines and buds
Translesional flow
Junctional zone (JZ)
JZ not thickened (regular or not visible)
Interrupted JZ in areas with lesions types (1-3)
Thickened (irregular or ill-defined)
Interrupted JZ (even in absence of localized lesions)
Treatment of Adenomyosis
Adenomyosis can only be cured definitively with surgical removal of the uterus. As adenomyosis is responsive to reproductive hormones, it reasonably abates following menopause when these hormones decrease. For women in their reproductive years, adenomyosis can typically be managed with the goals to provide pain relief, to restrict progression of the process, and to reduce significant menstrual bleeding.
Medications
NSAIDs – Nonsteroidal anti-inflammatory drugs, such as ibuprofen and naproxen, are commonly used in conjunction with other therapies for pain relief. NSAIDs inhibit the production of prostaglandins by decreasing the activity of the enzyme cyclooxygenase. Prostaglandins have been shown to be primarily responsible for dysmenorrhea or the cramping pelvic pain associated with menses.
Analgesic – Nonsteroidal anti-inflammatory drugs (NSAIDs) work by inhibiting the cyclooxygenase (COX-1 and COX-2) and decreasing the production of prostaglandins. NSAIDs have been proved to be effective in the treatment of primary dysmenorrhea by Gambone et al. [rx]. It is usually the first-line treatment for symptomatic pain relief for adenomyosis.
Oral Contraceptive Pills (OCPs) – Combined oral contraceptive pills work by inhibiting ovulation by suppressing the release of gonadotropins. Many studies have shown that they are effective in the treatment of dysmenorrhea. A prospective observational trial showed that continuous low-dose OCP was more effective than cyclical low-dose OCP in controlling symptoms in patients after surgical treatment for endometriosis [rx].
Danazol – Danazol is an isoxazole derivative of 12 alpha-ethinyl testosterone. It causes a hypogonadal state and thus is widely used for the treatment of endometriosis and abnormal uterine bleeding [rx]. However, data on its use in adenomyosis remains limited. This may be due to its unwanted adverse effects after systemic treatment.
Dienogest – Dienogest is a selective synthetic oral progestin that combines the pharmacological properties of 17-alpha-progesterone and 19 nor-progesterone with a pronounced local effect on endometrial tissue. Dienogest has been shown to be effective in the treatment of endometriosis-associated pelvic pain. A prospective clinical trial has shown dienogest to be a valuable alternative to depot triptorelin acetate for the treatment of premenopausal pelvic pains in women with uterine adenomyosis. [rx]
Levonorgestrel-Releasing Intrauterine Device (LNG-IUD) – LNG-IUD is an intrauterine device, which releases 20 micrograms of levonorgestrel per day. It has been shown to be an effective treatment for abnormal uterine bleeding. LNG-IUD acts locally and causes decidualization of the endometrium and adenomyotic deposits. LNG-IUD alleviates dysmenorrhea by improving uterine contractility and reducing local prostaglandin production within the endometrium. LNG-IUD appears to be an effective method in relieving dysmenorrhea associated with adenomyosis [rx] and more effective than the combined OC pill [rx], improved the quality of life [rx], and appears to be a promising alternative treatment to hysterectomy.
LNG-IUD – may be used in conjunction with other treatment modalities such as GnRH analog [rx] or transcervical resection of the endometrium (TCRE) [rx]. In the latter study, it was found that TCRE combined with LNG-IUD was more effective in reducing menstrual flow compared with the LNG-IUD alone although there was no significant difference in the amount of pain reduction between the two treatment strategies.
GnRH Agonists – GnRH agonists are effective in alleviating dysmenorrhea and relieving menorrhagia associated with adenomyosis [rx]. However, due to the undesirable climacteric side effects and risk of osteoporosis, treatment with GnRH agonists is usually restricted to a short duration of 3–6 months although the duration of use may be extended if add-back estrogen therapy is employed [rx]. Discontinuation of treatment usually leads to regrowth of the lesions and recurrence of symptoms.
Selective Estrogen Receptor Modulator (SERM) – Selective estrogen receptor modulators like tamoxifen or raloxifene have been tried in the treatment of endometriosis [rx] based on observations that SERMs may reduce endometriosis lesion in mouse [rx]; however, their value in the treatment of adenomyoma has not been formally explored.
Aromatase Inhibitors– Like endometriosis, adenomyotic deposits are estrogen-dependent. Aromatase inhibitors inhibit the conversion of estrogen from androgens, thereby lowering the synthesis of estrogen. A prospective randomized controlled study found that the efficacy of aromatase inhibitors (letrozole 2.5 mg/day) in reducing the volume of adenomyoma as well as improving adenomyosis symptoms was similar to that of GnRH agonists (goserelin 3.6 mg/month) [rx] [rx].
Ulipristal Acetate– Ulipristal acetate (UPA) is a potent selective progesterone receptor modulator. There is good evidence to suggest that it can be used to shrink fibroid and control menorrhagia [rx, rx]. It is possible that it may be similarly effective in the treatment of adenomyoma but literature data is lacking.
Antiplatelet Therapy – There is new evidence to suggest the role of antiplatelet therapy in treating adenomyosis. Emerging evidence suggests that endometriotic lesions are wounds undergoing repeated tissue injury and repair (ReTIAR), and platelets induce epithelial-mesenchymal transition (EMT) and fibroblast-to-myofibroblast transdifferentiation (FMT), leading ultimately to fibrosis. Adenomyotic lesions are thought to have similar pathogenesis to that of endometriosis. A recent study in mice suggests that antiplatelet treatment may suppress myometrial infiltration, improve generalized hyperalgesia, and reduce uterine hyperactivity [rx].
Uterine Artery Embolization – Uterine artery embolization (UAE) has been used to treat symptomatic fibroids since the 1990s. There is increasing evidence to suggest that it is also effective in the treatment of the management of adenomyosis. [rx][rx].
High-Intensity Focused Ultrasound – High intensity focused ultrasound (HIFU) is another nonsurgical treatment for uterine fibroids that focuses high-intensity ultrasound on the target lesion causing coagulative necrosis and shrinkage of the lesion. Both MRI and USG can be used for guidance for the procedure. MRI has better real-time thermal mapping during the HIFU treatment. [rx, rx] [rx].
Endomyometrial Ablation or Resection – There is a limited report on the use of laparoscopic or hysteroscopic endometrial in treating adenomyosis in the literature. The success rate of myometrial electrocoagulation ranges from 55 to 70% as reported [rx]. [rx]MRI treated with laparoscopic bipolar coagulation, having significant reduction or the resolution of dysmenorrhea or heavy menstrual bleeding.
GnRH Analogue Therapy before In Vitro Fertilization – Several studies have shown that pretreatment with GnRH analog before IVF treatment improved pregnancy outcome. Zhou et al. [rx] analyzed the clinical efficacy of leuprorelin acetate in the treatment of uterine adenomyosis with infertility. They found that, after 2–6 months of leuprorelin acetate therapy, the mean uterine volume was significantly reduced from 180 ± 73 cm3 to 86 ± 67 cm3, leading to an improvement in embryo implantation and clinical pregnancy rates.
Stimulation Protocol – In women without pre-IVF GnRH analog therapy as described above, a long GnRH analog protocol should be considered as it helps to induce decidualization of the adenomyotic deposits rendering the disease inactive. Tao et al. [rx] showed that GnRH antagonist protocol appears to be inferior to GnRH agonist long protocol cycle, and the latter appeared to be associated with increased pregnancy and decreased miscarriage rates.
Two-Staged In Vitro Fertilization – In women with adenomyosis, a two-staged in vitro fertilization could be considered. Patients can undergo ovarian stimulation, oocyte retrieval, and fertilization followed by frozen-thawed embryo transfer (FET) at a later stage. Prior to the FET, GnRH analog suppression therapy for 3 months or so leads to shrinkage of the adenomyosis. FET in the first HRT cycle following GnRH analog suppression therapy, before the adenomyosis lesion regrows to its pretreatment size and exerts its adverse impact on implantation, may improve the result.
Mock Embryo Transfer – Performing a mock embryo transfer is desirable in women with adenomyosis, as it may help to assess the uterine cavity length and position, choose the correct transfer catheter, and alert the clinicians any extra precautions (e.g., use of tenaculum or cervical dilatation). Mock embryo transfer is particularly desirable in those with an enlarged uterus or distorted uterine cavity.
Single Embryo Transfer – Adenomyosis has been reported to be associated with an increased incidence of preterm delivery, preeclampsia, and second-trimester miscarriage when compared with the control group [rx]. Consequently, multiple pregnancies should be avoided and so single embryo transfer should be advised. Women who had adenomyomectomy prior to IVF should also be advised to have SET to avoid multiple pregnancies with a view to minimizing the risk of scar rupture.
HRT Protocol in Frozen-Thawed Embryo Transfer (FET) Cycle – GnRH agonist pretreatment to suppress the pituitary ovarian axis prior to hormone replacement therapy to prepare the endometrium in FET cycles appeared to improve the outcome compared with hormone replacement therapy without downregulation. In a study including 339 patients with adenomyosis, 194 received long-term GnRH agonist plus HRT (downregulation + HRT) and 145 with HRT alone. [rx].
Uterine Contractility and Atosiban Therapy – Several functional studies showed that excessive uterine contractility (>5 contractions per minute) has been demonstrated in approximately 30% of patients undergoing embryo transfer and this may have a significant adverse impact on subsequent embryo implantation and clinical pregnancy rates [rx]. The incidence of abnormal contractility appeared to be higher in women with adenomyosis [rx] which may in part explain the higher incidence of reproductive failure observed in this group of women. Recurrent Implantation Failure – Recurrent implantation failure is diagnosed when there is a failure to achieve a clinical pregnancy after the transfer of at least four good-quality embryos in a minimum of three fresh or frozen cycles in a woman under the age of 40 years [rx]. It is known that adenomyosis is associated with recurrent implantation failure [rx]. Women with recurrent implantation failure should be offered a 3D scan or MRI to establish if there is adenomyosis; if adenomyosis is present, the above management strategies should be adopted to improve the outcome.
Hormones and hormone modulators
Levonorgestrel-releasing intrauterine devices or hormonal IUDs – such as the Mirena, are an effective treatment for adenomyosis.[rx] They reduce symptoms by causing decidualization of the endometrium, reducing or eliminating menstrual flow.[rx] Additionally, by helping downregulate estrogen receptors, hormonal IUDs shrink the clusters of endometrial tissue within the myometrium. This leads to reduced menstrual blood flow, helps the uterus contract more properly, and helps to reduce menstrual pain. The use of hormonal IUDs in patients with adenomyosis has been proven to reduce menstrual bleeding, improve anemia and iron levels, reduce pain, and even result in an improvement of adenomyosis with a smaller uterus on medical imaging.[rx][rx]
Oral contraceptives – reduce the menstrual pain and bleeding associated with adenomyosis. This may require taking continuous hormone therapy to reduce or eliminating menstrual flow. Oral contraceptives may even lead to short-term regression of adenomyosis.
Progesterone or Progestins – Progesterone counteracts estrogen and inhibits the growth of endometrial tissue. Such therapy can reduce or eliminate menstruation in a controlled and reversible fashion. Progestins are chemical variants of natural progesterone.
Gonadotropin-releasing hormone (GnRH) – agonists and danazol have been tried in order to relieve adenomyosis related symptoms and show some effect, but the studies are few, mainly with a retrospective study design and have small sample sizes.[rx] Long-time use of GnRH-analogues is often associated with heavy side effects, loss of bone density and increased risk of cardiovascular events, and therefore not feasible for young women. Furthermore, all present treatment options are irrelevant options for women trying to conceive. Exogenous progestogenic treatments have been found to be ineffective.[rx] In IVF-settings long down-regulation prior to IVF might have a positive effect on pregnancy rates.[rx]
Surgery
Uterine-sparing procedures
Uterine artery embolization (UAE) – In this minimally-invasive procedure, doctors intentionally block two large arteries that supply the uterus, called the uterine arteries. This is performed in order to dramatically reduce the blood supply to the uterus. By doing so, there is insufficient blood and thus oxygen present for the adenomyosis to develop and spread. 57-75% of women who undergo UAE for adenomyosis typically report long-term improvement in their menstrual pain and bleeding. However, there is a recurrence rate of symptoms in 35% of women following a UAE.
Myometrium or adenomyoma resection – In this procedure, surgeons remove a focal consolidation of adenomyosis known as an adenomyoma. To be successful this procedure requires that the adenomyosis is relatively focally isolated and with a minimal diffuse spread. Unfortunately, adenomyosis is commonly diffuse and the operation is successful only 50% of the time. The procedure is performed with either a laparoscope or hysteroscope.[rx]
Endometrial ablation techniques – are only for women who have completed their childbearing. The techniques either include physical resection and removal of the endometrium through a hysteroscope or focus on ablating or killing the endometrial layer of the uterus without its immediate removal. Endometrial ablation and resection techniques are most appropriate for shallow adenomyosis. The efficacy of the procedures is reduced if the adenomyosis is too widespread or deep. Furthermore, deep adenomyosis may become trapped behind a scarred region that was ablated, leading to further bleeding and pain. Endometrial resection is also limited to relatively shallow adenomyosis as significant bleeding may result from damage to large arteries that are present 5 mm deep within the myometrium.[rx]
Non-hysteroscopic procedures – These techniques do not require a hysteroscope are relatively fast, and many can be performed as an outpatient procedure.
Thermal balloon – Using a thin expanding balloon placed within the uterus, providers can introduce heated fluid and ablate the endometrium. This procedure has been shown to result in amenorrhea or complete cessation of menstrual bleeding for 12 months in 23% of patients. 16% of patients eventually experience treatment failure with pain or bleeding requiring additional treatments or a hysterectomy. [9]
Cryo-endometrial ablation (CEA) – A form of cryotherapy whereby using a small probe, providers can directly apply sub-zero temperatures within the uterus to freeze and ablate the endometrium.
Circulating Hot Water – Heated water directly introduced into the uterus is used to thermally ablate the endometrium.
Microwave ablation – Using a small probe introduced into the uterus, a provider uses microwave energy to ablate the endometrium.
High-energy radiofrequency ablation – Using a small expandable mesh placed within the uterus, providers use high-energy radio waves to ablate the endometrium.
Hysteroscopic procedures: These techniques all require the use of a hysteroscope to perform.
Wire-loop resection – Under direct visualization through a hysteroscope, a wire loop instrument charged with an electric current permits a provider to carefully remove the endometrium in strips.
Laser ablation – Under direct visualization through a hysteroscope, lasers are used to vaporize and ablate the endometrium.
Rollerball ablation – Under direct visualization through a hysteroscope, a metallic ball on the end of a probe is charged with electricity and rolled across the surface of the endometrium. This has been shown to have a coagulative effect to the depth of 2–3 mm into the myometrium. This destroys the endometrium and the nearby growth of dysfunctional smooth muscle. Deeper adenomyosis escapes this coagulative effect.[9]