Childhood Soft Tissue Sarcoma Treatment (PDQ®)–Health Professional Version
General Information About Childhood Soft Tissue Sarcoma
Dramatic improvements in survival have been achieved for children and adolescents with cancer. Between 1975 and 2020, childhood cancer mortality decreased by more than 50%.[1–3] Childhood and adolescent cancer survivors require close monitoring because cancer therapy side effects may persist or develop months or years after treatment. For information about the incidence, type, and monitoring of late effects in childhood and adolescent cancer survivors, see Late Effects of Treatment for Childhood Cancer.
Pediatric soft tissue sarcomas are a heterogenous group of malignant tumors that originate from primitive mesenchymal tissue and account for 6% of all childhood tumors (rhabdomyosarcomas, 3%; other soft tissue sarcomas, 3%).[2] For more information, see the Histopathological Classification of Childhood Soft Tissue Sarcoma section.
Rhabdomyosarcoma, a tumor of striated muscle, is the most common soft tissue sarcoma in children. It accounts for 50% of the soft tissue sarcomas in children aged 0 to 14 years.[2] For more information, see Childhood Rhabdomyosarcoma Treatment.
In pediatrics, the remaining soft tissue sarcomas are commonly referred to as nonrhabdomyosarcomatous soft tissue sarcomas (NRSTS) and account for approximately 3.5% of all childhood tumors.[2,4] This summary discusses the treatment of NRSTS.
NRSTS are often classified according to the normal tissue types from which they are derived. These types include various connective tissues, peripheral nervous system tissue, smooth muscle tissue, and vascular tissue. The classification also includes undifferentiated tumors that are not clearly related to specific tissue types. For more information about vascular tumors in children, see Childhood Vascular Tumors Treatment.
Incidence of Soft Tissue Sarcoma by Age and Histology
The distribution of soft tissue sarcomas by histology and age, based on the Surveillance, Epidemiology, and End Results (SEER) Program information from 2000 to 2015, is depicted in Table 1. The distribution of histological subtypes by age is also shown in Figure 2.
Table 1. Age Distribution of Soft Tissue Sarcomas in Children Aged 0 to 19 Years (SEER 2000–2015)a
Age <5 y
Age 5–9 y
Age 10–14 y
Age 15–19 y
Age <20 y
All Ages (Including Adults)
pPNET = peripheral primitive neuroectodermal tumors; SEER = Surveillance, Epidemiology, and End Results.
Fibrosarcomas, peripheral nerve, and other fibrous neoplasms
137
64
112
181
494
6,645
Fibroblastic and myofibroblastic tumors
114
33
41
77
265
4,228
Nerve sheath tumors
23
31
70
102
226
2,303
Other fibromatous neoplasms
0
0
1
2
3
114
Kaposi sarcoma
2
1
2
10
15
7,722
Other specified soft tissue sarcomas
237
238
559
865
1,899
49,004
Ewing tumor and Askin tumor of soft tissue
37
36
72
113
258
596
pPNET of soft tissue
24
23
42
56
145
402
Extrarenal rhabdoid tumor
75
8
9
4
96
205
Liposarcomas
4
6
37
79
126
10,749
Fibrohistiocytic tumors
43
73
142
223
481
13,531
Leiomyosarcomas
11
14
19
41
85
14,107
Synovial sarcomas
12
39
141
210
402
2,608
Blood vessel tumors
12
9
11
32
64
4,238
Osseous and chondromatous neoplasms of soft tissue
1
6
16
14
37
1,018
Alveolar soft parts sarcoma
4
5
22
33
64
211
Miscellaneous soft tissue sarcomas
14
19
48
60
141
1,339
Unspecified soft tissue sarcomas
80
53
146
175
454
13,614
Soft tissue sarcomas include both rhabdomyosarcomas and NRSTS. NRSTS are more common in adolescents and adults.[6] Most of the information regarding treatment and natural history of the disease in younger patients has been based on studies in adult patients. The distributions of soft tissue sarcomas by age according to stage (Figure 1), histological subtype (Figure 2), and tumor site (Figure 3) are shown below.[7]
EnlargeFigure 1. The distribution of soft tissue sarcomas by age according to stage.EnlargeFigure 2. The distribution of soft tissue sarcomas by age according to histological subtype.EnlargeFigure 3. The distribution of soft tissue sarcomas by age according to tumor site.
Risk Factors
Some genetic factors and external exposures have been associated with the development of NRSTS, including the following:
Genetic factors:
Li-Fraumeni syndrome: Patients with Li-Fraumeni syndrome (usually resulting from heritable cancer-associated changes of the TP53 tumor suppressor gene) have an increased risk of developing soft tissue tumors (mostly NRSTS), bone sarcomas, breast cancer, brain tumors, and acute leukemia.[8,9]
Familial adenomatous polyposis: Patients with familial adenomatous polyposis are at increased risk of developing desmoid-type fibromatosis.[10]
RB1 gene: Germline pathogenic variants of the RB1 gene have been associated with an increased risk of developing soft tissue sarcoma, particularly leiomyosarcoma, and the risk appears higher among those younger than 1 year who were treated with alkylating agents.[11,12]
SMARCB1 gene: Germline pathogenic variants or deletions of the SMARCB1 gene are associated with an increased risk of developing extrarenal rhabdoid tumors.[13] For more information about SMARCB1 and rhabdoid tumor predisposition syndrome type 1, see Rhabdoid Tumor Predisposition Syndrome Type 1.
Neurofibromatosis type 1: Approximately 4% of patients with neurofibromatosis type 1 develop malignant peripheral nerve sheath tumors, which usually develop after a long latency. Some patients develop multiple lesions.[14–16]
Werner syndrome: Werner syndrome is characterized by spontaneous chromosomal instability, resulting in increased susceptibility to cancer and premature aging. An excess of soft tissue sarcomas has been reported in patients with Werner syndrome.[17]
Tuberous sclerosis complex: Tuberous sclerosis complex is associated with the development of various tumors showing perivascular epithelioid cell differentiation (PEComas), including lymphangioleiomyomatosis and hepatic and renal angiomyolipomas.[18–20]
Adenosine deaminase–deficient severe combined immunodeficiency: Patients with adenosine deaminase–deficient severe combined immunodeficiency are at increased risk of developing multicentric dermatofibrosarcoma protuberans, which usually presents at an average age of 8.9 years.[21]
External exposures:
Radiation: Some NRSTS (particularly malignant fibrous histiocytoma) can develop within a previously irradiated site.[22–26]
Epstein-Barr virus (EBV) infection in patients with AIDS: Some NRSTS (e.g., leiomyosarcoma) have been linked to EBV infection in patients with AIDS.[22,27]
Clinical Presentation
NRSTS can develop in any part of the body, but they arise most commonly in the trunk and extremities.[28–30] Although rare, these tumors can arise in brain tissue and are treated according to the histological type.[31]
NRSTS can present initially as an asymptomatic solid mass, or they may be symptomatic because of local invasion or impact on adjacent anatomical structures. Systemic symptoms (e.g., fever, weight loss, and night sweats) are rare. Hypoglycemia and hypophosphatemic rickets have been reported in cases of hemangiopericytoma, which was identified as a solitary fibrous tumor and is now included within myofibroma in the revised World Health Organization (WHO) classification. Hyperglycemia has been noted in patients with fibrosarcoma of the lung.[32]
Diagnostic and Staging Evaluation
When a suspicious lesion is identified, it is crucial to perform a complete workup, followed by adequate biopsy. The lesion is imaged before initiating any intervention using the following procedures:
Plain films. Plain films can be used to rule out bone involvement and detect calcifications that may be seen in soft tissue tumors such as extraskeletal osteosarcoma or synovial sarcoma.
Computed tomography (CT). Chest CT is essential to assess the presence of metastases. An abdominal CT can be used to image intra-abdominal tumors, such as liposarcoma. Patients with NRSTS who were treated in 11 centers as part of the European paediatric Soft Tissue Sarcoma Study Group (EpSSG) were retrospectively assessed to evaluate the impact of indeterminate pulmonary nodules identified on chest CT.[33] Of the 206 patients examined, 109 (52.9%) did not have any nodules, 78 (38%) had at least one indeterminate nodule, and 19 (9.2%) had nodules meeting the definition of metastases. The 5-year event-free survival (EFS) rate was 78.5% (95% confidence interval [CI], 69.4%–85.1%) for patients without nodules and 69.6% (95% CI, 57.9%–78.7%) for patients with indeterminate nodules (P = .135). The 5-year overall survival (OS) rate was 87.4% (95% CI, 79.3%–92.5%) for patients without nodules and 79.0% (95% CI, 67.5%–86.8%) for patients with indeterminate nodules (P = .086).
Magnetic resonance imaging (MRI). MRI may be essential for a surgeon to achieve adequate surgical margins. MRI can be used to image intra-abdominal tumors, such as liposarcoma, and is essential for extremity lesions.
Positron emission tomography (PET) scan and bone scan. In a retrospective study, 46 PET scans were completed in 25 pediatric patients with soft tissue sarcoma.[34] The positive predictive value of finding metastatic disease was 89%, and the negative predictive value was 67%. A small retrospective study of nine patients with NRSTS suggested that PET-CT was more accurate and cost-effective than either modality alone in identifying distant metastatic disease.[35] The use of this modality in pediatric NRSTS has not been studied prospectively.
The imaging characteristics of some tumors can be highly suggestive of that particular diagnosis. For example, the imaging characteristics of pediatric low-grade fibromyxoid sarcoma and alveolar soft part sarcoma have been described and can aid in the diagnosis of these rare neoplasms.[36]
Biopsy strategies
Although NRSTS are pathologically distinct from rhabdomyosarcoma and Ewing sarcoma, the classification of childhood NRSTS type is often difficult. Core-needle biopsy, incisional biopsy, or excisional biopsy can be used to diagnose NRSTS. If possible, the surgeon who will perform the definitive resection needs to be involved in the biopsy decision. Poorly placed incisional or needle biopsies may adversely affect the ability to achieve negative margins.
Needle biopsy techniques must ensure adequate tissue sampling. Given the diagnostic importance of translocations and other molecular changes, a core-needle biopsy or small incisional biopsy that obtains adequate tumor tissue is crucial to allow for conventional histological and immunocytochemical analysis and other studies such as light and electron microscopy, cytogenetics, fluorescence in situ hybridization, and molecular pathology.[37,38]
The acquisition of multiple cores of tissue may be required. Of 530 suspected soft tissue masses in (largely adult) patients who underwent core-needle biopsies, 426 (80%) were proven to be soft tissue tumors, 225 (52.8%) of which were malignant. Core-needle biopsy was able to differentiate soft tissue sarcomas from benign lesions with a sensitivity of 96.3% and a specificity of 99.4%. Tumor subtype was accurately assigned in 89.5% of benign lesions and in 88% of soft tissue sarcomas. The biopsy complication rate was 0.4%.[39]
Considerations related to a biopsy procedure are as follows:
Core-needle biopsy for a deep-seated tumor can lead to formation of a hematoma, which affects subsequent resection and/or radiation (because the hematoma should be covered in the irradiated volume).
Fine-needle biopsy is usually not recommended because it is difficult to determine the accurate histological diagnosis and grade of the tumor in this heterogeneous group of tumors.
Image guidance using ultrasonography, CT scan, or MRI may be necessary to ensure a representative biopsy.[40] Image guidance is particularly helpful in deep lesions and to avoid cystic changes or necrotic tumors.[41]
Incisional biopsies must not compromise subsequent wide local excision.
Excisional biopsy of the lesion is only appropriate for small superficial lesions (<3 cm in size) and are discouraged.[42,43] If an excisional biopsy is contemplated, then MRI of the area is recommended to define the area of involvement as subsequent surgery or radiation therapy may be needed.
Various institutional series have demonstrated the feasibility and effectiveness of sentinel lymph node biopsy as a staging procedure in pediatric patients with soft tissue sarcomas.[44–49] The utility of sentinel node biopsy is currently limited to epithelioid sarcoma, clear cell sarcoma, and rhabdomyosarcoma of the trunk and extremities.[50]
In a prospective study of pediatric patients with sarcoma who underwent sentinel lymph node biopsy, 28 patients were examined. Sentinel lymph node biopsy was positive in 7 of the 28 patients, including 3 patients (43%) who had negative PET-CT scans. PET-CT overestimated and suggested nodal involvement in 14 patients, more than what was confirmed by sentinel lymph node biopsy. The findings from the sentinel lymph node biopsies resulted in altering therapy for all seven patients who were determined to have metastatic disease. As indicated by previous reports, epithelioid sarcoma and clear cell sarcoma were the two NRSTS included in this study.[50]
In the ARST0332 (NCT00346164) study, patients with epithelioid sarcoma, clear cell sarcoma, or radiographically enlarged nodes underwent regional node sampling. Nodal metastases were identified in 20 patients (3.8%), and all but one of these patients had radiographic evidence of nodal involvement. The most common histologies included epithelioid sarcoma (18%), angiosarcoma (17%), and clear cell sarcoma (14%). Patients with isolated nodal metastases had a similar outcome to those who did not have distant metastases (5-year OS rates, 85% vs. 87%). Sentinel lymph node biopsies were encouraged but not required for this study. A sentinel lymph node biopsy was not done in most patients because they had clinically enlarged nodes. Of note, three patients without clinical evidence of lymph node metastasis at study entry experienced lymph node basin failure. One of these patients had three lymph nodes in two different lymph node basins sampled by sentinel lymph node biopsy that were pathologically negative.[51]
Transverse extremity incisions are avoided to reduce skin loss at re-excision and because they require a greater cross-sectional volume of tissue to be covered in the radiation field. Other extensive surgical procedures are also avoided before definitive diagnosis.
For these reasons, open biopsy or multiple core-needle biopsies are strongly encouraged so that adequate tumor tissue can be obtained to allow crucial studies to be performed and to avoid limiting future treatment options.
Unplanned resection
In children with unplanned resection of NRSTS, primary re-excision is frequently recommended because many patients will have tumor present in the re-excision specimen.[52,53] A single-institution analysis of adolescents and adults compared patients who had unplanned excisions of soft tissue sarcoma to stage-matched controls. In this retrospective analysis, unplanned initial excision of soft tissue sarcoma resulted in increased risk of local recurrence, metastasis, and death. This increased risk was greatest for high-grade tumors.[54][Level of evidence C1] In this case, a second resection is expected.
Chromosomal abnormalities
Many NRSTS are characterized by chromosomal abnormalities. Some of these chromosomal translocations lead to a fusion of two disparate genes. The resulting fusion transcript can be readily detected by using polymerase chain reaction–based techniques, thus facilitating the diagnosis of those neoplasms that have translocations.
Some of the most frequent aberrations seen in NRSTS are listed in Table 2.
Table 2. Frequent Chromosomal Aberrations Seen in Nonrhabdomyosarcomatous Soft Tissue Sarcomaa
Histology
Chromosomal Aberrations
Genes Involved
aAdapted from Sandberg,[55] Slater et al.,[56] Mertens et al.,[57] Romeo,[58] and Schaefer et al.[59]
Presence of metastases and site of the metastatic tumor.
Resectability of the tumor.
Use of radiation therapy.
In a review of a large adult series of NRSTS, patients with superficial extremity sarcomas had a better prognosis than did patients with deep tumors. This may be a reflection of differences in resectability. Thus, in addition to grade and size, the depth of invasion of the tumor should be considered.[76]
Data specific to NRSTS in children and adolescents are difficult to separate. Several adult and pediatric series have shown that patients with large or invasive tumors have a significantly worse prognosis than do those with small, noninvasive tumors. A retrospective review of soft tissue sarcomas (rhabdomyosarcoma and NRSTS) in children and adolescents suggests that the 5-cm cutoff used for adults with soft tissue sarcoma may not be ideal for smaller children, especially infants. The review identified an interaction between tumor diameter and body surface area.[77] This relationship has been questioned in a rhabdomyosarcoma study and requires further study to determine the therapeutic implications of the observation.[78]
Some pediatric NRSTS are associated with a better outcome. For instance, patients with infantile fibrosarcoma who present at age 4 years or younger have an excellent prognosis. This excellent outcome occurs because surgery alone can cure a significant number of these patients and infantile fibrosarcoma is highly chemosensitive. This tumor also responds well to larotrectinib, a specific tropomyosin receptor kinase inhibitor.[22,79]
Prognosis based on the Children’s Oncology Group (COG) ARST0332 trial
Soft tissue sarcomas in older children and adolescents often behave similarly to those in adult patients.[22,80] A large, prospective, multinational COG study (ARST0332 [NCT00346164]) enrolled newly diagnosed patients younger than 30 years. Patients were assigned to treatment based on their risk group. Risk groups were defined by the presence of metastasis, tumor resectability and margins, and tumor size and grade. For more information, see Figure 4.[81][Level of evidence B4]
Each patient was assigned to one of three risk groups and one of four treatment groups. The risk groups were as follows:[81]
Low risk: Nonmetastatic R0 (resection was complete with negative microscopic margins) or R1 (microscopically positive margins) low-grade tumor, or ≤5 cm R1 high-grade tumor.
Intermediate risk: Nonmetastatic R0 or R1 >5 cm high-grade tumor, or unresected tumor of any size or grade.
Neoadjuvant chemoradiation therapy (chemotherapy and 45 Gy radiation therapy, then surgery and radiation therapy boost based on margins with continued chemotherapy) (n = 196).
Chemotherapy included six cycles of ifosfamide (3 g/m2 per dose) given intravenously on days 1 through 3 and five cycles of doxorubicin (37.5 mg/m2 per dose) given intravenously on days 1 to 2 every 3 weeks, with the sequence adjusted based on the timing of surgery or radiation therapy.
For the 550 patients enrolled, 529 evaluable patients were included in the analysis. At a median follow-up of 6.5 years (interquartile range [IQR], 4.9–7.9), the survival results are shown in Table 3.
Table 3. Survival Results for the Children’s Oncology Group ARST0332 Trial
5-Year Event-Free Survival
5-Year Overall Survival
Risk Group
Events/Patients
Estimate, % (95% CI)
Events/Patients
Estimate, % (95% CI)
CI = confidence interval; R0 = completely excised with negative microscopic margins; R1 = grossly excised but with positive microscopic margins; R2 = less than complete gross excision.
Low
26/222
88.9 (84.0–93.8)
10/222
96.2 (93.2–99.2)
Intermediate
84/227
65.0 (58.2–71.8)
55/227
79.2 (73.4–85.0)
High
63/80
21.2 (11.4–31.1)
52/80
35.5 (23.6–47.4)
Surgical Margin
R0
44/252
83.6 (78.3–89.0)
22/252
92.8 (89.1–96.5)
R1
29/81
66.2 (54.8–77.5)
17/81
79.7 (70.0–89.5)
R2
100/196
49.2 (41.4–57.0)
78/196
62.7 (55.2–70.3)
The COG ARST0332 trial was a risk-based stratification study. Overall, local control after radiation therapy was as follows: R0, 106 of 109 patients (97%); R1, 51 of 60 patients (85%); and R2/unresectable, 2 of 6 patients (33%). Local recurrence predictors included extent of delayed resection (P < .001), imaging response before delayed surgery (P < .001), histological subtype (P < .001), and no radiation therapy (P = .046). The 5-year EFS was significantly lower for patients unable to undergo R0 or R1 resection (P = .0003).[82]
Pediatric patients with unresected localized NRSTS have a poor outcome. Only about one-third of patients treated with multimodality therapy remain disease free.[73,83]; [84,85][Level of evidence C1] In an Italian review of 30 patients with NRSTS at visceral sites, only ten patients survived at 5 years. Unfavorable prognostic factors included inability to achieve complete resection, large tumor size, tumor invasion, histological subtype, and lung-pleura sites.[86][Level of evidence C1]
Prognosis based on the European paediatric Soft Tissue Sarcoma Study Group (EpSSG) NRSTS 2005 study
The EpSSG conducted a prospective trial for patients younger than 21 years with NRSTS. They reported an analysis of 206 patients with synovial sarcoma and 363 with adult-type NRSTS. Patients were treated according to assigned risk groups. For more information, see Figure 5.[87] With a median follow-up of 80 months (interquartile range, 54.3–111.3) for the 467 surviving patients, the 5-year EFS rate was 73.7% (95% CI, 69.7%–77.2%), and the OS rate was 83.8% (95% CI, 80.3%–86.7%). The survival by treatment groups are shown in Table 4.[87]
EnlargeFigure 5. Treatment plan for patients with synovial sarcoma or adult-type non-rhabdomyosarcoma soft tissue sarcomas. Patients were divided into four treatment groups based on surgical stage, tumour size, nodal involvement, tumour grade (according to the Fédération Nationale des Centres de Lutte Contre le Cancer grading system for adult-type non-rhabdomyosarcoma soft tissue sarcomas), and tumour site (for synovial sarcoma). I+D = ifosfamide (3.0 g/m2 per day intravenously for 3 days) plus doxorubicin (37.5 mg/m2 per day intravenously for 2 days). I = ifosfamide (3.0 g/m2 per day intravenously for 2 days). IRS = Intergroup Rhabdomyosarcoma Study. N1 = nodal involvement. S = delayed surgery. Reprinted from The Lancet Child & Adolescent Health, Volume 5, Issue 8, Ferrari A, van Noesel MM, Brennan B, et al., Paediatric non-rhabdomyosarcoma soft tissue sarcomas: the prospective NRSTS 2005 study by the European paediatric Soft Tissue Sarcoma Study Group (EpSSG), Pages 546-558, Copyright 2021, with permission from Elsevier.
Table 4. Survival Outcomes by Treatment Groups in the EpSSG NRSTS 2005 Study
Treatment Group
5-Year Event-Free Survival Rate (95% CI)
5-Year Overall Survival Rate (95% CI)
Local Recurrence Rate
CI = confidence interval; EpSSG = European paediatric Soft Tissue Sarcoma Study Group; NRSTS = nonrhabdomyosarcomatous soft tissue sarcomas.
Delayed surgery followed by radiation therapy (n = 104)
16
6
8
Delayed surgery alone (n = 48)
8
3
8
No local treatment (n = 16)
12
4
0
Preoperative radiation therapy followed by delayed surgery (n = 20)
4
0
6
The authors concluded that adjuvant therapy (radiation therapy and chemotherapy) could safely be omitted in the group of patients assigned to surgery alone. Their criteria included the following:[87]
Synovial cell: Intergroup Rhabdomyosarcoma Study (IRS) group I tumor size <5 cm.
Adult-type NRSTS: IRS group I tumor size <5 cm, any grade.
Adult-type NRSTS: IRS group I tumor size >5 cm, tumor grade I.
Adult-type NRSTS: IRS group II any tumor size, tumor grade I.
They also concluded that improving the outcome for patients with high-risk, initially resected, adult-type NRSTS and those with initially unresected disease remains a major clinical challenge.[87]
In a pooled analysis from U.S. and European pediatric centers, outcomes were better for patients whose tumor removal procedure was deemed complete than for patients whose tumor removal was incomplete. Outcomes were better for patients who received radiation therapy than for patients who did not.[84][Level of evidence C1]
Because long-term morbidity must be minimized while disease-free survival is maximized, the ideal therapy for each patient must be carefully and individually determined using these prognostic factors before initiating therapy.[29,88–92]
References
Smith MA, Altekruse SF, Adamson PC, et al.: Declining childhood and adolescent cancer mortality. Cancer 120 (16): 2497-506, 2014. [PUBMED Abstract]
National Cancer Institute: NCCR*Explorer: An interactive website for NCCR cancer statistics. Bethesda, MD: National Cancer Institute. Available online. Last accessed February 25, 2025.
Surveillance Research Program, National Cancer Institute: SEER*Explorer: An interactive website for SEER cancer statistics. Bethesda, MD: National Cancer Institute. Available online. Last accessed December 30, 2024.
Hawkins DS, Black JO, Orbach D, et al.: Nonrhabdomyosarcoma soft-tissue sarcomas. In: Blaney SM, Helman LJ, Adamson PC, eds.: Pizzo and Poplack’s Pediatric Oncology. 8th ed. Wolters Kluwer, 2020, pp 721-46.
Surveillance, Epidemiology, and End Results (SEER) Program: SEER*Stat Database: Incidence – SEER 18 Regs Research Data + Hurricane Katrina Impacted Louisiana Cases, Nov 2017 Sub (1973-2015 varying) – Linked To County Attributes – Total U.S., 1969-2016 Counties [Database]. National Cancer Institute, DCCPS, Surveillance Research Program, released April 2018, based on the November 2017 submission. Available online. Last accessed October 12, 2022.
Ferrari A, Sultan I, Huang TT, et al.: Soft tissue sarcoma across the age spectrum: a population-based study from the Surveillance Epidemiology and End Results database. Pediatr Blood Cancer 57 (6): 943-9, 2011. [PUBMED Abstract]
Chang F, Syrjänen S, Syrjänen K: Implications of the p53 tumor-suppressor gene in clinical oncology. J Clin Oncol 13 (4): 1009-22, 1995. [PUBMED Abstract]
Plon SE, Malkin D: Childhood cancer and hereditary. In: Pizzo PA, Poplack DG, eds.: Principles and Practice of Pediatric Oncology. 7th ed. Lippincott Williams and Wilkins, 2015, pp 13-31.
Groen EJ, Roos A, Muntinghe FL, et al.: Extra-intestinal manifestations of familial adenomatous polyposis. Ann Surg Oncol 15 (9): 2439-50, 2008. [PUBMED Abstract]
Kleinerman RA, Tucker MA, Abramson DH, et al.: Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst 99 (1): 24-31, 2007. [PUBMED Abstract]
Wong JR, Morton LM, Tucker MA, et al.: Risk of subsequent malignant neoplasms in long-term hereditary retinoblastoma survivors after chemotherapy and radiotherapy. J Clin Oncol 32 (29): 3284-90, 2014. [PUBMED Abstract]
Eaton KW, Tooke LS, Wainwright LM, et al.: Spectrum of SMARCB1/INI1 mutations in familial and sporadic rhabdoid tumors. Pediatr Blood Cancer 56 (1): 7-15, 2011. [PUBMED Abstract]
deCou JM, Rao BN, Parham DM, et al.: Malignant peripheral nerve sheath tumors: the St. Jude Children’s Research Hospital experience. Ann Surg Oncol 2 (6): 524-9, 1995. [PUBMED Abstract]
Stark AM, Buhl R, Hugo HH, et al.: Malignant peripheral nerve sheath tumours–report of 8 cases and review of the literature. Acta Neurochir (Wien) 143 (4): 357-63; discussion 363-4, 2001. [PUBMED Abstract]
Goto M, Miller RW, Ishikawa Y, et al.: Excess of rare cancers in Werner syndrome (adult progeria). Cancer Epidemiol Biomarkers Prev 5 (4): 239-46, 1996. [PUBMED Abstract]
Fricke BL, Donnelly LF, Casper KA, et al.: Frequency and imaging appearance of hepatic angiomyolipomas in pediatric and adult patients with tuberous sclerosis. AJR Am J Roentgenol 182 (4): 1027-30, 2004. [PUBMED Abstract]
Adriaensen ME, Schaefer-Prokop CM, Duyndam DA, et al.: Radiological evidence of lymphangioleiomyomatosis in female and male patients with tuberous sclerosis complex. Clin Radiol 66 (7): 625-8, 2011. [PUBMED Abstract]
Hornick JL, Fletcher CD: PEComa: what do we know so far? Histopathology 48 (1): 75-82, 2006. [PUBMED Abstract]
Kesserwan C, Sokolic R, Cowen EW, et al.: Multicentric dermatofibrosarcoma protuberans in patients with adenosine deaminase-deficient severe combined immune deficiency. J Allergy Clin Immunol 129 (3): 762-769.e1, 2012. [PUBMED Abstract]
Spunt SL, Million L, Coffin C: The nonrhabdomyosarcoma soft tissue sarcoma. In: Pizzo PA, Poplack DG, eds.: Principles and Practice of Pediatric Oncology. 7th ed. Lippincott Williams and Wilkins, 2015, pp 827-54.
Tukenova M, Guibout C, Hawkins M, et al.: Radiation therapy and late mortality from second sarcoma, carcinoma, and hematological malignancies after a solid cancer in childhood. Int J Radiat Oncol Biol Phys 80 (2): 339-46, 2011. [PUBMED Abstract]
Bartkowiak D, Humble N, Suhr P, et al.: Second cancer after radiotherapy, 1981-2007. Radiother Oncol 105 (1): 122-6, 2012. [PUBMED Abstract]
Casey DL, Friedman DN, Moskowitz CS, et al.: Second cancer risk in childhood cancer survivors treated with intensity-modulated radiation therapy (IMRT). Pediatr Blood Cancer 62 (2): 311-316, 2015. [PUBMED Abstract]
McClain KL, Leach CT, Jenson HB, et al.: Association of Epstein-Barr virus with leiomyosarcomas in children with AIDS. N Engl J Med 332 (1): 12-8, 1995. [PUBMED Abstract]
Dillon P, Maurer H, Jenkins J, et al.: A prospective study of nonrhabdomyosarcoma soft tissue sarcomas in the pediatric age group. J Pediatr Surg 27 (2): 241-4; discussion 244-5, 1992. [PUBMED Abstract]
Zeytoonjian T, Mankin HJ, Gebhardt MC, et al.: Distal lower extremity sarcomas: frequency of occurrence and patient survival rate. Foot Ankle Int 25 (5): 325-30, 2004. [PUBMED Abstract]
Benesch M, von Bueren AO, Dantonello T, et al.: Primary intracranial soft tissue sarcoma in children and adolescents: a cooperative analysis of the European CWS and HIT study groups. J Neurooncol 111 (3): 337-45, 2013. [PUBMED Abstract]
Giraudo C, Schoot R, Cardoen L, et al.: Indeterminate pulmonary nodules in non-rhabdomyosarcoma soft tissue sarcoma: A study of the European paediatric Soft Tissue Sarcoma Study Group. Cancer 130 (4): 597-608, 2024. [PUBMED Abstract]
Mody RJ, Bui C, Hutchinson RJ, et al.: FDG PET imaging of childhood sarcomas. Pediatr Blood Cancer 54 (2): 222-7, 2010. [PUBMED Abstract]
Tateishi U, Hosono A, Makimoto A, et al.: Accuracy of 18F fluorodeoxyglucose positron emission tomography/computed tomography in staging of pediatric sarcomas. J Pediatr Hematol Oncol 29 (9): 608-12, 2007. [PUBMED Abstract]
Sargar K, Kao SC, Spunt SL, et al.: MRI and CT of Low-Grade Fibromyxoid Sarcoma in Children: A Report From Children’s Oncology Group Study ARST0332. AJR Am J Roentgenol 205 (2): 414-20, 2015. [PUBMED Abstract]
Recommendations for the reporting of soft tissue sarcomas. Association of Directors of Anatomic and Surgical Pathology. Mod Pathol 11 (12): 1257-61, 1998. [PUBMED Abstract]
Strauss DC, Qureshi YA, Hayes AJ, et al.: The role of core needle biopsy in the diagnosis of suspected soft tissue tumours. J Surg Oncol 102 (5): 523-9, 2010. [PUBMED Abstract]
Chowdhury T, Barnacle A, Haque S, et al.: Ultrasound-guided core needle biopsy for the diagnosis of rhabdomyosarcoma in childhood. Pediatr Blood Cancer 53 (3): 356-60, 2009. [PUBMED Abstract]
Tuttle R, Kane JM: Biopsy techniques for soft tissue and bowel sarcomas. J Surg Oncol 111 (5): 504-12, 2015. [PUBMED Abstract]
Coffin CM, Dehner LP, O’Shea PA: Pediatric Soft Tissue Tumors: A Clinical, Pathological, and Therapeutic Approach. Williams and Wilkins, 1997.
Smith LM, Watterson J, Scott SM: Medical and surgical management of pediatric soft tissue tumors. In: Coffin CM, Dehner LP, O’Shea PA: Pediatric Soft Tissue Tumors: A Clinical, Pathological, and Therapeutic Approach. Williams and Wilkins, 1997, pp 360-71.
Neville HL, Andrassy RJ, Lally KP, et al.: Lymphatic mapping with sentinel node biopsy in pediatric patients. J Pediatr Surg 35 (6): 961-4, 2000. [PUBMED Abstract]
Kayton ML, Delgado R, Busam K, et al.: Experience with 31 sentinel lymph node biopsies for sarcomas and carcinomas in pediatric patients. Cancer 112 (9): 2052-9, 2008. [PUBMED Abstract]
Dall’Igna P, De Corti F, Alaggio R, et al.: Sentinel node biopsy in pediatric patients: the experience in a single institution. Eur J Pediatr Surg 24 (6): 482-7, 2014. [PUBMED Abstract]
Parida L, Morrisson GT, Shammas A, et al.: Role of lymphoscintigraphy and sentinel lymph node biopsy in the management of pediatric melanoma and sarcoma. Pediatr Surg Int 28 (6): 571-8, 2012. [PUBMED Abstract]
Alcorn KM, Deans KJ, Congeni A, et al.: Sentinel lymph node biopsy in pediatric soft tissue sarcoma patients: utility and concordance with imaging. J Pediatr Surg 48 (9): 1903-6, 2013. [PUBMED Abstract]
Wagner LM, Kremer N, Gelfand MJ, et al.: Detection of lymph node metastases in pediatric and adolescent/young adult sarcoma: Sentinel lymph node biopsy versus fludeoxyglucose positron emission tomography imaging-A prospective trial. Cancer 123 (1): 155-160, 2017. [PUBMED Abstract]
Alvarez E, He J, Spunt SL, et al.: Lymph node metastases in paediatric and young adult patients with non-rhabdomyosarcoma soft tissue sarcoma (NRSTS): Findings from Children’s Oncology Group (COG) study ARST0332. Eur J Cancer 180: 89-98, 2023. [PUBMED Abstract]
Chui CH, Spunt SL, Liu T, et al.: Is reexcision in pediatric nonrhabdomyosarcoma soft tissue sarcoma necessary after an initial unplanned resection? J Pediatr Surg 37 (10): 1424-9, 2002. [PUBMED Abstract]
Cecchetto G, Guglielmi M, Inserra A, et al.: Primary re-excision: the Italian experience in patients with localized soft-tissue sarcomas. Pediatr Surg Int 17 (7): 532-4, 2001. [PUBMED Abstract]
Qureshi YA, Huddy JR, Miller JD, et al.: Unplanned excision of soft tissue sarcoma results in increased rates of local recurrence despite full further oncological treatment. Ann Surg Oncol 19 (3): 871-7, 2012. [PUBMED Abstract]
Sandberg AA: Translocations in malignant tumors. Am J Pathol 159 (6): 1979-80, 2001. [PUBMED Abstract]
Slater O, Shipley J: Clinical relevance of molecular genetics to paediatric sarcomas. J Clin Pathol 60 (11): 1187-94, 2007. [PUBMED Abstract]
Ladanyi M, Lui MY, Antonescu CR, et al.: The der(17)t(X;17)(p11;q25) of human alveolar soft part sarcoma fuses the TFE3 transcription factor gene to ASPL, a novel gene at 17q25. Oncogene 20 (1): 48-57, 2001. [PUBMED Abstract]
Ladanyi M: The emerging molecular genetics of sarcoma translocations. Diagn Mol Pathol 4 (3): 162-73, 1995. [PUBMED Abstract]
Williams A, Bartle G, Sumathi VP, et al.: Detection of ASPL/TFE3 fusion transcripts and the TFE3 antigen in formalin-fixed, paraffin-embedded tissue in a series of 18 cases of alveolar soft part sarcoma: useful diagnostic tools in cases with unusual histological features. Virchows Arch 458 (3): 291-300, 2011. [PUBMED Abstract]
Antonescu CR, Dal Cin P, Nafa K, et al.: EWSR1-CREB1 is the predominant gene fusion in angiomatoid fibrous histiocytoma. Genes Chromosomes Cancer 46 (12): 1051-60, 2007. [PUBMED Abstract]
Hisaoka M, Ishida T, Kuo TT, et al.: Clear cell sarcoma of soft tissue: a clinicopathologic, immunohistochemical, and molecular analysis of 33 cases. Am J Surg Pathol 32 (3): 452-60, 2008. [PUBMED Abstract]
Barnoud R, Sabourin JC, Pasquier D, et al.: Immunohistochemical expression of WT1 by desmoplastic small round cell tumor: a comparative study with other small round cell tumors. Am J Surg Pathol 24 (6): 830-6, 2000. [PUBMED Abstract]
Wang LL, Perlman EJ, Vujanic GM, et al.: Desmoplastic small round cell tumor of the kidney in childhood. Am J Surg Pathol 31 (4): 576-84, 2007. [PUBMED Abstract]
Errani C, Zhang L, Sung YS, et al.: A novel WWTR1-CAMTA1 gene fusion is a consistent abnormality in epithelioid hemangioendothelioma of different anatomic sites. Genes Chromosomes Cancer 50 (8): 644-53, 2011. [PUBMED Abstract]
Haller F, Knopf J, Ackermann A, et al.: Paediatric and adult soft tissue sarcomas with NTRK1 gene fusions: a subset of spindle cell sarcomas unified by a prominent myopericytic/haemangiopericytic pattern. J Pathol 238 (5): 700-10, 2016. [PUBMED Abstract]
Jain S, Xu R, Prieto VG, et al.: Molecular classification of soft tissue sarcomas and its clinical applications. Int J Clin Exp Pathol 3 (4): 416-28, 2010. [PUBMED Abstract]
Mariño-Enríquez A, Wang WL, Roy A, et al.: Epithelioid inflammatory myofibroblastic sarcoma: An aggressive intra-abdominal variant of inflammatory myofibroblastic tumor with nuclear membrane or perinuclear ALK. Am J Surg Pathol 35 (1): 135-44, 2011. [PUBMED Abstract]
Lovly CM, Gupta A, Lipson D, et al.: Inflammatory myofibroblastic tumors harbor multiple potentially actionable kinase fusions. Cancer Discov 4 (8): 889-95, 2014. [PUBMED Abstract]
Agaimy A, Bieg M, Michal M, et al.: Recurrent Somatic PDGFRB Mutations in Sporadic Infantile/Solitary Adult Myofibromas But Not in Angioleiomyomas and Myopericytomas. Am J Surg Pathol 41 (2): 195-203, 2017. [PUBMED Abstract]
Spunt SL, Hill DA, Motosue AM, et al.: Clinical features and outcome of initially unresected nonmetastatic pediatric nonrhabdomyosarcoma soft tissue sarcoma. J Clin Oncol 20 (15): 3225-35, 2002. [PUBMED Abstract]
Spunt SL, Poquette CA, Hurt YS, et al.: Prognostic factors for children and adolescents with surgically resected nonrhabdomyosarcoma soft tissue sarcoma: an analysis of 121 patients treated at St Jude Children’s Research Hospital. J Clin Oncol 17 (12): 3697-705, 1999. [PUBMED Abstract]
Ferrari A, Casanova M, Collini P, et al.: Adult-type soft tissue sarcomas in pediatric-age patients: experience at the Istituto Nazionale Tumori in Milan. J Clin Oncol 23 (18): 4021-30, 2005. [PUBMED Abstract]
Brooks AD, Heslin MJ, Leung DH, et al.: Superficial extremity soft tissue sarcoma: an analysis of prognostic factors. Ann Surg Oncol 5 (1): 41-7, 1998 Jan-Feb. [PUBMED Abstract]
Ferrari A, Miceli R, Meazza C, et al.: Soft tissue sarcomas of childhood and adolescence: the prognostic role of tumor size in relation to patient body size. J Clin Oncol 27 (3): 371-6, 2009. [PUBMED Abstract]
Rodeberg DA, Stoner JA, Garcia-Henriquez N, et al.: Tumor volume and patient weight as predictors of outcome in children with intermediate risk rhabdomyosarcoma: a report from the Children’s Oncology Group. Cancer 117 (11): 2541-50, 2011. [PUBMED Abstract]
Hong DS, DuBois SG, Kummar S, et al.: Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 21 (4): 531-540, 2020. [PUBMED Abstract]
Spunt SL, Million L, Chi YY, et al.: A risk-based treatment strategy for non-rhabdomyosarcoma soft-tissue sarcomas in patients younger than 30 years (ARST0332): a Children’s Oncology Group prospective study. Lancet Oncol 21 (1): 145-161, 2020. [PUBMED Abstract]
Million L, Hayes-Jordan A, Chi YY, et al.: Local Control For High-Grade Nonrhabdomyosarcoma Soft Tissue Sarcoma Assigned to Radiation Therapy on ARST0332: A Report From the Childrens Oncology Group. Int J Radiat Oncol Biol Phys 110 (3): 821-830, 2021. [PUBMED Abstract]
O’Sullivan B, Davis AM, Turcotte R, et al.: Preoperative versus postoperative radiotherapy in soft-tissue sarcoma of the limbs: a randomised trial. Lancet 359 (9325): 2235-41, 2002. [PUBMED Abstract]
Ferrari A, Miceli R, Rey A, et al.: Non-metastatic unresected paediatric non-rhabdomyosarcoma soft tissue sarcomas: results of a pooled analysis from United States and European groups. Eur J Cancer 47 (5): 724-31, 2011. [PUBMED Abstract]
Smith KB, Indelicato DJ, Knapik JA, et al.: Definitive radiotherapy for unresectable pediatric and young adult nonrhabdomyosarcoma soft tissue sarcoma. Pediatr Blood Cancer 57 (2): 247-51, 2011. [PUBMED Abstract]
Ferrari A, Magni C, Bergamaschi L, et al.: Pediatric nonrhabdomyosarcoma soft tissue sarcomas arising at visceral sites. Pediatr Blood Cancer 64 (9): , 2017. [PUBMED Abstract]
Ferrari A, van Noesel MM, Brennan B, et al.: Paediatric non-rhabdomyosarcoma soft tissue sarcomas: the prospective NRSTS 2005 study by the European Pediatric Soft Tissue Sarcoma Study Group (EpSSG). Lancet Child Adolesc Health 5 (8): 546-558, 2021. [PUBMED Abstract]
Dillon PW, Whalen TV, Azizkhan RG, et al.: Neonatal soft tissue sarcomas: the influence of pathology on treatment and survival. Children’s Cancer Group Surgical Committee. J Pediatr Surg 30 (7): 1038-41, 1995. [PUBMED Abstract]
Pappo AS, Fontanesi J, Luo X, et al.: Synovial sarcoma in children and adolescents: the St Jude Children’s Research Hospital experience. J Clin Oncol 12 (11): 2360-6, 1994. [PUBMED Abstract]
Marcus KC, Grier HE, Shamberger RC, et al.: Childhood soft tissue sarcoma: a 20-year experience. J Pediatr 131 (4): 603-7, 1997. [PUBMED Abstract]
Pratt CB, Pappo AS, Gieser P, et al.: Role of adjuvant chemotherapy in the treatment of surgically resected pediatric nonrhabdomyosarcomatous soft tissue sarcomas: A Pediatric Oncology Group Study. J Clin Oncol 17 (4): 1219, 1999. [PUBMED Abstract]
Pratt CB, Maurer HM, Gieser P, et al.: Treatment of unresectable or metastatic pediatric soft tissue sarcomas with surgery, irradiation, and chemotherapy: a Pediatric Oncology Group study. Med Pediatr Oncol 30 (4): 201-9, 1998. [PUBMED Abstract]
Histopathological Classification of Childhood Soft Tissue Sarcoma
World Health Organization (WHO) Classification of Soft Tissue Tumors
The WHO classification system for cancer represents the common nomenclature for cancer worldwide. In the United States, it has been adopted by the American Joint Committee on Cancer (AJCC) for sarcoma staging and the College of American Pathologists (CAP) cancer protocols for bone and soft tissue sarcomas. The WHO published a revision to their classification of soft tissue and bone tumors in 2020. The classification had several updates to existing classification, nomenclature, grading, and risk stratification schemes. The revised classification includes newly described entities, and it uses molecular alterations in the classifications.[1]
The grading of soft tissue tumors has always been a controversial issue. The 2020 WHO classification represents the consensus of several soft tissue pathologists and geneticists, as well as a medical oncologist, radiologist, and surgeon. This edition further integrates morphological and genetic information into the classification. For example, a new category of tumors called NTRK-rearranged spindle cell neoplasms was included, but infantile fibrosarcoma was excluded from this group. Ewing sarcoma was removed from the bone tumor section and, instead, is in the undifferentiated small cell sarcomas of bone and soft tissue section. This classification reflects the variable presentation sites and the variety of translocations seen in Ewing sarcoma. This classification also separated Ewing sarcoma from entities such as CIC-rearranged sarcomas, BCOR-rearranged sarcomas, and EWSR1 gene fusions involving non-ETS partner genes.[1]
With the increased use of next-generation sequencing techniques and heightened awareness of recently approved tyrosine kinase inhibitors that target NTRK and other genes, newer subgroups of pediatric soft tissue lesions that are characterized by kinase fusions have been identified and share a similar morphological spectrum. Identifying these rare entities is important because some of them might be amenable to therapeutic targeting with novel agents. Some examples of these lesions are described below.[4]
Lipofibromatosis-like neural tumors are superficial tumors that commonly affect children, and the cells are immunoreactive for S100. These tumors commonly have NTRK1 fusions but rarely harbor RET or ALK fusions.
Spindle cell tumors with S100 and CD34 positivity that resemble intermediate-grade malignant peripheral nerve sheath tumors predominate in children and young adults and can affect bone and soft tissues. They have fusions in various genes, including RAF1, BRAF, NTRK1, and NTRK2.
Infantile fibrosarcoma–like lesions morphologically resemble infantile fibrosarcoma and most commonly affect patients younger than 2 years. They have a predilection for intraabdominal sites. They often exhibit alternate fusions, involving genes such as BRAF, NTRK1, and MET.
Spindle cell sarcomas with hemangiopericytic and myopericytic patterns can affect children and have NTRK1 fusions.
RAF1 fusion–positive spindle cell sarcomas can be seen in children and adults and often arise in the trunk. They rarely behave aggressively.
BRAF fusion–positive soft tissue tumors have been associated with infantile fibrosarcoma–like variants or spindle cell sarcomas that resemble malignant peripheral nerve sheath tumors. They have been reported in children and often involve the abdominal cavity.
RET fusion–positive tumors predominantly affect children and have a similar phenotype to NTRK fusion–positive tumors. They can display fibroblastic and neural-like differentiation. These tumors are sensitive to the highly selective small-molecule RET inhibitor selpercatinib.[5]
References
WHO Classification of Tumours Editorial Board: WHO Classification of Tumours. Volume 3: Soft Tissue and Bone Tumours. 5th ed., IARC Press, 2020.
Steelman C, Katzenstein H, Parham D, et al.: Unusual presentation of congenital infantile fibrosarcoma in seven infants with molecular-genetic analysis. Fetal Pediatr Pathol 30 (5): 329-37, 2011. [PUBMED Abstract]
Evans HL: Low-grade fibromyxoid sarcoma: a clinicopathologic study of 33 cases with long-term follow-up. Am J Surg Pathol 35 (10): 1450-62, 2011. [PUBMED Abstract]
Antonescu CR: Emerging soft tissue tumors with kinase fusions: An overview of the recent literature with an emphasis on diagnostic criteria. Genes Chromosomes Cancer 59 (8): 437-444, 2020. [PUBMED Abstract]
Ortiz MV, Gerdemann U, Raju SG, et al.: Activity of the Highly Specific RET Inhibitor Selpercatinib (LOXO-292) in Pediatric Patients With Tumors Harboring RET Gene Alterations. JCO Precis Oncol 4: , 2020. [PUBMED Abstract]
Staging and Grading Systems for Childhood Soft Tissue Sarcoma
Assessment of disease extent has an important role in predicting the clinical outcome and determining the most effective therapy for pediatric soft tissue sarcomas. As yet, there is no well-accepted assessment system that is applicable to all childhood sarcomas. The system from the American Joint Committee on Cancer (AJCC) that is used for adults has not been validated in pediatric studies.
No standardized staging system for pediatric nonrhabdomyosarcomatous soft tissue sarcomas (NRSTS) exists, but two systems are currently used to assess disease extent:[1]
Surgico-pathological group system: The surgico-pathological group system used by the Intergroup Rhabdomyosarcoma Study is based on the amount, or extent, of tumor that remains after initial surgery and whether the disease has metastasized. This group system was used in early pediatric trials.[2] For more information, see the Intergroup Rhabdomyosarcoma Study Clinical Group System section.
TNM staging system: The TNM staging system is a collaborative effort between the AJCC (United States) and the International Union Against Cancer (worldwide). Staging is based on the extent of the tumor (T), the extent of spread to the lymph nodes (N), the presence of metastasis (M), and the tumor grade. For the staging of soft tissue sarcoma from the eighth edition of the AJCC Cancer Staging Manual, see Tables 6, 7, 8, and 9.[3–7] The last Children’s Oncology Group (COG) trial used the sixth edition AJCC Cancer Staging Manual for soft tissue sarcoma (with central pathology review).[1] A review of children with NRSTS was performed with data from the Surveillance, Epidemiology, and End Results (SEER) Program and identified 941 patients between 1988 and 2007.[8] The COG risk stratification was validated in this cohort.
Intergroup Rhabdomyosarcoma Study Clinical Group System
Nonmetastatic disease
Group I: Localized tumor completely resected with histologically negative margins.
Group II: Grossly resected tumor with microscopic residual tumor at the margin(s) and/or extension into regional lymph nodes.
Group IIA: Localized, grossly resected tumor with microscopic residual disease.
Group IIB: Regional disease with involved nodes completely resected with no microscopic disease. The most proximal (to the patient, most distal to the tumor) regional lymph node must be negative.
Group IIC: Regional disease with involved nodes grossly resected but with evidence of residual microscopic disease at the primary site and/or histologic involvement of the most proximal regional lymph node in the dissection.
Group III: Localized tumor, incompletely resected, or biopsy only, with gross residual tumor.
Metastatic disease
Group IV: Any localized or regional tumor with distant metastases present at the time of diagnosis. This includes the presence of malignant cells in effusions (pleural, peritoneal) and/or cerebrospinal fluid (rare).
Recurrent/progressive disease
Any soft tissue sarcoma that recurs after initial treatment or progresses after radiation therapy, chemotherapy, or initial surgery.
TNM Staging System
The eighth edition of the AJCC Cancer Staging Manual has designated staging by the four criteria of tumor size, nodal status, histological grade, and metastasis and by anatomical primary tumor site (head and neck; trunk and extremities; abdomen and thoracic visceral organs; retroperitoneum; and unusual histologies and sites) (see Tables 6, 7, 8, and 9).[3–7] For information about unusual histologies and sites, see the AJCC Cancer Staging Manual.[7]
Table 6. Definition of Primary Tumor (T) for Soft Tissue Sarcoma of the Trunk, Extremities, and Retroperitoneum; Head and Neck; and Abdomen and Thoracic Visceral Organsa
T Category
Soft Tissue Sarcoma of the Trunk, Extremities, and Retroperitoneum
Soft Tissue Sarcoma of the Head and Neck
Soft Tissue Sarcoma of the Abdomen and Thoracic Visceral Organs
aAdapted from O’Sullivan et al.,[3] Yoon et al.,[4] Raut et al.,[5] and Pollock et al.[6]
TX
Primary tumor cannot be assessed.
Primary tumor cannot be assessed.
Primary tumor cannot be assessed.
T0
No evidence of primary tumor.
T1
Tumor ≤5 cm in greatest dimension.
Tumor ≤2 cm.
Organ confined.
T2
Tumor >5 cm and ≤10 cm in greatest dimension.
Tumor >2 to ≤4 cm.
Tumor extension into tissue beyond organ.
T2a
Invades serosa or visceral peritoneum.
T2b
Extension beyond serosa (mesentery).
T3
Tumor >10 cm and ≤15 cm in greatest dimension.
Tumor >4 cm.
Invades another organ.
T4
Tumor >15 cm in greatest dimension.
Tumor with invasion of adjoining structures.
Multifocal involvement.
T4a
Tumor with orbital invasion, skull base/dural invasion, invasion of central compartment viscera, involvement of facial skeleton, or invasion of pterygoid muscles.
Multifocal (2 sites).
T4b
Tumor with brain parenchymal invasion, carotid artery encasement, prevertebral muscle invasion, or central nervous system involvement via perineural spread.
Multifocal (3–5 sites).
T4c
Multifocal (>5 sites).
Table 7. Definition of Regional Lymph Node (N) for Soft Tissue Sarcoma of the Head and Neck; Trunk and Extremities; Abdomen and Thoracic Visceral Organs; and Retroperitoneuma
aAdapted from O’Sullivan et al.,[3] Yoon et al.,[4] Raut et al.,[5] and Pollock et al.[6]
bFor soft tissue sarcoma of the abdomen and thoracic visceral organs, N0 = no lymph node involvement or unknown lymph node status and N1 = lymph node involvement present.
N0
No regional lymph node metastasis or unknown lymph node status.b
N1
Regional lymph node metastasis.b
Table 8. Definition of Distant Metastasis (M) for Soft Tissue Sarcoma of the Head and Neck; Trunk and Extremities; Abdomen and Thoracic Visceral Organs; and Retroperitoneuma
aAdapted from O’Sullivan et al.,[3] Yoon et al.,[4] Raut et al.,[5] and Pollock et al.[6]
bFor soft tissue sarcoma of the abdomen and thoracic visceral organs, M0 = no metastases and M1 = metastases present.
M0
No distant metastasis.b
M1
Distant metastasis.b
Table 9. AJCC Prognostic Stage Groups for Soft Tissue Sarcoma of the Trunk, Extremities, and Retroperitoneuma
Stage
T
N
M
Grade
T = primary tumor; N = regional lymph node; M = distant metastasis.
aAdapted from Yoon et al. [4] and Pollock et al.[6]
bStage IIIB for soft tissue sarcoma of the retroperitoneum; stage IV for soft tissue sarcoma of the trunk and extremities.
IA
T1
N0
M0
G1, GX
IB
T2, T3, T4
N0
M0
G1, GX
II
T1
N0
M0
G2, G3
IIIA
T2
N0
M0
G2, G3
IIIB
T3, T4
N0
M0
G2, G3
IIIB/IVb
Any T
N1
M0
Any G
IV
Any T
Any N
M1
Any G
Soft Tissue Sarcoma Tumor Pathological Grading System
In most cases of soft tissue sarcomas, accurate histopathological classification alone does not yield optimal information about their clinical behavior. Therefore, several histological parameters are evaluated in the grading process, including the following:
Degree of cellularity.
Cellular pleomorphism.
Mitotic activity.
Degree of necrosis.
Invasive growth.
This process is used to improve the correlation between histological findings and clinical outcome.[9] In children, grading of soft tissue sarcoma is complicated by certain factors, such as prognosis, patient age, extent of surgical resection, and ability to metastasize. For example, children younger than 4 years with infantile fibrosarcoma and hemangiopericytoma have a good prognosis, and angiomatoid fibrous histiocytoma and dermatofibrosarcoma protuberans can recur locally if incompletely excised but usually do not metastasize.
Testing the validity of a grading system within the pediatric population is difficult because of the rarity of these neoplasms. In 1986, the Pediatric Oncology Group (POG) conducted a prospective study on pediatric NRSTS and devised the POG grading system. Analysis of outcomes for patients with localized NRSTS demonstrated that patients with grade 3 tumors fared significantly worse than those with grade 1 or grade 2 lesions. This finding suggests that this system can accurately predict the clinical behavior of NRSTS.[9–11]
The POG and Fédération Nationale des Centres de Lutte Contre Le Cancer (FNCLCC) grading systems have proven to be of prognostic value in pediatric and adult NRSTS.[12–16] The COG uses the FNCLCC clinically. In a study of 130 tumors from children and adolescents with NRSTS enrolled in three prospective clinical trials, a correlation was found between the POG-assigned grade and the FNCLCC-assigned grade. However, grading did not correlate in all cases; 44 patients whose tumors received discrepant grades (POG grade 3, FNCLCC grade 1 or 2) had outcomes between concurrent grade 3 and grades 1 and 2. A mitotic index of 10 or greater emerged as an important prognostic factor.[17]
The COG ARST0332 (NCT00346164) trial compared the POG and FNCLCC pathological grading systems as part of a prospective risk-based strategy. The study found that, in addition to tumor depth and invasiveness, the FNCLCC grade was strongly associated with event-free survival and overall survival.[18] The closed COG ARST1321 (NCT02180867) trial used the FNCLCC system to assign histological grade.
The FNCLCC Sarcoma Group is described below. The POG grading system is no longer used.
FNCLCC grading system
The FNCLCC histological grading system was developed for adults with soft tissue sarcoma. The purpose of the grading system is to predict which patients will develop metastasis and subsequently benefit from postoperative chemotherapy.[19,20] For information about the FNCLCC histological grading system for adults, see the FNCLCC histological grade section in Soft Tissue Sarcoma Treatment.
References
American Joint Committee on Cancer: AJCC Cancer Staging Manual. 6th ed. Springer, 2002.
Maurer HM, Beltangady M, Gehan EA, et al.: The Intergroup Rhabdomyosarcoma Study-I. A final report. Cancer 61 (2): 209-20, 1988. [PUBMED Abstract]
O’Sullivan B, Maki RG, Agulnik M, et al.: Soft tissue sarcoma of the head and neck. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp 499-505.
Yoon SS, Maki RG, Asare EA, et al.: Soft tissue sarcoma of the trunk and extremities. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp 507-15.
Raut CP, Maki RG, Baldini EH, et al.: Soft tissue sarcoma of the abdomen and thoracic visceral organs. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp 517-21.
Pollock RE, Maki RG, Baldini EH, et al.: Soft tissue sarcoma of the retroperitoneum. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp 531-7.
Maki RG, Folpe AL, Guadagnolo BA, et al.: Soft tissue sarcoma – unusual histologies and sites. In: Amin MB, Edge SB, Greene FL, et al., eds.: AJCC Cancer Staging Manual. 8th ed. Springer; 2017, pp 539-45.
Waxweiler TV, Rusthoven CG, Proper MS, et al.: Non-Rhabdomyosarcoma Soft Tissue Sarcomas in Children: A Surveillance, Epidemiology, and End Results Analysis Validating COG Risk Stratifications. Int J Radiat Oncol Biol Phys 92 (2): 339-48, 2015. [PUBMED Abstract]
Parham DM, Webber BL, Jenkins JJ, et al.: Nonrhabdomyosarcomatous soft tissue sarcomas of childhood: formulation of a simplified system for grading. Mod Pathol 8 (7): 705-10, 1995. [PUBMED Abstract]
Recommendations for the reporting of soft tissue sarcomas. Association of Directors of Anatomic and Surgical Pathology. Mod Pathol 11 (12): 1257-61, 1998. [PUBMED Abstract]
Skytting B, Meis-Kindblom JM, Larsson O, et al.: Synovial sarcoma–identification of favorable and unfavorable histologic types: a Scandinavian sarcoma group study of 104 cases. Acta Orthop Scand 70 (6): 543-54, 1999. [PUBMED Abstract]
Pisters PW, Leung DH, Woodruff J, et al.: Analysis of prognostic factors in 1,041 patients with localized soft tissue sarcomas of the extremities. J Clin Oncol 14 (5): 1679-89, 1996. [PUBMED Abstract]
Coindre JM, Terrier P, Bui NB, et al.: Prognostic factors in adult patients with locally controlled soft tissue sarcoma. A study of 546 patients from the French Federation of Cancer Centers Sarcoma Group. J Clin Oncol 14 (3): 869-77, 1996. [PUBMED Abstract]
Pappo AS, Fontanesi J, Luo X, et al.: Synovial sarcoma in children and adolescents: the St Jude Children’s Research Hospital experience. J Clin Oncol 12 (11): 2360-6, 1994. [PUBMED Abstract]
Pratt CB, Maurer HM, Gieser P, et al.: Treatment of unresectable or metastatic pediatric soft tissue sarcomas with surgery, irradiation, and chemotherapy: a Pediatric Oncology Group study. Med Pediatr Oncol 30 (4): 201-9, 1998. [PUBMED Abstract]
Khoury JD, Coffin CM, Spunt SL, et al.: Grading of nonrhabdomyosarcoma soft tissue sarcoma in children and adolescents: a comparison of parameters used for the Fédération Nationale des Centers de Lutte Contre le Cancer and Pediatric Oncology Group Systems. Cancer 116 (9): 2266-74, 2010. [PUBMED Abstract]
Spunt SL, Million L, Chi YY, et al.: A risk-based treatment strategy for non-rhabdomyosarcoma soft-tissue sarcomas in patients younger than 30 years (ARST0332): a Children’s Oncology Group prospective study. Lancet Oncol 21 (1): 145-161, 2020. [PUBMED Abstract]
Coindre JM, Terrier P, Guillou L, et al.: Predictive value of grade for metastasis development in the main histologic types of adult soft tissue sarcomas: a study of 1240 patients from the French Federation of Cancer Centers Sarcoma Group. Cancer 91 (10): 1914-26, 2001. [PUBMED Abstract]
Guillou L, Coindre JM, Bonichon F, et al.: Comparative study of the National Cancer Institute and French Federation of Cancer Centers Sarcoma Group grading systems in a population of 410 adult patients with soft tissue sarcoma. J Clin Oncol 15 (1): 350-62, 1997. [PUBMED Abstract]
Treatment Option Overview for Childhood Soft Tissue Sarcoma
Because of the rarity of pediatric nonrhabdomyosarcomatous soft tissue sarcomas (NRSTS), treatment should be coordinated by a multidisciplinary team that includes oncologists (pediatric or medical), pathologists, surgeons, and radiation oncologists for all children, adolescents, and young adults with these tumors. In addition, to better define the tumors’ natural history and response to therapy, entry into national or institutional treatment protocols should be considered for children with rare neoplasms. Information about ongoing clinical trials is available from the NCI website.
The Children’s Oncology Group (COG) performed a prospective nonrandomized trial (ARST0332 [NCT00346164]) for patients with soft tissue sarcomas.[1]
Surgical resection of the primary tumor was classified as follows:
R0 if the resection was complete with negative microscopic margins.
R1 if the margins were microscopically positive.
R2 if the resection left macroscopic residual tumor.
Patients were assigned to one of the following three risk groups:
Low risk: Nonmetastatic R0 or R1 low-grade, or ≤5 cm R1 high-grade tumor.
Intermediate risk: Nonmetastatic R0 or R1 >5 cm high-grade, or unresected tumor of any size or grade.
High risk: Metastatic tumor.
The treatment groups were as follows:
Surgery alone.
Radiation therapy (55.8 Gy).
Chemoradiation therapy (chemotherapy and 55.8 Gy radiation therapy).
Neoadjuvant chemoradiation therapy (chemotherapy and 45 Gy radiation therapy, then surgery and radiation therapy boost based on margins with continued chemotherapy).
Chemotherapy included six cycles of intravenous (IV) ifosfamide (3 g/m2 per dose) on days 1 through 3 and five cycles of IV doxorubicin (37.5 mg/m2 per dose) on days 1 to 2 every 3 weeks, with the sequence adjusted based on timing of surgery or radiation therapy.
The analysis included 529 evaluable patients: low risk (n = 222), intermediate risk (n = 227), and high risk (n = 80). Patients underwent surgery alone (n = 205), radiation therapy (n = 17), chemoradiation therapy (n = 111), and neoadjuvant chemoradiation therapy (n = 196).
At a median follow-up of 6.5 years (interquartile range [IQR], 4.9–7.9), the 5-year event-free survival (EFS) and overall survival (OS) rates, by risk group, were as follows:
Low-risk group: EFS rate, 88.9% (95% confidence interval [CI], 84.0%–93.8%) and OS rate, 96.2% (95% CI, 93.2%–99.2%).
Intermediate-risk group: EFS rate, 65.0% (95% CI, 58.2%–71.8%) and OS rate, 79.2% (95% CI, 73.4%–85.0%).
High-risk group: EFS rate, 21.2% (95% CI, 11.4%–31.1%) and OS rate, 35.5% (95% CI, 23.6%–47.4%).
The authors concluded that pretreatment clinical features can be used to effectively define treatment failure risk and stratify young patients with NRSTS for risk-adapted therapy. Most low-risk patients can be cured without adjuvant therapy, avoiding known long-term treatment complications. Survival remains suboptimal for intermediate-risk and high-risk patients, and novel therapies are needed for these patients.
Surgery
Surgical resection of the primary tumor is the predominant therapy for most NRSTS. In the COG ARST0332 (NCT00346164) study, approximately 37% of patients younger than 30 years were treated with surgery alone.[1] Another 36% of patients had surgical resection after neoadjuvant chemoradiation therapy. Involvement of a surgeon with special expertise in the resection of soft tissue sarcomas is highly desirable.
After an appropriate biopsy and pathological diagnosis, every attempt is made to resect the primary tumor. Completeness of resection predicts outcome. In the COG ARST0332 study, complete resections with negative microscopic margins (R0) resulted in the best outcomes.[1]
The 5-year EFS rates for patients treated with surgery and other modalities were the following:
84% for patients who had R0 resections.
66% for patients who had R1 resections.
49% for patients who had R2 resections.
The 5-year OS rates for patients treated with surgery and other modalities were the following:
93% for patients who had R0 resections.
80% for patients who had R1 resections.
63% for patients who had R2 resections.
The 5-year EFS rates for patients treated with surgery only were the following:
96% for patients with low-grade tumors who had R0 resections.
81% for patients with low-grade tumors who had R1 resections.
84% for patients with high-grade tumors that were smaller than 5 cm and had R0 resections.
The COG reported results for the subset of patients with low-grade NRSTS enrolled in the ARST0332 study.[2] Low-risk patients were treated with surgery alone. Intermediate- and high-risk patients received ifosfamide/doxorubicin and radiation therapy, with definitive resection either before or after 12 weeks of chemotherapy and radiation therapy.
Table 10. Survival Results From the ARST0332 Study
Risk Group
5-Year Event-Free Survival Rate
5-Year Overall Survival Rate
Low risk
90%
100%
Intermediate risk
55%
78%
High risk
25%
25%
In low-risk patients, local-only recurrences were seen in 10% of patients. No patients with margins of resection greater than 1 mm had local recurrences.
Sixteen of 17 intermediate- and high-risk patients who completed neoadjuvant chemotherapy and radiation therapy underwent gross-total tumor resection, and 80% had negative margins.
In the intermediate- and high-risk groups, events included one local recurrence and seven metastatic recurrences.
The timing of surgery depends on an assessment of the feasibility and morbidity of surgery. In the COG ARST0332 study, if the central review surgeon deemed the tumor unresectable without loss of limb, form, or function, the patient was treated in arm C with neoadjuvant radiation therapy. Surgery was performed 4 to 6 weeks after the completion of radiation therapy. This early surgery allowed for decreased morbidity, better wound healing, and more complete surgical resection. The outcomes in the COG ARST0332 study were nearly identical for intermediate-risk patients who achieved an R0 or R1 resection with up-front surgery or surgery after neoadjuvant chemoradiation therapy (70% vs. 71%, respectively). An R0 resection was more likely to occur after neoadjuvant therapy.[1] These observations are true even for high-grade tumors, where the ability to achieve R0 or R1 resections was the major predictor of EFS. Treatment with neoadjuvant chemoradiation therapy resulted in lower doses of radiation therapy and achieved greater rates of R0 resections.[3] Resectability should be determined at the time of diagnosis. While there should be an emphasis on achieving negative margins without loss of form or function, given the variability of chemosensitivity of such diverse tumors, it may be better to tailor the resection to histology for each patient.
If the initial operation fails to achieve pathologically negative tissue margins or if the initial surgery was done without the knowledge that cancer was present, a re-excision of the affected area is performed to obtain clear, but not necessarily wide, margins.[4–7] This surgical tenet is true even if no mass is detected by magnetic resonance imaging after initial surgery.[8]; [9][Level of evidence C1]
Regional lymph node metastases at diagnosis are unusual and are most often seen in patients with epithelioid and clear cell sarcomas.[10,11] Sentinel lymph node biopsy as a staging procedure in soft tissue sarcoma remains controversial. However, it may help manage selected cases in adults with clear cell sarcoma and epithelioid sarcoma. There are insufficient data to support the use of sentinel lymph node biopsy in the management of pediatric patients with other NRSTS.[12–17]
Radiation Therapy
Considerations for radiation therapy are based on the potential for surgery, with or without chemotherapy, to obtain local control without severe injury to critical organs, compromise of function, or significant cosmetic or psychological impairment. This will vary according to the following:
Patient variables (e.g., age and sex).
Tumor variables (e.g., histopathology, site, size, and grade).
Surgery and subsequent margin status.
Expectations for radiation-induced morbidities (e.g., impaired bone or muscle development, organ damage, or subsequent neoplasms).
Radiation therapy can be given preoperatively or postoperatively. It can also be used as definitive therapy in rare situations in which surgical resection is not performed.[18] Radiation field size and dose will be based on patient and tumor variables and the surgical procedure.[19] Radiation therapy is associated with improved OS compared with surgery alone when delivered preoperatively or postoperatively.[20]
Brachytherapy and intraoperative radiation may be applicable in select situations.[21–23]; [24][Level of evidence C2]
Preoperative radiation therapy
Preoperative radiation therapy has been associated with excellent local control rates.[25–27] The advantages of this approach include treating smaller tissue volumes without the need to treat a postsurgical bed and somewhat lower radiation doses because relative hypoxia from surgical disruption of vasculature and scarring is not present. Preoperative radiation therapy has been associated with an increased rate of wound complications in adults, primarily in lower extremity tumors. However, the degree of these complications is questionable.[28] Conversely, preoperative radiation therapy may lead to less fibrosis than with postoperative approaches, perhaps because of the smaller treatment volume and dose.[29] Radiation techniques, like proton-beam radiation therapy can facilitate normal tissue sparing. Compared with 3-dimensional conformal radiation therapy, intensity-modulated radiation therapy may decrease radiation dose to the skin and epiphysis when irradiating extremity sarcomas, which can translate into decreased fibrosis or growth impairment.[30,31]
Postoperative radiation therapy
Radiation therapy can also be given postoperatively. In general, radiation is indicated for patients with inadequate surgical margins and for larger, high-grade tumors.[32,33] This is particularly important in high-grade tumors with tumor margins smaller than 1 cm.[34,35]; [36][Level of evidence C3] With combined R0 (negative margin) surgery and radiation therapy, local control of the primary tumor can be achieved in about 90% of patients with extremity sarcomas, 70% to 75% of patients with retroperitoneal sarcomas, and 80% of patients overall.[21,37–40]
Retroperitoneal sarcomas are unique in that the radiosensitivity of the bowel increases the risk of injury and makes postoperative radiation therapy less desirable.[41,42] Postoperative adhesions and bowel immobility can increase the risk of damage from any given radiation dose. This contrasts with the preoperative approach in which the tumor often displaces bowel outside of the radiation field, and any exposed bowel is more mobile, which decreases exposure to specific bowel segments.
Dose and volume
Radiation volume and dose depend on the patient, tumor, and surgical variables noted above, as well as the following:
Patient age and growth potential.
Ability to avoid critical organs, epiphyseal plates, and lymphatics (but not the neurovascular bundles that are relatively radiation tolerant).
Functional/cosmetic outcome.
Radiation doses are typically 45 Gy to 50 Gy preoperatively, and as high as 60 Gy to very small volumes at highest risk when postoperative resection margins are predicted to be microscopically or grossly positive. Planned brachytherapy is an option if the resection is predicted to be subtotal. This can be accomplished with a simultaneously integrated boost dose (i.e., higher dose area within the larger lower dose volume) or administered with a small field of radiation after the initial volume is treated with a dose of 45 Gy to 50 Gy. However, data documenting the efficacy of a postoperative boost to areas with microscopically positive margins are lacking.[43] The postoperative radiation dose is 55 Gy to 60 Gy for R0 resections, up to 65 Gy for R1 resections (microscopic positive margins), and higher when unresectable gross residual disease exists, depending on overall treatment goals (e.g., definitive local control vs. palliation).
The COG analyzed local recurrence (LR) for NRSTS after radiation therapy in patients treated in the ARST0332 trial.[3] Patients younger than 30 years with high-grade NRSTS received radiation therapy (55.8 Gy) for an R1, 5 cm or smaller tumor (arm B); radiation therapy (55.8 Gy) with chemotherapy for an R0/R1, larger than 5 cm tumor (arm C); or neoadjuvant radiation therapy (45 Gy) with chemotherapy plus delayed surgery, chemotherapy, and postoperative boost to 10.8 Gy for an R0, smaller than 5 mm margins tumor or R1 tumor, or 19.8 Gy for R2 or unresected tumors (arm D).
Of 193 eligible patients, 24 had local recurrences (arm B: 1 of 15 [6.7%], arm C: 7 of 65 [10.8%], arm D: 16 of 113 [14.2%]) with a median time to local recurrence of 1.1 years (range, 0.11–5.27 years).
Of 95 patients eligible for delayed surgery after neoadjuvant therapy, 89 (93.7%) achieved R0/R1 margins.
Overall local control after radiation therapy were as follows: R0, 106 of 109 (97%); R1, 51 of 60 (85%); and R2/unresectable, 2 of 6 (33%).
The authors concluded that risk-based treatment for young patients with high-grade NRSTS treated on ARST0332 produced very high local control, particularly after R0 resection (97%), despite lower-than-standard radiation therapy doses.
Radiation margins are typically 2 cm to 4 cm longitudinally and encompass fascial planes axially.[44,45]
Radiation therapy was used in the COG ARST1321 trial.
Chemotherapy
The role of postoperative chemotherapy remains unclear.[46]
Evidence (lack of clarity regarding postoperative chemotherapy):
A meta-analysis of data from all randomized trials of adults with soft tissue sarcoma observed the following:[47]
Recurrence-free survival was better with postoperative chemotherapy for patients with high-grade tumors larger than 5 cm.
In a European trial, adults with completely resected soft tissue sarcoma were randomly assigned to observation or postoperative chemotherapy with ifosfamide and doxorubicin.[48][Level of evidence A1]
Postoperative chemotherapy was not associated with improved EFS or OS.
It is difficult to extrapolate this trial to pediatric patients because the trial included: (1) a wide variety of histologies; (2) a relatively low dose of ifosfamide; (3) patients assigned to chemotherapy had definitive radiation delayed until completion of chemotherapy; and (4) almost one-half of the patients in the trial had intermediate-grade tumors.
In the discussion section, the authors merged their patients with previously published series, including those from the European meta-analysis, and concluded that the results suggested a benefit for postoperative chemotherapy.
The largest prospective pediatric trial failed to demonstrate any benefit with postoperative vincristine, dactinomycin, cyclophosphamide, and doxorubicin.[37]
Although this was not a randomized study, results at 2.6 years showed that patients with high-risk (>5 cm and high grade), grossly resected, nonmetastatic tumors who were treated with radiation therapy and postoperative doxorubicin and ifosfamide had a 5-year EFS rate of 67.2% and an OS rate of 78%. However, this study did not have a comparison group of patients who did not receive chemotherapy.
In patients with metastatic disease treated with preoperative chemotherapy and radiation therapy, the estimated 5-year EFS rate was 21.2%, and the OS rate was 35.5%.
Targeted Therapy
The use of angiogenesis and mammalian target of rapamycin (mTOR) inhibitors has been explored in the treatment of adult patients with soft tissue sarcomas but not in pediatric patients. Other targeted therapy agents are reported for specific tumor types in the following sections.
Evidence (targeted therapy in adults with soft tissue sarcoma):
In a trial of 711 adult patients who achieved a response or stable disease after chemotherapy, patients were randomly assigned to receive ridaforolimus (a rapamycin inhibitor) or placebo.[49]
The administration of ridaforolimus was associated with a 3-week improvement in progression-free survival (PFS) when compared with placebo.
In another trial of 371 randomly assigned adult patients with metastatic soft tissue sarcoma that progressed after chemotherapy, pazopanib (a multitargeted receptor tyrosine kinase inhibitor) was compared with placebo.[50]
The median PFS for the pazopanib arm was 4.6 months, compared with 1.6 months for the placebo arm. OS was not different between the two arms.
In a study of 182 previously treated adult patients with recurrent liposarcoma, leiomyosarcoma, synovial sarcoma, and other sarcomas, patients were randomly assigned to receive either regorafenib or placebo.[51]
Patients with nonadipocytic tumors who were treated with regorafenib had significant improvements in PFS when compared with patients who were treated with placebo.
The COG and NRG Oncology cancer consortia conducted a randomized trial of pazopanib added to neoadjuvant chemotherapy (doxorubicin and ifosfamide) and preoperative radiation therapy in pediatric (older than 2 years) and adult patients with NRSTS. Patients with intermediate- or high-grade disease whose tumors were larger than 5 cm were eligible. The end point of the trial was pathological tumor response after adjuvant therapy. Study entry was closed early because the planned interim analysis showed that the pathological response boundary was crossed. Eighty-one patients were enrolled, but only 42 (52%) were available for response data (17 patients from each group discontinued therapy for either progression, unacceptable toxicity, or patient or physician choice).[52,53]
Four of 18 patients (22%) in the control group had greater than 90% necrosis at resection, compared with 14 of 24 patients (58%) in the group treated with pazopanib, meeting the criteria for early stopping of the study.
Toxicity was greater in the pazopanib group, mainly resulting from increased myelosuppression. Wound complications were also more frequent in the pazopanib group.
With longer follow-up, the investigators were able to analyze the secondary objectives of OS and EFS.[54] At a median follow-up of 3.3 years (range, 0.1–5.8 years), the 3-year EFS rate for all patients in the intent-to-treat analysis was 52.5% for patients who received pazopanib and 50.6% for those who did not (log-rank P = .8677). The 3-year OS rate was 75.7% for patients who received pazopanib and 65.4% for the control group (log-rank P = .1919). However, the study was not powered to evaluate these end points.
References
Spunt SL, Million L, Chi YY, et al.: A risk-based treatment strategy for non-rhabdomyosarcoma soft-tissue sarcomas in patients younger than 30 years (ARST0332): a Children’s Oncology Group prospective study. Lancet Oncol 21 (1): 145-161, 2020. [PUBMED Abstract]
Douglass DP, Coffin CM, Randall RL, et al.: Clinical features and outcomes of young patients with low-grade non-rhabdomyosarcoma soft tissue sarcomas treated with a risk-based strategy: A report from Children’s Oncology Group study ARST0332. Pediatr Blood Cancer 71 (8): e31062, 2024. [PUBMED Abstract]
Million L, Hayes-Jordan A, Chi YY, et al.: Local Control For High-Grade Nonrhabdomyosarcoma Soft Tissue Sarcoma Assigned to Radiation Therapy on ARST0332: A Report From the Childrens Oncology Group. Int J Radiat Oncol Biol Phys 110 (3): 821-830, 2021. [PUBMED Abstract]
Sugiura H, Takahashi M, Katagiri H, et al.: Additional wide resection of malignant soft tissue tumors. Clin Orthop (394): 201-10, 2002. [PUBMED Abstract]
Cecchetto G, Guglielmi M, Inserra A, et al.: Primary re-excision: the Italian experience in patients with localized soft-tissue sarcomas. Pediatr Surg Int 17 (7): 532-4, 2001. [PUBMED Abstract]
Chui CH, Spunt SL, Liu T, et al.: Is reexcision in pediatric nonrhabdomyosarcoma soft tissue sarcoma necessary after an initial unplanned resection? J Pediatr Surg 37 (10): 1424-9, 2002. [PUBMED Abstract]
Paulino AC, Ritchie J, Wen BC: The value of postoperative radiotherapy in childhood nonrhabdomyosarcoma soft tissue sarcoma. Pediatr Blood Cancer 43 (5): 587-93, 2004. [PUBMED Abstract]
Kaste SC, Hill A, Conley L, et al.: Magnetic resonance imaging after incomplete resection of soft tissue sarcoma. Clin Orthop (397): 204-11, 2002. [PUBMED Abstract]
Chandrasekar CR, Wafa H, Grimer RJ, et al.: The effect of an unplanned excision of a soft-tissue sarcoma on prognosis. J Bone Joint Surg Br 90 (2): 203-8, 2008. [PUBMED Abstract]
Daigeler A, Kuhnen C, Moritz R, et al.: Lymph node metastases in soft tissue sarcomas: a single center analysis of 1,597 patients. Langenbecks Arch Surg 394 (2): 321-9, 2009. [PUBMED Abstract]
Mazeron JJ, Suit HD: Lymph nodes as sites of metastases from sarcomas of soft tissue. Cancer 60 (8): 1800-8, 1987. [PUBMED Abstract]
Neville HL, Andrassy RJ, Lally KP, et al.: Lymphatic mapping with sentinel node biopsy in pediatric patients. J Pediatr Surg 35 (6): 961-4, 2000. [PUBMED Abstract]
Kayton ML, Delgado R, Busam K, et al.: Experience with 31 sentinel lymph node biopsies for sarcomas and carcinomas in pediatric patients. Cancer 112 (9): 2052-9, 2008. [PUBMED Abstract]
Dall’Igna P, De Corti F, Alaggio R, et al.: Sentinel node biopsy in pediatric patients: the experience in a single institution. Eur J Pediatr Surg 24 (6): 482-7, 2014. [PUBMED Abstract]
Parida L, Morrisson GT, Shammas A, et al.: Role of lymphoscintigraphy and sentinel lymph node biopsy in the management of pediatric melanoma and sarcoma. Pediatr Surg Int 28 (6): 571-8, 2012. [PUBMED Abstract]
Alcorn KM, Deans KJ, Congeni A, et al.: Sentinel lymph node biopsy in pediatric soft tissue sarcoma patients: utility and concordance with imaging. J Pediatr Surg 48 (9): 1903-6, 2013. [PUBMED Abstract]
Haas RL, Gronchi A, van de Sande MAJ, et al.: Perioperative Management of Extremity Soft Tissue Sarcomas. J Clin Oncol 36 (2): 118-124, 2018. [PUBMED Abstract]
Crompton JG, Ogura K, Bernthal NM, et al.: Local Control of Soft Tissue and Bone Sarcomas. J Clin Oncol 36 (2): 111-117, 2018. [PUBMED Abstract]
Nussbaum DP, Rushing CN, Lane WO, et al.: Preoperative or postoperative radiotherapy versus surgery alone for retroperitoneal sarcoma: a case-control, propensity score-matched analysis of a nationwide clinical oncology database. Lancet Oncol 17 (7): 966-975, 2016. [PUBMED Abstract]
Merchant TE, Parsh N, del Valle PL, et al.: Brachytherapy for pediatric soft-tissue sarcoma. Int J Radiat Oncol Biol Phys 46 (2): 427-32, 2000. [PUBMED Abstract]
Schomberg PJ, Gunderson LL, Moir CR, et al.: Intraoperative electron irradiation in the management of pediatric malignancies. Cancer 79 (11): 2251-6, 1997. [PUBMED Abstract]
Nag S, Shasha D, Janjan N, et al.: The American Brachytherapy Society recommendations for brachytherapy of soft tissue sarcomas. Int J Radiat Oncol Biol Phys 49 (4): 1033-43, 2001. [PUBMED Abstract]
Viani GA, Novaes PE, Jacinto AA, et al.: High-dose-rate brachytherapy for soft tissue sarcoma in children: a single institution experience. Radiat Oncol 3: 9, 2008. [PUBMED Abstract]
Virkus WW, Mollabashy A, Reith JD, et al.: Preoperative radiotherapy in the treatment of soft tissue sarcomas. Clin Orthop (397): 177-89, 2002. [PUBMED Abstract]
Zagars GK, Ballo MT, Pisters PW, et al.: Preoperative vs. postoperative radiation therapy for soft tissue sarcoma: a retrospective comparative evaluation of disease outcome. Int J Radiat Oncol Biol Phys 56 (2): 482-8, 2003. [PUBMED Abstract]
Dickie C, Parent A, Griffin AM, et al.: The value of adaptive preoperative radiotherapy in management of soft tissue sarcoma. Radiother Oncol 122 (3): 458-463, 2017. [PUBMED Abstract]
O’Sullivan B, Davis AM, Turcotte R, et al.: Preoperative versus postoperative radiotherapy in soft-tissue sarcoma of the limbs: a randomised trial. Lancet 359 (9325): 2235-41, 2002. [PUBMED Abstract]
Davis AM, O’Sullivan B, Turcotte R, et al.: Late radiation morbidity following randomization to preoperative versus postoperative radiotherapy in extremity soft tissue sarcoma. Radiother Oncol 75 (1): 48-53, 2005. [PUBMED Abstract]
Rao AD, Chen Q, Million L, et al.: Preoperative Intensity Modulated Radiation Therapy Compared to Three-Dimensional Conformal Radiation Therapy for High-Grade Extremity Sarcomas in Children: Analysis of the Children’s Oncology Group Study ARST0332. Int J Radiat Oncol Biol Phys 103 (1): 38-44, 2019. [PUBMED Abstract]
Seddon B, Grange FL, Simões R, et al.: The IMRiS Trial: A Phase 2 Study of Intensity Modulated Radiation Therapy in Extremity Soft Tissue Sarcoma. Int J Radiat Oncol Biol Phys 120 (4): 978-989, 2024. [PUBMED Abstract]
Marcus KC, Grier HE, Shamberger RC, et al.: Childhood soft tissue sarcoma: a 20-year experience. J Pediatr 131 (4): 603-7, 1997. [PUBMED Abstract]
Delaney TF, Kepka L, Goldberg SI, et al.: Radiation therapy for control of soft-tissue sarcomas resected with positive margins. Int J Radiat Oncol Biol Phys 67 (5): 1460-9, 2007. [PUBMED Abstract]
Blakely ML, Spurbeck WW, Pappo AS, et al.: The impact of margin of resection on outcome in pediatric nonrhabdomyosarcoma soft tissue sarcoma. J Pediatr Surg 34 (5): 672-5, 1999. [PUBMED Abstract]
Skytting B: Synovial sarcoma. A Scandinavian Sarcoma Group project. Acta Orthop Scand Suppl 291: 1-28, 2000. [PUBMED Abstract]
Hua C, Gray JM, Merchant TE, et al.: Treatment planning and delivery of external beam radiotherapy for pediatric sarcoma: the St. Jude Children’s Research Hospital experience. Int J Radiat Oncol Biol Phys 70 (5): 1598-606, 2008. [PUBMED Abstract]
Pratt CB, Pappo AS, Gieser P, et al.: Role of adjuvant chemotherapy in the treatment of surgically resected pediatric nonrhabdomyosarcomatous soft tissue sarcomas: A Pediatric Oncology Group Study. J Clin Oncol 17 (4): 1219, 1999. [PUBMED Abstract]
Karakousis CP, Driscoll DL: Treatment and local control of primary extremity soft tissue sarcomas. J Surg Oncol 71 (3): 155-61, 1999. [PUBMED Abstract]
Zagars GK, Ballo MT, Pisters PW, et al.: Prognostic factors for disease-specific survival after first relapse of soft-tissue sarcoma: analysis of 402 patients with disease relapse after initial conservative surgery and radiotherapy. Int J Radiat Oncol Biol Phys 57 (3): 739-47, 2003. [PUBMED Abstract]
Raut CP, Miceli R, Strauss DC, et al.: External validation of a multi-institutional retroperitoneal sarcoma nomogram. Cancer 122 (9): 1417-24, 2016. [PUBMED Abstract]
Baldini EH, Wang D, Haas RL, et al.: Treatment Guidelines for Preoperative Radiation Therapy for Retroperitoneal Sarcoma: Preliminary Consensus of an International Expert Panel. Int J Radiat Oncol Biol Phys 92 (3): 602-12, 2015. [PUBMED Abstract]
Bishop AJ, Zagars GK, Torres KE, et al.: Combined Modality Management of Retroperitoneal Sarcomas: A Single-Institution Series of 121 Patients. Int J Radiat Oncol Biol Phys 93 (1): 158-65, 2015. [PUBMED Abstract]
Al Yami A, Griffin AM, Ferguson PC, et al.: Positive surgical margins in soft tissue sarcoma treated with preoperative radiation: is a postoperative boost necessary? Int J Radiat Oncol Biol Phys 77 (4): 1191-7, 2010. [PUBMED Abstract]
Wang D, Bosch W, Kirsch DG, et al.: Variation in the gross tumor volume and clinical target volume for preoperative radiotherapy of primary large high-grade soft tissue sarcoma of the extremity among RTOG sarcoma radiation oncologists. Int J Radiat Oncol Biol Phys 81 (5): e775-80, 2011. [PUBMED Abstract]
Bahig H, Roberge D, Bosch W, et al.: Agreement among RTOG sarcoma radiation oncologists in contouring suspicious peritumoral edema for preoperative radiation therapy of soft tissue sarcoma of the extremity. Int J Radiat Oncol Biol Phys 86 (2): 298-303, 2013. [PUBMED Abstract]
Ferrari A: Role of chemotherapy in pediatric nonrhabdomyosarcoma soft-tissue sarcomas. Expert Rev Anticancer Ther 8 (6): 929-38, 2008. [PUBMED Abstract]
Adjuvant chemotherapy for localised resectable soft-tissue sarcoma of adults: meta-analysis of individual data. Sarcoma Meta-analysis Collaboration. Lancet 350 (9092): 1647-54, 1997. [PUBMED Abstract]
Woll PJ, Reichardt P, Le Cesne A, et al.: Adjuvant chemotherapy with doxorubicin, ifosfamide, and lenograstim for resected soft-tissue sarcoma (EORTC 62931): a multicentre randomised controlled trial. Lancet Oncol 13 (10): 1045-54, 2012. [PUBMED Abstract]
Demetri GD, Chawla SP, Ray-Coquard I, et al.: Results of an international randomized phase III trial of the mammalian target of rapamycin inhibitor ridaforolimus versus placebo to control metastatic sarcomas in patients after benefit from prior chemotherapy. J Clin Oncol 31 (19): 2485-92, 2013. [PUBMED Abstract]
van der Graaf WT, Blay JY, Chawla SP, et al.: Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 379 (9829): 1879-86, 2012. [PUBMED Abstract]
Mir O, Brodowicz T, Italiano A, et al.: Safety and efficacy of regorafenib in patients with advanced soft tissue sarcoma (REGOSARC): a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 17 (12): 1732-1742, 2016. [PUBMED Abstract]
Weiss AR, Chen YL, Scharschmidt TJ, et al.: Pathological response in children and adults with large unresected intermediate-grade or high-grade soft tissue sarcoma receiving preoperative chemoradiotherapy with or without pazopanib (ARST1321): a multicentre, randomised, open-label, phase 2 trial. Lancet Oncol 21 (8): 1110-1122, 2020. [PUBMED Abstract]
Kayton ML, Weiss AR, Xue W, et al.: Neoadjuvant pazopanib in nonrhabdomyosarcoma soft tissue sarcomas (ARST1321): A report of major wound complications from the Children’s Oncology Group and NRG Oncology. J Surg Oncol 127 (5): 871-881, 2023. [PUBMED Abstract]
Weiss AR, Chen YL, Scharschmidt TJ, et al.: Outcomes After Preoperative Chemoradiation With or Without Pazopanib in Non-Rhabdomyosarcoma Soft Tissue Sarcoma: A Report From Children’s Oncology Group and NRG Oncology. J Clin Oncol 41 (31): 4842-4848, 2023. [PUBMED Abstract]
Special Considerations for the Treatment of Children With Soft Tissue Sarcoma
Cancer in children and adolescents is rare, although the overall incidence has slowly increased since 1975.[1] Children and adolescents with cancer should be referred to medical centers that have a multidisciplinary team of cancer specialists with experience treating the cancers that occur during childhood and adolescence. This multidisciplinary team approach incorporates the skills of the following pediatric specialists and others to ensure that children receive treatment, supportive care, and rehabilitation to achieve optimal survival and quality of life:
Primary care physicians.
Pediatric surgeons.
Pathologists.
Pediatric radiation oncologists.
Pediatric medical oncologists and hematologists.
Rehabilitation specialists.
Pediatric oncology nurses.
Social workers.
Child-life professionals.
Psychologists.
Nutritionists.
For specific information about supportive care for children and adolescents with cancer, see the summaries on Supportive and Palliative Care.
The American Academy of Pediatrics has outlined guidelines for pediatric cancer centers and their role in the treatment of children and adolescents with cancer.[2] At these centers, clinical trials are available for most types of cancer that occur in children and adolescents, and the opportunity to participate is offered to most patients and their families. Multidisciplinary evaluation in pediatric cancer centers that have surgical and radiotherapeutic expertise is of critical importance to ensure the best clinical outcome for these patients. Although surgery with or without radiation therapy can be curative for a significant proportion of patients, the addition of chemotherapy might benefit subsets of children with the disease. Therefore, enrollment in clinical trials is encouraged. Clinical trials for children and adolescents with cancer are generally designed to compare potentially better therapy with current standard therapy. Other types of clinical trials test novel therapies when there is no standard therapy for a cancer diagnosis. Most of the progress in identifying curative therapies for childhood cancers has been achieved through clinical trials. Information about ongoing clinical trials is available from the NCI website.
Many therapeutic strategies for children and adolescents with soft tissue tumors are similar to those for adult patients, although there are important differences. For example, the biology of the neoplasm in pediatric patients may differ dramatically from that of the adult lesion. Additionally, limb-sparing procedures are more difficult to perform in pediatric patients. The morbidity associated with radiation therapy, particularly in infants and young children, may be much greater than that observed in adults.[3]
Improved outcomes with multimodality therapy in adults and children with soft tissue sarcomas over the past 20 years have caused increasing concern about the potential long-term side effects of this therapy in children. To maximize tumor control and minimize long-term morbidity, treatment must be individualized for children and adolescents with nonrhabdomyosarcomatous soft tissue sarcoma. These patients should be enrolled in prospective studies that accurately assess any potential complications.[4]
References
Smith MA, Altekruse SF, Adamson PC, et al.: Declining childhood and adolescent cancer mortality. Cancer 120 (16): 2497-506, 2014. [PUBMED Abstract]
American Academy of Pediatrics: Standards for pediatric cancer centers. Pediatrics 134 (2): 410-4, 2014. Also available online. Last accessed February 25, 2025.
Suit H, Spiro I: Radiation as a therapeutic modality in sarcomas of the soft tissue. Hematol Oncol Clin North Am 9 (4): 733-46, 1995. [PUBMED Abstract]
Hawkins DS, Black JO, Orbach D, et al.: Nonrhabdomyosarcoma soft-tissue sarcomas. In: Blaney SM, Helman LJ, Adamson PC, eds.: Pizzo and Poplack’s Pediatric Oncology. 8th ed. Wolters Kluwer, 2020, pp 721-46.
Treatment of Adipocytic Tumors
Adipocytic tumors account for less than 10% of soft tissue lesions in patients younger than 20 years. The most common adipocytic tumors in children are lipomas and lipoblastomas.
Table 11 summarizes the adipocytic neoplasms seen in pediatric patients and includes information about their corresponding clinico-pathological and molecular features.[1]
Table 11. Adipocytic Neoplasms in Pediatric Populationa
M = male; F = female; HGMA2 = high-mobility group AT-hook 2; PLAG1 = pleomorphic adenoma gene 1; MDM2 = mouse double minute 2 homolog; FUS = fused in sarcoma; DDIT3 = DNA damage inducible transcript 3.
aReprinted from Seminars in Diagnostic Pathology, Volume 36, Issue 2, Putra J, Al-Ibraheemi A, Adipocytic tumors in Children: A contemporary review, Pages 95–104, Copyright 2019, with permission from Elsevier.[1]
Benign
Lipoma
64%–70% (including variants)
• Solitary: M = F
Trunk.
Monotonous sheets of mature adipocytes.
Chromosomes 12q (HMGA2), 13q and 6p.
• Multiple: M > F
• Uncommon in the first 2 decades of life.
• Most common between age 40–60 years.
Angiolipoma
4%–28%
• M > F
Trunk and extremities.
• Mature adipocytic proliferation.
—
• Most common in late teens or early twenties.
• Vascular proliferation (capillary proliferation with fibrin thrombi).
Lipoblastoma
18%–30%
• M > F
Trunk and extremities.
• Lobular architecture.
Chromosome 8q (PLAG1) rearrangement.
• Zones of maturation.
• <3 years old (90%)
• Primitive stellate cells.
• Multivacuolated lipoblasts.
• Myxoid area with prominent plexiform vessels.
Hibernoma
2%
• M = F
Back (scapular area), chest wall, axilla and inguinal regions.
• Lobular architecture.
Chromosome 11q13-21 rearrangement.
• Rare in the first 2 decades of life (5%).
• Different types of cells: brown fat cells, multivacuolated lipoblasts, mature fat cells.
• 60% occur in the 3rd and 4th decades of life.
• Prominent capillary network (less pronounced than lipoblastoma and myxoid liposarcoma).
Supernumerary ring and giant marker chromosome 12q14-15 (MDM2).
• Extremely rare in children (associated with Li-Fraumeni syndrome).
• Significant variation in size.
• Peak incidence is 6th decade of life.
• Hyperchromatic nuclei with atypia.
Malignant
Myxoid liposarcoma
4%
• F > M
Extremities, trunk, head and neck and abdominal regions.
• Nodular architecture.
Recurrent t(12;16)(q13;p11) resulting in FUS::DDIT3 gene fusion.
• Mixture of round to spindle nonlipogenic cells and lipoblasts.
• The most common liposarcoma in children (2nd decade of life), but less frequent than adults.
• Prominent myxoid stroma with chicken-wire vasculature.
• Variants seen in children: pleomorphic and spindle cell subtypes.
• Peak incidence is 4th and 5th decades of life.
• Progression to round cell morphology is uncommon in children.
Dedifferentiated liposarcoma
Rare
• Reported in an 8-year old with a well-differentiated liposarcoma.[4]
• Lower extremity in a single case report of pediatric patient.[4]
• Transition from a well-differentiated liposarcoma to nonlipogenic, high-grade sarcoma.
Supernumerary ring and giant marker chromosome 12q14-15 (MDM2).
• Dedifferentiation occurs in up to 10% of well-differentiated liposarcomas in adults.
• Retroperitoneum (adults).
• Heterologous differentiation (5%–10%).
• Peak incidence is 6th decade of life.
Pleomorphic liposarcoma
Rare/not reported
• Peak incidence of pleomorphic liposarcoma is 7th decade of life.
• Extremities (adults).
• Pleomorphic lipoblasts.
—
• The subtype has been reported in the settings of Li-Fraumeni [5] and Muir-Torre syndromes.[6]
• Background of a high-grade, pleomorphic sarcoma (non-lipogenic).
Liposarcoma, Well-Differentiated, Not Otherwise Specified (NOS)
Liposarcoma is rare in the pediatric population and accounts for 3% of soft tissue sarcoma in patients younger than 20 years (see Table 1).
In a review of 182 pediatric patients with adult-type sarcomas, only 14 had a diagnosis of liposarcoma.[7] One retrospective study identified 34 patients younger than 22 years from 1960 to 2011.[8] There were roughly equal numbers of male and female patients, and the median age was 18 years. In an international clinico-pathological review, the characteristics of 82 cases of pediatric liposarcoma were reported.[9] The median age was 15.5 years, and females were more commonly affected. In both reports, most patients had myxoid liposarcoma.[8,9]
A literature review of 275 cases of pediatric liposarcoma showed that:[10]
Myxoid liposarcoma was the most common histology (68%), followed by well-differentiated liposarcoma (10.5%).
Twelve percent of patients died of disease, and most of the deaths occurred in patients with the pleomorphic and myxoid pleomorphic subtypes.
About 70% of patients with myxoid and well-differentiated liposarcoma were treated with surgery only. The overall clinical outcomes for these groups of patients were excellent, with no evidence of disease in 114 of 127 patients.
In contrast, more than 50% of patients with pleomorphic liposarcoma received radiation therapy and chemotherapy in addition to surgery, and their clinical outcome was suboptimal, with no evidence of disease in only 5 of 10 patients.
Germline TP53 pathogenic variants were seen in two patients with myxoid pleomorphic liposarcoma and two patients with well-differentiated liposarcoma who had a family history compatible with Li-Fraumeni syndrome.
Clinical presentation
Most liposarcomas in the pediatric and adolescent age range are low grade and located subcutaneously. Metastasis to lymph nodes is uncommon, and most metastases are pulmonary. Tumors arising in the periphery are more likely to be low grade and myxoid. Tumors arising centrally are more likely to be high grade, pleomorphic, and present with metastasis or recur with metastasis.
Histopathological classification
The World Health Organization (WHO) classification for liposarcoma is as follows:[11]
Intermediate (locally aggressive).
Atypical lipomatous tumor. These tumors do not metastasize unless they undergo dedifferentiation.
Malignant.
Dedifferentiated liposarcoma.
Myxoid liposarcoma. Pure myxoid liposarcomas are characterized by a t(12;16)(q13;p11) translocation and can metastasize, but patients usually have an excellent outcome when they do not have a round cell component.[12] Myxoid liposarcoma is the most common subtype of liposarcoma in the pediatric population.[8,9]
Pleomorphic liposarcoma. This is an uncommon type of liposarcoma and primarily arises in older adults.
Myxoid pleomorphic liposarcoma. This rare entity occurs primarily in children, adolescents, and young adults. It commonly presents in the mediastinum and is clinically aggressive.
Liposarcoma, well-differentiated, NOS.
Genomic alterations
Atypical lipomatous tumor. This entity is characterized by supernumerary ring and giant marker chromosomes that contain chromosomal region 12q14-q15, which includes MDM2. MDM2 amplification can be detected in virtually all cases of atypical lipomatous tumor/well-differentiated liposarcoma, with nearby genes such as CDK4 and FRS2 commonly being coamplified with MDM2.[13]
Dedifferentiated liposarcoma. This entity, like atypical lipomatous tumor, is characterized by MDM2 amplification and the supernumerary ring and giant marker chromosomes containing the chromosomal region 12q14-q15. Dedifferentiated liposarcoma contains a high number of segmental copy number alterations, but has few gene variants.[14]
Myxoid liposarcoma. This entity is characterized by the t(12;16)(q13;p11) translocation that produces the FUS::DDIT3 gene fusion.[14] In a small percentage of cases, EWSR1 substitutes for FUS, producing the EWSR1::DDIT3 gene fusion (t(12;22)(q13;q12)). DDIT3 (previously called CHOP and GADD153) is a stress-induced gene that has an inhibitory effect on adipogenesis.[15] Myxoid liposarcoma is the most common subtype of liposarcoma in the pediatric population. Most pediatric cases show the FUS::DDIT3 gene fusion.[8,9,16]
Pleomorphic liposarcoma. This entity is primarily a disease of older adults and lacks either DDIT3 gene rearrangements or MDM2 amplification. Cases of pleomorphic liposarcoma typically have multiple chromosomal imbalances, including variants in TP53 and NF1 observed in some cases.[17]
Myxoid pleomorphic liposarcoma. This entity most commonly presents in the adolescent and young adult population and lacks the DDIT3 gene rearrangement of myxoid liposarcoma and the MDM2 amplification of atypical lipomatous tumor and dedifferentiated liposarcoma.[9,16,18] Instead, myxoid pleomorphic liposarcoma presents with multiple chromosomal gains and losses. Loss of Rb expression is commonly observed, sometimes in association with loss of chromosome 13q14 where RB1 is located.[18,19] Although most cases of myxoid pleomorphic liposarcoma lack TP53 variants, a minority have TP53 variants that are associated with Li-Fraumeni syndrome in some cases.[20–22]
Prognosis
Higher grade or central tumors are associated with a significantly higher risk of death. In an international retrospective review, the 5-year survival rate was 42% for patients with central tumors. Seven of ten patients with pleomorphic myxoid liposarcoma died of their disease.[9] In a retrospective study of 14 patients, the 5-year survival rate was 78%. Tumor grade, histological subtype, and primary location correlated with survival.[8]
Treatment of liposarcoma
Treatment options for liposarcoma include the following:
Surgery. If the tumor is not completely removed or locally recurs, a second surgery may be performed.[23–25]
Surgery is the most important treatment for liposarcoma. After complete surgical resection of well-differentiated or myxoid liposarcoma, the event-free survival (EFS) and overall survival (OS) rates are roughly 90%.[29] If initial surgery is incomplete, re-excision should be performed to achieve a wide margin of resection.[23–25] Local recurrences have been seen and are controlled with a second resection of the tumor, particularly for low-grade liposarcomas.
Chemotherapy
Chemotherapy has been used to decrease the size of liposarcoma before surgery to facilitate complete resection, particularly in central tumors.[30,31] The role of postoperative chemotherapy for liposarcoma is poorly defined. Postoperative therapy for completely resected myxoid liposarcomas does not appear to be needed. Even with the use of postoperative chemotherapy, the survival of patients with pleomorphic liposarcomas remains poor.[32]
There are limited data to support the use of trabectedin in pediatric patients.[33] Trabectedin has produced encouraging responses in adults with advanced myxoid liposarcoma.[34] In one study, adult patients with recurrent liposarcoma and leiomyosarcoma were randomly assigned to treatment with either trabectedin or dacarbazine. Patients treated with trabectedin had a 45% reduction in disease progression.[35][Level of evidence B1]
Treatment with eribulin, a nontaxane microtubule dynamics inhibitor, significantly improved survival in adult patients with recurrent liposarcoma compared with dacarbazine. The median OS was 15.6 months for patients who received eribulin, versus 8.4 months for patients who received dacarbazine. Survival differences were more pronounced in patients with dedifferentiated and pleomorphic liposarcoma. Eribulin was effective in prolonging survival of patients with either high-grade or intermediate-grade tumors.[36][Level of evidence A1] A pediatric phase I trial of eribulin did not accrue any patients with liposarcoma.[37]
Surgery and radiation therapy
Radiation therapy is also considered either preoperatively or postoperatively, depending on the cosmetic/functional consequences of additional surgery and radiation therapy.[38,39]
Targeted therapy
In a phase II, single-arm, multicenter study, 41 adult patients with unresectable or metastatic high-grade or intermediate-grade liposarcoma were treated with pazopanib at a dose of 800 mg daily.[28][Level of evidence B4]
The progression-free survival (PFS) rate at 12 weeks was 68.3%, which was significantly greater than the null hypothesis value of 40%.
Forty-four percent of patients experienced tumor control. One patient had a partial response, and 17 patients had stable disease.
At 24 weeks, 39% of the patients remained progression free. The median PFS was 4.4 months, and median OS was 12.6 months.
References
Putra J, Al-Ibraheemi A: Adipocytic tumors in Children: A contemporary review. Semin Diagn Pathol 36 (2): 95-104, 2019. [PUBMED Abstract]
Coffin CM, Alaggio R: Adipose and myxoid tumors of childhood and adolescence. Pediatr Dev Pathol 15 (1 Suppl): 239-54, 2012. [PUBMED Abstract]
Dehner LP, Gru AA: Nonepithelial Tumors and Tumor-like Lesions of the Skin and Subcutis in Children. Pediatr Dev Pathol 21 (2): 150-207, 2018 Mar-Apr. [PUBMED Abstract]
Yozu M, Symmans P, Dray M, et al.: Muir-Torre syndrome-associated pleomorphic liposarcoma arising in a previous radiation field. Virchows Arch 462 (3): 355-60, 2013. [PUBMED Abstract]
Palit A, Inamadar AC: Circumferential skin folds in a child: a case of Michelin tire baby syndrome. Indian J Dermatol Venereol Leprol 73 (1): 49-51, 2007 Jan-Feb. [PUBMED Abstract]
Goucha S, Khaled A, Zéglaoui F, et al.: Nevus lipomatosus cutaneous superficialis: Report of eight cases. Dermatol Ther (Heidelb) 1 (2): 25-30, 2011. [PUBMED Abstract]
Ferrari A, Casanova M, Collini P, et al.: Adult-type soft tissue sarcomas in pediatric-age patients: experience at the Istituto Nazionale Tumori in Milan. J Clin Oncol 23 (18): 4021-30, 2005. [PUBMED Abstract]
Stanelle EJ, Christison-Lagay ER, Sidebotham EL, et al.: Prognostic factors and survival in pediatric and adolescent liposarcoma. Sarcoma 2012: 870910, 2012. [PUBMED Abstract]
Alaggio R, Coffin CM, Weiss SW, et al.: Liposarcomas in young patients: a study of 82 cases occurring in patients younger than 22 years of age. Am J Surg Pathol 33 (5): 645-58, 2009. [PUBMED Abstract]
Baday YI, Navai SA, Hicks MJ, et al.: Pediatric liposarcoma: A case series and literature review. Pediatr Blood Cancer 68 (12): e29327, 2021. [PUBMED Abstract]
Fletcher CDM, Bridge JA, Hogendoorn P, et al., eds.: WHO Classification of Tumours of Soft Tissue and Bone. 4th ed. IARC Press, 2013.
Sreekantaiah C, Karakousis CP, Leong SP, et al.: Cytogenetic findings in liposarcoma correlate with histopathologic subtypes. Cancer 69 (10): 2484-95, 1992. [PUBMED Abstract]
Kanojia D, Nagata Y, Garg M, et al.: Genomic landscape of liposarcoma. Oncotarget 6 (40): 42429-44, 2015. [PUBMED Abstract]
Powers MP, Wang WL, Hernandez VS, et al.: Detection of myxoid liposarcoma-associated FUS-DDIT3 rearrangement variants including a newly identified breakpoint using an optimized RT-PCR assay. Mod Pathol 23 (10): 1307-15, 2010. [PUBMED Abstract]
Han J, Murthy R, Wood B, et al.: ER stress signalling through eIF2α and CHOP, but not IRE1α, attenuates adipogenesis in mice. Diabetologia 56 (4): 911-24, 2013. [PUBMED Abstract]
Peng R, Li N, Lan T, et al.: Liposarcoma in children and young adults: a clinicopathologic and molecular study of 23 cases in one of the largest institutions of China. Virchows Arch 479 (3): 537-549, 2021. [PUBMED Abstract]
Barretina J, Taylor BS, Banerji S, et al.: Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet 42 (8): 715-21, 2010. [PUBMED Abstract]
Creytens D, Folpe AL, Koelsche C, et al.: Myxoid pleomorphic liposarcoma-a clinicopathologic, immunohistochemical, molecular genetic and epigenetic study of 12 cases, suggesting a possible relationship with conventional pleomorphic liposarcoma. Mod Pathol 34 (11): 2043-2049, 2021. [PUBMED Abstract]
Hofvander J, Jo VY, Ghanei I, et al.: Comprehensive genetic analysis of a paediatric pleomorphic myxoid liposarcoma reveals near-haploidization and loss of the RB1 gene. Histopathology 69 (1): 141-147, 2016. [PUBMED Abstract]
Sinclair TJ, Thorson CM, Alvarez E, et al.: Pleomorphic myxoid liposarcoma in an adolescent with Li-Fraumeni syndrome. Pediatr Surg Int 33 (5): 631-635, 2017. [PUBMED Abstract]
Francom CR, Leoniak SM, Lovell MA, et al.: Head and neck pleomorphic myxoid liposarcoma in a child with Li-Fraumeni syndrome. Int J Pediatr Otorhinolaryngol 123: 191-194, 2019. [PUBMED Abstract]
Zare SY, Leivo M, Fadare O: Recurrent Pleomorphic Myxoid Liposarcoma in a Patient With Li-Fraumeni Syndrome. Int J Surg Pathol 28 (2): 225-228, 2020. [PUBMED Abstract]
Sugiura H, Takahashi M, Katagiri H, et al.: Additional wide resection of malignant soft tissue tumors. Clin Orthop (394): 201-10, 2002. [PUBMED Abstract]
Cecchetto G, Guglielmi M, Inserra A, et al.: Primary re-excision: the Italian experience in patients with localized soft-tissue sarcomas. Pediatr Surg Int 17 (7): 532-4, 2001. [PUBMED Abstract]
Chui CH, Spunt SL, Liu T, et al.: Is reexcision in pediatric nonrhabdomyosarcoma soft tissue sarcoma necessary after an initial unplanned resection? J Pediatr Surg 37 (10): 1424-9, 2002. [PUBMED Abstract]
Bahig H, Roberge D, Bosch W, et al.: Agreement among RTOG sarcoma radiation oncologists in contouring suspicious peritumoral edema for preoperative radiation therapy of soft tissue sarcoma of the extremity. Int J Radiat Oncol Biol Phys 86 (2): 298-303, 2013. [PUBMED Abstract]
Baldini EH, Wang D, Haas RL, et al.: Treatment Guidelines for Preoperative Radiation Therapy for Retroperitoneal Sarcoma: Preliminary Consensus of an International Expert Panel. Int J Radiat Oncol Biol Phys 92 (3): 602-12, 2015. [PUBMED Abstract]
Samuels BL, Chawla SP, Somaiah N, et al.: Results of a prospective phase 2 study of pazopanib in patients with advanced intermediate-grade or high-grade liposarcoma. Cancer 123 (23): 4640-4647, 2017. [PUBMED Abstract]
La Quaglia MP, Spiro SA, Ghavimi F, et al.: Liposarcoma in patients younger than or equal to 22 years of age. Cancer 72 (10): 3114-9, 1993. [PUBMED Abstract]
Ferrari A, Casanova M, Spreafico F, et al.: Childhood liposarcoma: a single-institutional twenty-year experience. Pediatr Hematol Oncol 16 (5): 415-21, 1999 Sep-Oct. [PUBMED Abstract]
Cecchetto G, Alaggio R, Dall’Igna P, et al.: Localized unresectable non-rhabdo soft tissue sarcomas of the extremities in pediatric age: results from the Italian studies. Cancer 104 (9): 2006-12, 2005. [PUBMED Abstract]
Huh WW, Yuen C, Munsell M, et al.: Liposarcoma in children and young adults: a multi-institutional experience. Pediatr Blood Cancer 57 (7): 1142-6, 2011. [PUBMED Abstract]
Baruchel S, Pappo A, Krailo M, et al.: A phase 2 trial of trabectedin in children with recurrent rhabdomyosarcoma, Ewing sarcoma and non-rhabdomyosarcoma soft tissue sarcomas: a report from the Children’s Oncology Group. Eur J Cancer 48 (4): 579-85, 2012. [PUBMED Abstract]
Gronchi A, Bui BN, Bonvalot S, et al.: Phase II clinical trial of neoadjuvant trabectedin in patients with advanced localized myxoid liposarcoma. Ann Oncol 23 (3): 771-6, 2012. [PUBMED Abstract]
Demetri GD, von Mehren M, Jones RL, et al.: Efficacy and Safety of Trabectedin or Dacarbazine for Metastatic Liposarcoma or Leiomyosarcoma After Failure of Conventional Chemotherapy: Results of a Phase III Randomized Multicenter Clinical Trial. J Clin Oncol 34 (8): 786-93, 2016. [PUBMED Abstract]
Demetri GD, Schöffski P, Grignani G, et al.: Activity of Eribulin in Patients With Advanced Liposarcoma Demonstrated in a Subgroup Analysis From a Randomized Phase III Study of Eribulin Versus Dacarbazine. J Clin Oncol 35 (30): 3433-3439, 2017. [PUBMED Abstract]
Schafer ES, Rau RE, Berg S, et al.: A phase 1 study of eribulin mesylate (E7389), a novel microtubule-targeting chemotherapeutic agent, in children with refractory or recurrent solid tumors: A Children’s Oncology Group Phase 1 Consortium study (ADVL1314). Pediatr Blood Cancer 65 (8): e27066, 2018. [PUBMED Abstract]
Lee ATJ, Thway K, Huang PH, et al.: Clinical and Molecular Spectrum of Liposarcoma. J Clin Oncol 36 (2): 151-159, 2018. [PUBMED Abstract]
Beane JD, Yang JC, White D, et al.: Efficacy of adjuvant radiation therapy in the treatment of soft tissue sarcoma of the extremity: 20-year follow-up of a randomized prospective trial. Ann Surg Oncol 21 (8): 2484-9, 2014. [PUBMED Abstract]
Treatment of Chondro-osseous Tumors
Chondro-osseous tumors have several subtypes, including the following:
Osseous and chondromatous neoplasms account for 0.8% of soft tissue sarcomas in patients younger than 20 years (see Table 1). Mesenchymal chondrosarcoma is more common in the head and neck region.
Histopathological features and genomic alterations
Mesenchymal chondrosarcoma is a rare tumor characterized by small round cells and hyaline cartilage, and it more commonly affects young adults.
Mesenchymal chondrosarcoma has been associated with a consistent chromosomal rearrangement. A retrospective analysis of cases of mesenchymal chondrosarcoma identified a HEY1::NCOA2 gene fusion in 10 of 15 tested specimens.[1] This gene fusion was not associated with chromosomal changes that could be detected by karyotyping. In one instance, translocation t(1;5)(q42;q32) was identified in a case of mesenchymal chondrosarcoma and shown to be associated with a novel IRF2BP::CDX1 gene fusion.[2]
A retrospective study analyzed 13 patients with mesenchymal chondrosarcoma, all with confirmed HEY1::NCOA2 gene fusions.[3]
The median age of presentation was 19 years.
Five patients with mesenchymal chondrosarcomas (39%) had an intraosseous presentation (skull, maxilla, palate, and mandible), while the remaining eight cases occurred in the brain/meninges, orbit, and nasal cavity.
Microscopically, head and neck mesenchymal chondrosarcomas were characterized by primitive round cells arranged in a distinctive nested architecture and a rich staghorn vasculature.
A cartilaginous component of hyaline cartilage islands and/or single chondrocytes were present in 69% of cases.
A combined immunoprofile of CD99(+)/SATB2(+)/CD34(-)/STAT6(-) was typically noted.
Prognostic factors and prognosis
A retrospective survey of European institutions identified 113 children and adults with mesenchymal chondrosarcoma. Factors associated with better outcomes included the following:[4][Level of evidence C1]
Lack of metastatic disease at initial presentation.
Clear resection margins.
Administration of postoperative chemotherapy after resection for patients with initially localized disease.
A retrospective analysis of Surveillance, Epidemiology, and End Results (SEER) Program data from 1973 to 2011 identified 205 patients with mesenchymal chondrosarcoma; 82 patients had skeletal primary tumors, and 123 patients had extraskeletal tumors.[5] The outcomes of patients with skeletal and extraskeletal primary tumors were the same. Factors associated with outcomes included the following:
Primary site: The 5-year overall survival (OS) rate was 50% for patients with appendicular tumors, 37% for patients with axial tumors, and 74% for patients with cranial tumors.
Metastases and tumor size: Presence of metastatic disease and larger tumor size were independently associated with an increased risk of death.
A single-institution retrospective review identified 43 cases of mesenchymal chondrosarcoma from 1979 to 2010.[6] Thirty patients with localized disease were evaluated. The mean age at diagnosis was 33 years (range, 11–65 years).
The 5-year OS rate was 51%, and the 10-year OS rate was 37%.
Younger age (<30 years) and male sex were associated with poorer OS and disease-free survival (DFS).
Patients who did not receive adjuvant radiation therapy were more likely to have a local recurrence.
Treatment of extraskeletal mesenchymal chondrosarcoma
Treatment options for extraskeletal mesenchymal chondrosarcoma include the following:
Surgery.
Surgery preceded or followed by radiation therapy.[7,8]
Chemotherapy followed by surgery and additional chemotherapy. Radiation therapy may also be given.
A review of 15 patients younger than 26 years included 11 patients with soft tissue lesions from the German Cooperative Soft Tissue Sarcoma Study Group and 4 patients with primary bone lesions from the German-Austrian-Swiss Cooperative Osteosarcoma Study Group protocols. The review suggested that complete surgical removal, or incomplete resection followed by radiation therapy, was necessary for local control.[9][Level of evidence C1]
A single-institution, retrospective review identified 12 pediatric patients with mesenchymal chondrosarcoma.[10] Eleven patients presented with localized disease, and one patient presented with pulmonary nodules. Six patients received preoperative chemotherapy. All patients received postoperative chemotherapy (most commonly ifosfamide/doxorubicin) with or without radiation therapy (median dose, 59.4 Gy).
The NCOA2 rearrangement was documented in these patients’ tumors.
The study confirmed that surgical resection is necessary for cure.
At a median follow-up of 4.8 years, the 5-year DFS rate was 68.2% (95% confidence interval [CI], 39.8%–96.6%), and the OS rate was 88.9% (95% CI, 66.9%–100%).
A Japanese study of patients with extraskeletal myxoid chondrosarcoma and mesenchymal chondrosarcoma randomly assigned patients to treatment with either trabectedin or best supportive care.[11] The median age of patients was 38 years (range, 21–77 years).
The OS was higher for the patients assigned to receive trabectedin than for patients assigned to receive best supportive care.
Osteosarcoma, Extraskeletal
Extraskeletal osteosarcoma is extremely rare in the pediatric and adolescent population. Osseous and chondromatous neoplasms account for 0.8% of soft tissue sarcomas in patients younger than 20 years (see Table 1).
Genomic alterations
A review of 32 adult patients with extraskeletal osteosarcomas consistently revealed several alterations.[12] Frequent genomic alterations included copy number losses in CDKN2A (70%), TP53 (56%), and RB1 (49%). Variants were identified that affected methylation/demethylation (40%), chromatin remodeling (27%), and the WNT/SHH pathways (27%). Cases with simultaneous TP53 and RB1 biallelic copy number losses were associated with worse DFS and OS.
Prognostic factors and prognosis
Extraskeletal osteosarcoma is associated with a high risk of local recurrence and pulmonary metastasis.[13]
A single-institution retrospective review identified 43 patients with extraskeletal osteosarcoma; 37 patients had localized disease, and 6 patients presented with metastatic disease. The median age was 55 years (range, 7–81 years). Seventy-five percent of patients received chemotherapy.[14]
The median progression-free survival (PFS) was 21 months.
The median OS was 50 months.
There was a trend toward better survival for patients who received chemotherapy, and a statistically significant improvement in survival for patients who received chemotherapy that included cisplatin.
In a review of 274 patients with extraskeletal osteosarcoma, the median age at diagnosis was 57 years (range, 12–91 years).[15][Level of evidence C1]
The 5-year DFS and OS rates were significantly better for those who received chemotherapy.
The use of an osteosarcoma-type regimen was associated with improved response rates.
The European Musculoskeletal Oncology Society performed a retrospective analysis of 266 eligible patients with extraskeletal osteosarcoma treated between 1981 and 2014. Fifty patients (19%) presented with metastatic disease.[15]
An analysis of the 211 patients who achieved complete remission after surgical resection of the primary tumor showed a 5-year OS rate of 51% and a DFS rate of 43%.
There was a favorable trend for survival among patients who were treated with chemotherapy that is usually employed for patients with osseous osteosarcoma.
In a multivariable analysis, factors associated with better prognosis included younger age (<40 years), smaller tumors, and use of chemotherapy.
An analysis of SEER Program data from 1973 to 2009 identified 256 patients (6%) with extraskeletal osteosarcoma among 4,173 patients with high-grade osteosarcoma.[16]
Compared with skeletal osteosarcoma, patients with extraskeletal osteosarcoma were more likely to be older, female, have an axial primary tumor, and have regional lymph node involvement.
Adverse prognostic features included presence of metastatic disease, larger tumor size, older age, and axial primary tumor site.
Treatment of extraskeletal osteosarcoma
Treatment options for extraskeletal osteosarcoma include the following:
Wang L, Motoi T, Khanin R, et al.: Identification of a novel, recurrent HEY1-NCOA2 fusion in mesenchymal chondrosarcoma based on a genome-wide screen of exon-level expression data. Genes Chromosomes Cancer 51 (2): 127-39, 2012. [PUBMED Abstract]
Nyquist KB, Panagopoulos I, Thorsen J, et al.: Whole-transcriptome sequencing identifies novel IRF2BP2-CDX1 fusion gene brought about by translocation t(1;5)(q42;q32) in mesenchymal chondrosarcoma. PLoS One 7 (11): e49705, 2012. [PUBMED Abstract]
Xu B, Rooper LM, Dermawan JK, et al.: Mesenchymal chondrosarcoma of the head and neck with HEY1::NCOA2 fusion: A clinicopathologic and molecular study of 13 cases with emphasis on diagnostic pitfalls. Genes Chromosomes Cancer 61 (11): 670-677, 2022. [PUBMED Abstract]
Frezza AM, Cesari M, Baumhoer D, et al.: Mesenchymal chondrosarcoma: prognostic factors and outcome in 113 patients. A European Musculoskeletal Oncology Society study. Eur J Cancer 51 (3): 374-81, 2015. [PUBMED Abstract]
Schneiderman BA, Kliethermes SA, Nystrom LM: Survival in Mesenchymal Chondrosarcoma Varies Based on Age and Tumor Location: A Survival Analysis of the SEER Database. Clin Orthop Relat Res 475 (3): 799-805, 2017. [PUBMED Abstract]
Kawaguchi S, Weiss I, Lin PP, et al.: Radiation therapy is associated with fewer recurrences in mesenchymal chondrosarcoma. Clin Orthop Relat Res 472 (3): 856-64, 2014. [PUBMED Abstract]
Bahig H, Roberge D, Bosch W, et al.: Agreement among RTOG sarcoma radiation oncologists in contouring suspicious peritumoral edema for preoperative radiation therapy of soft tissue sarcoma of the extremity. Int J Radiat Oncol Biol Phys 86 (2): 298-303, 2013. [PUBMED Abstract]
Baldini EH, Wang D, Haas RL, et al.: Treatment Guidelines for Preoperative Radiation Therapy for Retroperitoneal Sarcoma: Preliminary Consensus of an International Expert Panel. Int J Radiat Oncol Biol Phys 92 (3): 602-12, 2015. [PUBMED Abstract]
Dantonello TM, Int-Veen C, Leuschner I, et al.: Mesenchymal chondrosarcoma of soft tissues and bone in children, adolescents, and young adults: experiences of the CWS and COSS study groups. Cancer 112 (11): 2424-31, 2008. [PUBMED Abstract]
Bishop MW, Somerville JM, Bahrami A, et al.: Mesenchymal Chondrosarcoma in Children and Young Adults: A Single Institution Retrospective Review. Sarcoma 2015: 608279, 2015. [PUBMED Abstract]
Morioka H, Takahashi S, Araki N, et al.: Results of sub-analysis of a phase 2 study on trabectedin treatment for extraskeletal myxoid chondrosarcoma and mesenchymal chondrosarcoma. BMC Cancer 16: 479, 2016. [PUBMED Abstract]
Jour G, Wang L, Middha S, et al.: The molecular landscape of extraskeletal osteosarcoma: A clinicopathological and molecular biomarker study. J Pathol Clin Res 2 (1): 9-20, 2016. [PUBMED Abstract]
Sordillo PP, Hajdu SI, Magill GB, et al.: Extraosseous osteogenic sarcoma. A review of 48 patients. Cancer 51 (4): 727-34, 1983. [PUBMED Abstract]
Paludo J, Fritchie K, Haddox CL, et al.: Extraskeletal Osteosarcoma: Outcomes and the Role of Chemotherapy. Am J Clin Oncol 41 (9): 832-837, 2018. [PUBMED Abstract]
Longhi A, Bielack SS, Grimer R, et al.: Extraskeletal osteosarcoma: A European Musculoskeletal Oncology Society study on 266 patients. Eur J Cancer 74: 9-16, 2017. [PUBMED Abstract]
Thampi S, Matthay KK, Boscardin WJ, et al.: Clinical Features and Outcomes Differ between Skeletal and Extraskeletal Osteosarcoma. Sarcoma 2014: 902620, 2014. [PUBMED Abstract]
Treatment of Fibroblastic and Myofibroblastic Tumors
Fibroblastic and myofibroblastic tumors have several subtypes, including the following:
Desmoid-type fibromatosis has previously been called desmoid tumor or aggressive fibromatosis.
Desmoid-type fibromatosis has an extremely low potential to metastasize. The tumors are locally infiltrating, and surgical control can be challenging because of difficulty obtaining margins of resection that contain the entire infiltrating tumor.
Desmoid-type fibromatosis has a high potential for local recurrence. These tumors also have a highly variable natural history, including well documented examples of spontaneous regression.[1,2]
Genomic alterations
Most desmoid tumors are sporadic, but a small proportion may occur in association with a variant in the APC gene (associated with intestinal polyps and a high incidence of colon cancer). In a study of 519 patients older than 10 years with a diagnosis of desmoid-type fibromatosis, 39 patients (7.5%, a possible underestimation) were found to have familial adenomatous polyposis (FAP).[3] The patients with FAP and desmoid-type fibromatosis were younger, more often male, and had more abdominal wall or mesenteric tumors than did patients with desmoid-type fibromatosis without FAP.
Variants in exon 3 of the CTNNB1 gene are seen in more than 80% of desmoid-type fibromatosis cases. The 45F variant in exon 3 of the CTNNB1 gene has been associated with an increased risk of disease recurrence.[4]
Currently, there are no general recommendations for genetic testing in children with desmoid-type fibromatosis. Pathological and molecular characteristics of the tumor only provide guidance for screening.
A patient should be referred to a genetic counselor if there is a family history of colon cancer, congenital hyperplasia of the retinal pigment epithelium is present,[5,6] or the desmoid-type fibromatosis occurs in the abdomen or abdominal wall.[3] If the tumor has a somatic CTNNB1 variant, screening is not necessary, because the APC gene variant has not been described in this setting. If a CTNNB1 variant is not identified, screening for the APC variant may be warranted.[7,8]
Pediatric desmoid tumors can harbor additional variants in the AKT, BRAF V600E, TP53, and RET genes.[9] For more information, see the Familial Adenomatous Polyposis (FAP) section in Genetics of Colorectal Cancer.
Treatment of desmoid-type fibromatosis
Evaluating the benefit of treatment interventions for desmoid-type fibromatosis has been extremely difficult, because desmoid-type fibromatosis has a highly variable natural history, with partial regressions seen in up to 20% of patients.[2] Large adult series and smaller pediatric series have reported long periods of disease stabilization and even regression without systemic therapy.[10,11]; [12][Level of evidence C2] For instance, in a large placebo-controlled trial of sorafenib in adult patients with desmoid tumors, the patients who received no therapy (observation/placebo) demonstrated a 20% partial regression rate, and 46% of the patients in the placebo group had no progression at 1 year.[2]
Treatment options for desmoid-type fibromatosis include the following:
Because of the variable natural history of desmoid tumors, as outlined above, observation is sometimes a viable option. This is particularly the case for lesions that are asymptomatic, do not pose a danger to vital organs, or are incompletely resected.[11,13–19]
A global consensus meeting that involved sarcoma experts with experience in both adult and pediatric desmoid tumor was organized to define the appropriate management of these tumors. The Desmoid Tumor Working Group suggested that an initial active surveillance approach does not influence the efficacy of subsequent treatments. They suggested that active therapy should only be considered in cases of persistent progression or symptoms. Active surveillance includes continuous monitoring with a first magnetic resonance imaging within 1 to 2 months of diagnosis, followed by scans in 3- to 6-month intervals. When the disease is located in critical structures that may pose significant morbidity, such as the mesentery and head and neck region, earlier treatment decisions should be made.[20]
Evidence (observation vs. initial surgery):
A nonrandomized prospective cohort study included 771 patients with desmoid-type fibromatosis who were referred for second opinion and molecular analysis from 22 referral centers in France. The study compared initial surgery with initial observation.[21][Level of evidence C2]
There was no difference in event-free survival (EFS) rates between the two groups (53% vs. 58%; P = .415).
Among patients with favorable tumor locations (defined as abdominal wall, intra-abdominal, breast, digestive viscera, and lower limb), the 2-year EFS rate was similar in patients who underwent surgery (70%) or were observed (63%; P = .41).
Among patients with tumors in unfavorable locations (defined as chest wall, head and neck, and upper limb), the 2-year EFS rate was significantly better for those treated nonsurgically (52%) compared with those who underwent initial surgery (25%; P = .001).
There were 173 patients with desmoid-type fibromatosis who were treated on European paediatric Soft Tissue Sarcoma Study Group (EpSSG) studies since 2005. Thirteen patients (8%) had biopsies only (no further treatment), 65 patients (42%) received chemotherapy only, 31 patients (20%) underwent surgery only, 36 patients (23%) had both chemotherapy and surgery, and 9 patients (6%) received radiation therapy in addition to other therapies.[22][Level of evidence C2]
All patients were alive at the time of analysis.
The authors concluded that the conservative nonsurgical approach did not compromise outcome in pediatric patients.
Chemotherapy, for unresectable or recurrent tumors
The following chemotherapy regimens have been used to treat desmoid-type fibromatosis:
Methotrexate and vinblastine: This combination produced objective responses in about one-third of patients with unresectable or recurrent desmoid-type fibromatosis.[23]
Doxorubicin and dacarbazine: A series of mainly adult patients with FAP and unresectable desmoid-type fibromatosis that were unresponsive to hormone therapy showed that doxorubicin plus dacarbazine followed by meloxicam (an NSAID) can be safely administered and can induce responses.[24]
Pegylated liposomal doxorubicin: In a study of 11 patients, 4 patients achieved an objective response and 7 patients had stable disease.[25] In a series of five patients, a median progression-free interval of 29 months was reported.[26]
Hydroxyurea: A retrospective analysis reported the results of 16 children with previously treated desmoid tumors who were treated with hydroxyurea. Before hydroxyurea, seven patients had tumor progression, two patients had increased pain, and seven patients had both. Tumor shrinkage occurred in 37.5% of patients (with 18.7% partial remissions), and symptom improvement occurred in 68.7% of patients.[27]
Vinorelbine: A retrospective review of 24 patients with desmoid-type fibromatosis (median age, 13.9 years; range, 1–23 years) received oral vinorelbine at eight centers of the Société Française des Cancers de l’Enfant between 2005 and 2020. For the 23 evaluable patients, 13% had partial responses, 78% had disease stabilization, and 9% had disease progression. The progression-free survival (PFS) rate was 89.3% at 24 months.[28]
Tyrosine kinase inhibitors
Targeted therapy has been used to treat children and adults with desmoid-type fibromatosis.
Evidence (sorafenib):
An international, prospective, phase III, double-blind study was conducted through the National Clinical Trials Network to evaluate the efficacy of sorafenib in patients with unresectable progressive or symptomatic desmoid tumors. Eighty-seven patients were enrolled (aged 18–72 years). Patients were randomly assigned in a 2:1 fashion to receive either sorafenib or placebo. Crossover to sorafenib was permitted after disease progression.[2][Level of evidence B1]
The objective response rate was 33% (95% confidence interval [CI], 20%–48%) in the sorafenib arm and 20% (95% CI, 8%–38%) in the placebo arm.
The median time to objective response was 9.5 months for patients treated with sorafenib and 13.3 months for patients who received the placebo.
The 2-year PFS rate was 81% for patients treated with sorafenib, compared with 36% for patients who received the placebo.
Evidence (pazopanib):
One small series included six patients (aged 3–21 years) with desmoid-type fibromatosis who were treated with pazopanib.[29]
Symptomatic improvement and stable disease were reported in all patients.
A randomized noncomparative study included adult patients with desmoid tumors who were treated with either pazopanib or methotrexate/vinblastine.[30]
About 84% of the patients who received pazopanib had no progression at 6 months.
NOTCH pathway/gamma-secretase inhibitors
The NOTCH pathway has been implicated in the development of desmoid tumors.[31] The NOTCH pathway/gamma-secretase inhibitor nirogacestat has been evaluated in adult and pediatric patients.
Evidence (nirogacestat):
One study included 17 adult patients with desmoid tumors, 15 of whom had variants in the APC or CTNNB1 genes.[32][Level of evidence C3]
Five patients (29%) achieved a confirmed partial response to nirogacestat.
Four adult patients experienced grade 1 irregular menstruation.
In a clinical trial (NCT03785964), 75% of women of childbearing potential reported events related to ovarian dysfunction.[33]
A small series included four patients younger than 20 years who received nirogacestat on a compassionate basis.[34][Level of evidence C3]
Three patients had a durable benefit, defined as a complete response (n = 1), partial response (n = 1), or stable disease (n = 1).
No patients experienced grades 3 or 4 adverse events.
The U.S. Food and Drug Administration (FDA) approved nirogacestat for the treatment of patients aged 18 years and older with progressive desmoid tumors who require systemic therapy. The approval was based on a prospective, randomized, placebo-controlled trial conducted by a consortium.[33]
In 142 patients, nirogacestat had a significant PFS benefit over placebo (hazard ratio for disease progression or death, 0.29; 95% CI, 0.15–0.55; P < .001).
The likelihood of being event free at 2 years was 76% with nirogacestat and 44% with placebo.
Nirogacestat was associated with significant benefits related to PFS, objective response, pain, symptom burden, physical functioning, role functioning, and health-related quality of life in adults with progressing desmoid tumors.
Nirogacestat was associated with a significant risk of ovarian failure.
NSAIDs
NSAIDs such as sulindac have been used in single cases for desmoid-type fibromatosis. The responses seen were usually disease stabilization.[35]
Antiestrogen treatment
Antiestrogen treatment, usually tamoxifen, plus sulindac has also resulted in disease stabilization.[36] A prospective trial of the combination of tamoxifen and sulindac reported few side effects, although asymptomatic ovarian cysts were common in girls. This combination showed relatively little activity, as measured by rates of response and PFS.[37][Level of evidence B4]
Surgery
Surgical resection should be used judiciously in patients with desmoid tumors because spontaneous regression can occur in up to 20% of cases. Surgical resection is recommended when tumor enlargement threatens the airway or when symptoms such as pain are persistent. A watch-and-wait strategy is otherwise preferred.
If surgery is chosen, the intent is to achieve clear margins. However, a retrospective review of children who underwent surgery for desmoid-type fibromatosis at St. Jude Children’s Research Hospital (SJCRH) reported no correlation between surgical margins and risk of recurrence.[19] In this study, 10 of 39 patients experienced a recurrence after surgery, with a median interval time of 2.5 years.
Radiation therapy
Radiation has been used for unresectable and symptomatic desmoid-type fibromatosis or postoperatively for tumors with inadequate resections if progression would have morbid consequences. The potential long-term complications of radiation therapy, especially subsequent neoplasms, make this modality less appealing in younger patients.[38]
Postoperative radiation therapy can be considered when recurrence or progression would entail additional surgery that might cause functional or cosmetic compromise and if radiation is considered acceptable in terms of morbidities.
Treatment options under clinical evaluation
Information about National Cancer Institute (NCI)-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Dermatofibrosarcoma Protuberans NOS
Dermatofibrosarcoma is a rare tumor that can be present in all age groups, but many of the reported cases arise in children.[39–41] A review of 451 cases in children younger than 20 years in the SEER Program database found that the incidence was 1 case per 1 million. The incidence was highest among Black patients aged 15 to 19 years. The most common sites were the trunk and extremities, which is similar to what is found in adults.
Ninety-five percent of patients underwent surgery. The overall survival (OS) rate was 100% at 5 years, 98% at 15 years, and 97% at 30 years. Male patients had decreased survival compared with female patients (P < .05).[42][Level of evidence C1]
Genomic alterations
The tumor has a consistent chromosomal translocation t(17;22)(q22;q13) that juxtaposes the COL1A1 gene with the PDGFRB gene.
Treatment of dermatofibrosarcoma protuberans
Guidelines for workup and management of dermatofibrosarcoma protuberans have been published.[43]
Treatment options for dermatofibrosarcoma protuberans include the following:
Most patients with dermatofibrosarcoma tumors can be cured by complete surgical resection. Wide excision with negative margins or Mohs/modified-Mohs surgery will prevent most tumors from recurring.[44] Despite the locally aggressive behavior of the tumor, lymph node or visceral metastasis rarely occurs.
Evidence (surgery):
The EpSSG prospective NRSTS 2005 (NCT00334854) trial identified 46 patients with dermatofibrosarcoma protuberans.[45] The median age at diagnosis was 6.9 years (range, 0.4–17.5 years). All patients had localized disease, 93% of patients had small tumors (<5 cm), and 76% of patients had Intergroup Rhabdomyosarcoma Study (IRS) group I tumors.
All patients underwent up-front surgery, and 32 patients required two procedures.
There were 11 patients with IRS group II tumors, 2 of whom went on to have a local recurrence.
After a median follow-up of 49 months (range, 4.2–130.9 months), all patients were alive at the time of this report.
The 5-year EFS rate was 92.6% (95% CI, 78.8%–97.6%), and the OS rate was 100%.
In retrospective reviews, postoperative radiation therapy after incomplete excision may have decreased the likelihood of recurrence.[46,47] Metastatic disease is more likely after multiple recurrences, and radiation or other adjuvant therapy should be considered in patients with recurrences that cannot be managed surgically.[40,42]
Targeted therapy (imatinib)
When surgical resection cannot be accomplished or the tumor is recurrent, treatment with imatinib (a tyrosine kinase inhibitor) has been effective in adult patients.[48–50]
Evidence (imatinib):
A systematic review of nine studies examined 152 adult patients with histologically proven dermatofibrosarcoma protuberans who were treated with imatinib.[51]
The study demonstrated a complete response rate of 5.2%, a partial response rate of 55.2%, and a stable disease rate of 27.6%.
There were no differences in the response rates based on imatinib dosing of either 400 mg or 800 mg per day.
Inflammatory Myofibroblastic Tumor and Epithelioid Inflammatory Myofibroblastic Sarcoma
Inflammatory myofibroblastic tumor is a rare mesenchymal tumor that is more common in children and adolescents.[52–54]
Clinical presentation
Inflammatory myofibroblastic tumors are rare tumors that affect soft tissues and visceral organs of children and young adults.[55] They rarely metastasize but tend to be locally invasive. Usual anatomical sites of disease include soft tissue, lungs, spleen, colon, and breast.[52] A review of 42 cases of pediatric inflammatory myofibroblastic tumor of the bladder was published in 2015.[56]
Epithelioid inflammatory myofibroblastic sarcoma is an uncommon subtype of inflammatory myofibroblastic tumors that shows a male predominance and can present from infancy through adulthood.[57–59] This subtype shows epithelioid morphology and a perinuclear or nuclear membrane pattern of immunostaining for ALK.[57] The most common site of presentation is the abdomen, although other primary sites have been reported.[57–59]
Genomic alterations
Roughly one-half of inflammatory myofibroblastic tumors exhibit a clonal variant that activates the ALK gene (encodes a receptor tyrosine kinase) at chromosome 2p23.[60]
Most cases of epithelioid inflammatory myofibroblastic sarcoma have RANBP2::ALK gene fusions. RRBP1::ALK gene fusions have also been reported.[57–59] Because RANBP2 localizes to the nuclear pore, this likely explains the perinuclear or nuclear membrane pattern of staining noted for ALK in epithelioid inflammatory myofibroblastic sarcoma.
ROS1 and PDGFRB kinase fusions were identified in 8 of 11 patients (73%) who were negative for ALK by immunohistochemistry.[61][Level of evidence C3]
Prognosis
Inflammatory myofibroblastic tumors recur frequently but are rarely metastatic.[52–54] Studies of children with inflammatory myofibroblastic tumor show 5-year survival rates higher than 80%.[62]
Epithelioid inflammatory myofibroblastic sarcoma is an aggressive tumor that is generally treated with surgery. Before the availability of ALK inhibitors, disease progression and high mortality rates were common.[57,58,63] Epithelioid inflammatory myofibroblastic sarcoma generally responds to ALK inhibitors but progression on therapy has been observed, which is consistent with the aggressive clinical behavior of the tumor.[59]
Treatment of inflammatory myofibroblastic tumor
Treatment options for inflammatory myofibroblastic tumor include the following:
Four patients achieved continuous remission after complete resection.
Three patients with residual disease experienced disease recurrence but later achieved continuous remission.
One patient with metastatic disease responded to multiagent chemotherapy.
In another study of 31 patients who underwent complete surgical resection, 4 patients had local disease recurrences.[62]
Of the 4 patients with local recurrences, all patients were alive after surgical re-resection (3 patients) or adjuvant chemotherapy and resection (1 patient).
A review of German studies identified 37 patients younger than 21 years with inflammatory myofibroblastic tumors.[66][Level of evidence C1]
Of 20 patients, 17 had complete resections with no recurrences. Surgical resections can be limited to those procedures that preserve form and function.
All other patients were treated with a combination of surgery and various chemotherapy regimens.
The overall 5-year EFS rate was 75%, and the OS rate was 91%.
A series of 32 patients aged 18 years and younger found that complete excision was the mainstay of therapy, although some patients were treated with steroids or cytotoxic chemotherapy.[67][Level of evidence C1]
The OS rate was 94%.
Three patients had relapsed disease, two of whom died of the disease.
When complete excision was performed, with or without other treatments such as steroids, there was a high survival rate for these patients.
The benefit of chemotherapy has been noted in case reports.[68] A prospective registry of children with inflammatory myofibroblastic tumor from the EpSSG (2005–2016) found an EFS rate of 82.9% and an OS rate of 98.1% at 5 years in all patients. The response rate for patients who received systemic therapy (chemotherapy or ALK inhibitor therapy) was 63% when used as front-line therapy and 66% when used as second-line therapy. Eight of ten patients who received vinblastine and low-dose methotrexate and all five patients who received ALK inhibitors (all of whom had ALK-positive tumors) responded to treatment.[62]
A retrospective, international, multicenter study analyzed patients younger than 21 years with ROS1-altered inflammatory myofibroblastic tumors who were enrolled in either the EpSSG NRSTS-2005 study or the Soft Tissue Sarcoma Registry. Primary surgery was recommended if a microscopic radical resection without disfigurement was feasible. Of the 19 patients, 12 received neoadjuvant systemic therapy as first-line treatment (high-dose steroids, n = 2; vinorelbine/vinblastine with methotrexate, n = 6; ROS1 inhibitors, n = 8). With a median follow-up of 2.8 years, seven patients had an event. The 3-year EFS rate was 41% (95% CI, 11%–71%), and the OS rate was 100%. While many patients in this series received crizotinib, the specific ROS1 inhibitor used for each patient was not specified.[69]
Steroid therapy or NSAID therapy
There are case reports of response to either steroids or NSAIDs.[62,70,71]
Targeted therapy (ALK inhibitors)
Inflammatory myofibroblastic tumors respond to ALK inhibitor therapy, as follows:
Crizotinib
Evidence (crizotinib):
For pediatric patients with measurable disease, the use of crizotinib achieved partial tumor responses in three of six patients with ALK-translocated inflammatory myofibroblastic tumors.[72]
A case report of a patient aged 16 years with metastatic/multifocal ALK-positive inflammatory myofibroblastic tumor demonstrated a complete response and a 3-year disease-free interval with crizotinib therapy.[73]
One study included 14 patients with inflammatory myofibroblastic tumors who were treated with crizotinib.[74][Level of evidence C3]
Five patients had complete responses, seven had partial responses, and the remaining two had stable disease.
No patient had relapsed disease at the time the article was published.
Two adult patients with ALK-rearranged inflammatory myofibroblastic tumor achieved partial responses with crizotinib.[75][Level of evidence C3]
An extensive review confirmed the effectiveness of crizotinib in children with inflammatory myofibroblastic tumors in various locations.[76]
The FDA approved crizotinib for patients aged 1 year and older with unresectable, recurrent, or refractory inflammatory ALK-positive myofibroblastic tumors.
Ceritinib
Evidence (ceritinib):
Two pediatric patients enrolled in a clinical trial responded to treatment with ceritinib.[77]
One patient had a complete response that was durable for multiple years on continuing therapy.
The other patient had a partial response when the drug was discontinued for severe liver and renal toxicity.
In a multicenter phase I study of ceritinib, 7 of 10 patients with inflammatory myofibroblastic tumor had objective responses to ceritinib.[78]
In a phase I trial of ceritinib for adult patients who were previously treated with ALK inhibitors, one patient with inflammatory myofibroblastic tumor had a partial response.[79]
Alectinib
A case report described the successful treatment of a patient with an inflammatory myofibroblastic tumor and a FN1::ALK gene fusion using alectinib, a second-generation ALK inhibitor.[80]
There are two distinct types of fibrosarcoma in children and adolescents, as follows:
Infantile fibrosarcoma is a malignant fibroblastic tumor usually characterized by ETV6::NTRK3 gene fusions.
Adult-type fibrosarcoma is composed of monomorphic fibroblastic tumor cells. Some of the genomic features of adult-type fibrosarcoma have been recently described.[81]
These are two distinct pathological diagnoses and require different treatments.
Clinical presentation
Infantile fibrosarcoma usually occurs in children younger than 1 year. This tumor usually presents with a rapidly growing mass, often noted at birth or even seen in the prenatal ultrasound. The tumors are frequently quite large at the time of presentation.[82] Hypercalcemia secondary to elevated levels of parathyroid hormone–related protein has been reported as a presenting feature of this disease in newborns.[83]
These tumors have a low incidence of metastases at diagnosis.
Genomic alterations
The tumor usually has a characteristic cytogenetic translocation t(12;15)(p13;q25) to create the ETV6::NTRK3 fusion gene. Infantile fibrosarcoma shares this translocation and a virtually identical histological appearance with mesoblastic nephroma.
Infantile fibrosarcoma occasionally occurs in children up to age 4 years. A tumor with similar morphology has been identified in older children. In these older children, the tumors do not have the ETV6::NTRK3 fusion that is characteristic of the tumors in younger patients.[84] BRAF intragenic deletions have been described in cases of infantile fibrosarcoma. Some of these tumors also contained NTRK3 fusions.[85] One study described four young children (aged 2–10 years) with tumors that were histologically classified as infantile fibrosarcoma and had ALK rearrangements.[86]
The Associazione Italiana Ematologia Oncologia Pediatrica analyzed a cohort of 44 pediatric patients with tumors classified as infantile fibrosarcomas/congenital mesoblastic nephromas. Eight infantile fibrosarcoma–like mesenchymal tumors found to be negative for the ETV6::NTRK3 fusion gene were analyzed by RNA sequencing to identify novel driver events. They identified three fusion genes involving RAF1: GOLGA4::RAF1, LRRFIP2::RAF1, and CLIP1::RAF1. The three fusion proteins retained the entire catalytic domain of the RAF1 kinase.[87]
Treatment of infantile fibrosarcoma
Treatment options for infantile fibrosarcoma include the following:
Complete resection is curative in most patients with infantile fibrosarcoma. However, the large size of the lesion frequently makes resection without major functional consequences impossible. For instance, tumors of the extremities often require amputation for complete excision.
The European pediatric group has reported that observation may also be an option in patients with microscopic residual disease after surgery.[88] Twelve patients with microscopic residual disease received no further therapy and two patients experienced disease relapse. One patient obtained a complete remission after chemotherapy. Postoperative chemotherapy was administered to patients with higher group disease and those who progressed. In a subsequent study, only one of seven patients with microscopic residual disease progressed during observation. That patient achieved complete remission with chemotherapy.[89][Level of evidence C1]
Preoperative chemotherapy has made a more conservative surgical approach possible. Agents active in this setting include vincristine, dactinomycin, cyclophosphamide, and ifosfamide.[90,91]; [89,92,93][Level of evidence C1] Three older studies of patients with infantile fibrosarcoma suggested that an alkylator-free regimen was effective and used as the first treatment choice in patients with macroscopic disease.[88,89,94] However, newer results of studies using NTRK inhibitors have suggested that kinase inhibitors are an appropriate initial therapy.
Targeted therapy
Larotrectinib
Larotrectinib is an oral ATP-competitive inhibitor of TRK A, B, and C.
Evidence (larotrectinib):
A phase I/II trial of larotrectinib was completed in patients with recurrent infantile fibrosarcoma who harbored an NTRK gene fusion.[95]
Durable objective responses were seen in all eight patients, and responses occurred at a median of 1.7 months.
Most toxicities were grades 1 and 2, which included transaminitis, leukopenia, neutropenia, and vomiting. There were no grade 4 or grade 5 events attributed to larotrectinib.
In another study, three of five patients who achieved a partial response after neoadjuvant larotrectinib underwent a complete surgical resection with negative margins.[96,97]; [98][Level of evidence C2]
These three patients achieved an excellent pathological response (>98% treatment effect) and remained disease free 7 to 15 months after surgery.
In a follow-up report, 159 patients with TRK fusion–positive tumors were enrolled in three phase I/II trials. There were 28 patients with infantile fibrosarcoma who were treated with single-agent larotrectinib.[99][Level of evidence C2]
The response rate was 96%.
The Children’s Oncology Group conducted a phase II histology-agnostic trial of larotrectinib in children with NTRK fusion–positive solid tumors.[100] The study enrolled 33 patients, 18 with infantile fibrosarcoma and 15 with other solid tumors.
The overall response rate within six cycles was 94% (17 of 18; 95% adjusted CI, 72.7%–98.6%) for children with infantile fibrosarcoma and 60% (9 of 15; 95% CI, 32.3%–83.7%) for children with other solid tumors.
Six percent (2 of 33; 95% CI, 0.7%–22.2%) of patients developed progressive disease while on therapy.
The 2-year EFS and OS rates among these groups were 82.2% (95% CI, 54.3%–93.9%) and 93.8% (95% CI, 63.2%–99.1%) for infantile fibrosarcoma and 80% (95% CI, 50.0%–93.1%) and 93.3% (95% CI, 61.3%–99.0%) for other solid tumors, respectively.
Patients who underwent surgical resection of their tumor had prolonged EFS, with only 1 of these 16 patients experiencing disease progression.
Other TRK inhibitors
LOXO-195: In a clinical trial, 1 of 8 pediatric patients with an ETV6::NTRK3–rearranged infantile fibrosarcoma developed progressive disease after 8 months of larotrectinib therapy and was found to have an acquired G623R resistance variant. The patient was treated with LOXO-195, a selective TRK inhibitor designed to overcome acquired resistance mediated by recurrent kinase domain variants. The patient experienced a transient partial response.[101] LOXO-195 is no longer being developed.
VEGFR inhibitor
Pazopanib: A patient aged 2 months with infantile fibrosarcoma was initially treated with chemotherapy. At disease progression, a response was seen with pazopanib therapy.[102]
Treatment options under clinical evaluation
Information about NCI-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Fibrosarcoma NOS
These tumors lack the translocation seen in infantile fibrosarcomas. They present like most nonrhabdomyosarcomas, and the management approach is similar.
Myxofibrosarcoma
Myxofibrosarcoma is a rare lesion, especially in childhood. It is typically treated with complete surgical resection.
Low-Grade Fibromyxoid Sarcoma
Low-grade fibromyxoid sarcoma is a histologically deceptive soft tissue neoplasm that most commonly affects young and middle-aged adults. It is commonly located deep within the extremities.[103–105]
A Children’s Oncology Group (COG) trial (ARST0332 [NCT00346164]) enrolled 11 patients with this tumor type. The median age at diagnosis was 13 years, and males were more commonly affected. The most common tumor sites were the lower and upper extremities (n = 9). None of the patients developed local or distant disease recurrence at a median follow up of 2.7 years.[106]
Genomic alterations
Low-grade fibromyxoid sarcoma is characterized by a FUS::CREB3L2 gene translocation and, rarely, alternative gene translocations such as FUS::CREB3L1 and EWSR1::CREB3L1.[107,108]
Prognosis
In a review of 33 patients (3 were younger than 18 years) with low-grade fibromyxoid sarcoma, 21 patients developed a local recurrence after intervals of up to 15 years (median, 3.5 years). Fifteen patients developed metastases up to 45 years (median, 5 years) from diagnosis, most commonly to the lungs and pleura. This finding emphasizes the need for continued follow-up of these patients.[103] Even after metastases occur, the disease course may be indolent.[109]
In another report, 14 of 73 patients were younger than 18 years. In this series with a relatively short follow-up (median of 24 months), only 8 of 54 patients with adequate follow-up developed local (9%) or distant (6%) disease recurrence. This report suggested that the behavior of this tumor might be significantly better than previously reported.[110] However, because late metastases can occur, careful monitoring of these patients is warranted.
A study by the Ultra-Rare Sarcoma Working Group examined 32 patients with rare occurrences of distant metastases (20 with metastases at diagnosis, 12 after initial resection). Most metastases occurred in the lungs. Treatments varied, and minimal responses were observed to anthracycline-based and gemcitabine-based regimens with trabectidin. However, there were few patients in each treatment group.[111]
Treatment of low-grade fibromyxoid sarcoma
Treatment options for low-grade fibromyxoid sarcoma include the following:
Surgery.
Low-grade fibromyxoid sarcoma is not very chemosensitive, and the limited treatment information suggests that surgery is the treatment of choice.[109]
Evidence (surgery):
The German Cooperative Weichteilsarkom Studiengruppe (CWS) reported study results for 31 patients younger than 21 years with low-grade fibromyxoid sarcoma.[104][Level of evidence C2]
The 5-year EFS rate was 71% (95% CI, ±18.6%), the 5-year local relapse-free survival rate was 76% (95% CI, ±17.6%), and the 5-year OS rate was 100%.
Among 24 patients who had R0 resections (complete resection with negative microscopic margins), 5 patients (21%) experienced disease relapses (3 local, 1 metastatic, and 1 combined).
Among seven patients who had R1 resections (margins were microscopically positive), three patients (43%) experienced local disease relapses.
There are little data regarding the use of chemotherapy and/or radiation therapy in this disease. One report suggests that trabectedin may be effective in the treatment of low-grade fibromyxoid sarcoma.[112]
Sclerosing Epithelioid Fibrosarcoma
Sclerosing epithelioid fibrosarcoma is a rare malignant sarcoma that commonly harbors EWSR1 gene fusions and has an aggressive clinical course. The tumor responds poorly to chemotherapy.[113,114]
Genomic characteristics
Sclerosing epithelioid fibrosarcoma most commonly has the EWSR1::CREB3L1 gene fusion. However, EWSR1 may have other partners, including CREB3L2 and CREB3L3.[115,116] Gene fusions involving FUS (including the FUS::CREB3L2 fusion associated with low-grade fibromyxoid sarcoma) and PAX5 (e.g., PAX5::CREB3L1) are uncommon but can occur.[116,117] For cases of sclerosing epithelioid fibrosarcoma that lack MUC4 expression, EWSR1 gene fusions are generally absent, while a gene fusion involving YAP1 and KMT2A is commonly observed.[113,115,118,119] Sclerosing epithelioid fibrosarcoma has more structural and chromosomal segmental alterations than low-grade fibromyxoid fibrosarcoma.[115]
Treatment of sclerosing epithelioid fibrosarcoma
Treatment options for sclerosing epithelioid fibrosarcoma include the following:
Surgery.
The tumor responds poorly to chemotherapy.[120] Therefore, it is typically treated with complete surgical excision. Long-term follow-up is recommended because late local recurrences and metastases can occur.
References
Lewis JJ, Boland PJ, Leung DH, et al.: The enigma of desmoid tumors. Ann Surg 229 (6): 866-72; discussion 872-3, 1999. [PUBMED Abstract]
Gounder MM, Mahoney MR, Van Tine BA, et al.: Sorafenib for Advanced and Refractory Desmoid Tumors. N Engl J Med 379 (25): 2417-2428, 2018. [PUBMED Abstract]
Nieuwenhuis MH, Casparie M, Mathus-Vliegen LM, et al.: A nation-wide study comparing sporadic and familial adenomatous polyposis-related desmoid-type fibromatoses. Int J Cancer 129 (1): 256-61, 2011. [PUBMED Abstract]
Lazar AJ, Tuvin D, Hajibashi S, et al.: Specific mutations in the beta-catenin gene (CTNNB1) correlate with local recurrence in sporadic desmoid tumors. Am J Pathol 173 (5): 1518-27, 2008. [PUBMED Abstract]
Rossato M, Rigotti M, Grazia M, et al.: Congenital hypertrophy of the retinal pigment epithelium (CHRPE) and familial adenomatous polyposis (FAP). Acta Ophthalmol Scand 74 (4): 338-42, 1996. [PUBMED Abstract]
Baker RH, Heinemann MH, Miller HH, et al.: Hyperpigmented lesions of the retinal pigment epithelium in familial adenomatous polyposis. Am J Med Genet 31 (2): 427-35, 1988. [PUBMED Abstract]
Kattentidt Mouravieva AA, Geurts-Giele IR, de Krijger RR, et al.: Identification of Familial Adenomatous Polyposis carriers among children with desmoid tumours. Eur J Cancer 48 (12): 1867-74, 2012. [PUBMED Abstract]
Wang WL, Nero C, Pappo A, et al.: CTNNB1 genotyping and APC screening in pediatric desmoid tumors: a proposed algorithm. Pediatr Dev Pathol 15 (5): 361-7, 2012 Sep-Oct. [PUBMED Abstract]
Baday YI, Navai SA, Hicks MJ, et al.: Pediatric liposarcoma: A case series and literature review. Pediatr Blood Cancer 68 (12): e29327, 2021. [PUBMED Abstract]
Merchant NB, Lewis JJ, Woodruff JM, et al.: Extremity and trunk desmoid tumors: a multifactorial analysis of outcome. Cancer 86 (10): 2045-52, 1999. [PUBMED Abstract]
Faulkner LB, Hajdu SI, Kher U, et al.: Pediatric desmoid tumor: retrospective analysis of 63 cases. J Clin Oncol 13 (11): 2813-8, 1995. [PUBMED Abstract]
Honeyman JN, Theilen TM, Knowles MA, et al.: Desmoid fibromatosis in children and adolescents: a conservative approach to management. J Pediatr Surg 48 (1): 62-6, 2013. [PUBMED Abstract]
Bonvalot S, Ternès N, Fiore M, et al.: Spontaneous regression of primary abdominal wall desmoid tumors: more common than previously thought. Ann Surg Oncol 20 (13): 4096-102, 2013. [PUBMED Abstract]
Bonvalot S, Eldweny H, Haddad V, et al.: Extra-abdominal primary fibromatosis: Aggressive management could be avoided in a subgroup of patients. Eur J Surg Oncol 34 (4): 462-8, 2008. [PUBMED Abstract]
Merchant TE, Nguyen D, Walter AW, et al.: Long-term results with radiation therapy for pediatric desmoid tumors. Int J Radiat Oncol Biol Phys 47 (5): 1267-71, 2000. [PUBMED Abstract]
Zelefsky MJ, Harrison LB, Shiu MH, et al.: Combined surgical resection and iridium 192 implantation for locally advanced and recurrent desmoid tumors. Cancer 67 (2): 380-4, 1991. [PUBMED Abstract]
Weiss AJ, Lackman RD: Low-dose chemotherapy of desmoid tumors. Cancer 64 (6): 1192-4, 1989. [PUBMED Abstract]
Klein WA, Miller HH, Anderson M, et al.: The use of indomethacin, sulindac, and tamoxifen for the treatment of desmoid tumors associated with familial polyposis. Cancer 60 (12): 2863-8, 1987. [PUBMED Abstract]
Soto-Miranda MA, Sandoval JA, Rao B, et al.: Surgical treatment of pediatric desmoid tumors. A 12-year, single-center experience. Ann Surg Oncol 20 (11): 3384-90, 2013. [PUBMED Abstract]
Desmoid Tumor Working Group: The management of desmoid tumours: A joint global consensus-based guideline approach for adult and paediatric patients. Eur J Cancer 127: 96-107, 2020. [PUBMED Abstract]
Penel N, Le Cesne A, Bonvalot S, et al.: Surgical versus non-surgical approach in primary desmoid-type fibromatosis patients: A nationwide prospective cohort from the French Sarcoma Group. Eur J Cancer 83: 125-131, 2017. [PUBMED Abstract]
Orbach D, Brennan B, Bisogno G, et al.: The EpSSG NRSTS 2005 treatment protocol for desmoid-type fibromatosis in children: an international prospective case series. Lancet Child Adolesc Health 1 (4): 284-292, 2017. [PUBMED Abstract]
Skapek SX, Ferguson WS, Granowetter L, et al.: Vinblastine and methotrexate for desmoid fibromatosis in children: results of a Pediatric Oncology Group Phase II Trial. J Clin Oncol 25 (5): 501-6, 2007. [PUBMED Abstract]
Gega M, Yanagi H, Yoshikawa R, et al.: Successful chemotherapeutic modality of doxorubicin plus dacarbazine for the treatment of desmoid tumors in association with familial adenomatous polyposis. J Clin Oncol 24 (1): 102-5, 2006. [PUBMED Abstract]
Constantinidou A, Jones RL, Scurr M, et al.: Pegylated liposomal doxorubicin, an effective, well-tolerated treatment for refractory aggressive fibromatosis. Eur J Cancer 45 (17): 2930-4, 2009. [PUBMED Abstract]
Ananth P, Werger A, Voss S, et al.: Liposomal doxorubicin: Effective treatment for pediatric desmoid fibromatosis. Pediatr Blood Cancer 64 (7): , 2017. [PUBMED Abstract]
Ferrari A, Orbach D, Affinita MC, et al.: Evidence of hydroxyurea activity in children with pretreated desmoid-type fibromatosis: A new option in the armamentarium of systemic therapies. Pediatr Blood Cancer 66 (1): e27472, 2019. [PUBMED Abstract]
Kornreich L, Orbach D, Nicolas N, et al.: Oral vinorelbine in young patients with desmoid-type fibromatosis. Tumori 109 (5): 511-518, 2023. [PUBMED Abstract]
Agresta L, Kim H, Turpin BK, et al.: Pazopanib therapy for desmoid tumors in adolescent and young adult patients. Pediatr Blood Cancer 65 (6): e26968, 2018. [PUBMED Abstract]
Toulmonde M, Pulido M, Ray-Coquard I, et al.: Pazopanib or methotrexate-vinblastine combination chemotherapy in adult patients with progressive desmoid tumours (DESMOPAZ): a non-comparative, randomised, open-label, multicentre, phase 2 study. Lancet Oncol 20 (9): 1263-1272, 2019. [PUBMED Abstract]
Shang H, Braggio D, Lee YJ, et al.: Targeting the Notch pathway: A potential therapeutic approach for desmoid tumors. Cancer 121 (22): 4088-96, 2015. [PUBMED Abstract]
Kummar S, O’Sullivan Coyne G, Do KT, et al.: Clinical Activity of the γ-Secretase Inhibitor PF-03084014 in Adults With Desmoid Tumors (Aggressive Fibromatosis). J Clin Oncol 35 (14): 1561-1569, 2017. [PUBMED Abstract]
Gounder M, Ratan R, Alcindor T, et al.: Nirogacestat, a γ-Secretase Inhibitor for Desmoid Tumors. N Engl J Med 388 (10): 898-912, 2023. [PUBMED Abstract]
Takahashi T, Prensner JR, Robson CD, et al.: Safety and efficacy of gamma-secretase inhibitor nirogacestat (PF-03084014) in desmoid tumor: Report of four pediatric/young adult cases. Pediatr Blood Cancer 67 (10): e28636, 2020. [PUBMED Abstract]
Meazza C, Bisogno G, Gronchi A, et al.: Aggressive fibromatosis in children and adolescents: the Italian experience. Cancer 116 (1): 233-40, 2010. [PUBMED Abstract]
Hansmann A, Adolph C, Vogel T, et al.: High-dose tamoxifen and sulindac as first-line treatment for desmoid tumors. Cancer 100 (3): 612-20, 2004. [PUBMED Abstract]
Skapek SX, Anderson JR, Hill DA, et al.: Safety and efficacy of high-dose tamoxifen and sulindac for desmoid tumor in children: results of a Children’s Oncology Group (COG) phase II study. Pediatr Blood Cancer 60 (7): 1108-12, 2013. [PUBMED Abstract]
Rutenberg MS, Indelicato DJ, Knapik JA, et al.: External-beam radiotherapy for pediatric and young adult desmoid tumors. Pediatr Blood Cancer 57 (3): 435-42, 2011. [PUBMED Abstract]
Buckley PG, Mantripragada KK, Benetkiewicz M, et al.: A full-coverage, high-resolution human chromosome 22 genomic microarray for clinical and research applications. Hum Mol Genet 11 (25): 3221-9, 2002. [PUBMED Abstract]
Valdivielso-Ramos M, Torrelo A, Campos M, et al.: Pediatric dermatofibrosarcoma protuberans in Madrid, Spain: multi-institutional outcomes. Pediatr Dermatol 31 (6): 676-82, 2014 Nov-Dec. [PUBMED Abstract]
Gooskens SL, Oranje AP, van Adrichem LN, et al.: Imatinib mesylate for children with dermatofibrosarcoma protuberans (DFSP). Pediatr Blood Cancer 55 (2): 369-73, 2010. [PUBMED Abstract]
Rubio GA, Alvarado A, Gerth DJ, et al.: Incidence and Outcomes of Dermatofibrosarcoma Protuberans in the US Pediatric Population. J Craniofac Surg 28 (1): 182-184, 2017. [PUBMED Abstract]
Miller SJ, Alam M, Andersen JS, et al.: Dermatofibrosarcoma protuberans. J Natl Compr Canc Netw 10 (3): 312-8, 2012. [PUBMED Abstract]
Meguerditchian AN, Wang J, Lema B, et al.: Wide excision or Mohs micrographic surgery for the treatment of primary dermatofibrosarcoma protuberans. Am J Clin Oncol 33 (3): 300-3, 2010. [PUBMED Abstract]
Brennan B, Zanetti I, De Salvo GL, et al.: Dermatofibrosarcoma protuberans in children and adolescents: The European Paediatric Soft Tissue Sarcoma Study Group prospective trial (EpSSG NRSTS 2005). Pediatr Blood Cancer 67 (10): e28351, 2020. [PUBMED Abstract]
Dagan R, Morris CG, Zlotecki RA, et al.: Radiotherapy in the treatment of dermatofibrosarcoma protuberans. Am J Clin Oncol 28 (6): 537-9, 2005. [PUBMED Abstract]
Sun LM, Wang CJ, Huang CC, et al.: Dermatofibrosarcoma protuberans: treatment results of 35 cases. Radiother Oncol 57 (2): 175-81, 2000. [PUBMED Abstract]
Price VE, Fletcher JA, Zielenska M, et al.: Imatinib mesylate: an attractive alternative in young children with large, surgically challenging dermatofibrosarcoma protuberans. Pediatr Blood Cancer 44 (5): 511-5, 2005. [PUBMED Abstract]
McArthur GA, Demetri GD, van Oosterom A, et al.: Molecular and clinical analysis of locally advanced dermatofibrosarcoma protuberans treated with imatinib: Imatinib Target Exploration Consortium Study B2225. J Clin Oncol 23 (4): 866-73, 2005. [PUBMED Abstract]
Rutkowski P, Van Glabbeke M, Rankin CJ, et al.: Imatinib mesylate in advanced dermatofibrosarcoma protuberans: pooled analysis of two phase II clinical trials. J Clin Oncol 28 (10): 1772-9, 2010. [PUBMED Abstract]
Navarrete-Dechent C, Mori S, Barker CA, et al.: Imatinib Treatment for Locally Advanced or Metastatic Dermatofibrosarcoma Protuberans: A Systematic Review. JAMA Dermatol 155 (3): 361-369, 2019. [PUBMED Abstract]
Brodlie M, Barwick SC, Wood KM, et al.: Inflammatory myofibroblastic tumours of the respiratory tract: paediatric case series with varying clinical presentations. J Laryngol Otol 125 (8): 865-8, 2011. [PUBMED Abstract]
Xiao Y, Zhou S, Ma C, et al.: Radiological and histopathological features of hepatic inflammatory myofibroblastic tumour: analysis of 10 cases. Clin Radiol 68 (11): 1114-20, 2013. [PUBMED Abstract]
Karnak I, Senocak ME, Ciftci AO, et al.: Inflammatory myofibroblastic tumor in children: diagnosis and treatment. J Pediatr Surg 36 (6): 908-12, 2001. [PUBMED Abstract]
Collin M, Charles A, Barker A, et al.: Inflammatory myofibroblastic tumour of the bladder in children: a review. J Pediatr Urol 11 (5): 239-45, 2015. [PUBMED Abstract]
Mariño-Enríquez A, Wang WL, Roy A, et al.: Epithelioid inflammatory myofibroblastic sarcoma: An aggressive intra-abdominal variant of inflammatory myofibroblastic tumor with nuclear membrane or perinuclear ALK. Am J Surg Pathol 35 (1): 135-44, 2011. [PUBMED Abstract]
Lee JC, Li CF, Huang HY, et al.: ALK oncoproteins in atypical inflammatory myofibroblastic tumours: novel RRBP1-ALK fusions in epithelioid inflammatory myofibroblastic sarcoma. J Pathol 241 (3): 316-323, 2017. [PUBMED Abstract]
Trahair T, Gifford AJ, Fordham A, et al.: Crizotinib and Surgery for Long-Term Disease Control in Children and Adolescents With ALK-Positive Inflammatory Myofibroblastic Tumors. JCO Precis Oncol 3: , 2019. [PUBMED Abstract]
Coffin CM, Hornick JL, Fletcher CD: Inflammatory myofibroblastic tumor: comparison of clinicopathologic, histologic, and immunohistochemical features including ALK expression in atypical and aggressive cases. Am J Surg Pathol 31 (4): 509-20, 2007. [PUBMED Abstract]
Lovly CM, Gupta A, Lipson D, et al.: Inflammatory myofibroblastic tumors harbor multiple potentially actionable kinase fusions. Cancer Discov 4 (8): 889-95, 2014. [PUBMED Abstract]
Casanova M, Brennan B, Alaggio R, et al.: Inflammatory myofibroblastic tumor: The experience of the European pediatric Soft Tissue Sarcoma Study Group (EpSSG). Eur J Cancer 127: 123-129, 2020. [PUBMED Abstract]
Yu L, Liu J, Lao IW, et al.: Epithelioid inflammatory myofibroblastic sarcoma: a clinicopathological, immunohistochemical and molecular cytogenetic analysis of five additional cases and review of the literature. Diagn Pathol 11 (1): 67, 2016. [PUBMED Abstract]
Devaney KO, Lafeir DJ, Triantafyllou A, et al.: Inflammatory myofibroblastic tumors of the head and neck: evaluation of clinicopathologic and prognostic features. Eur Arch Otorhinolaryngol 269 (12): 2461-5, 2012. [PUBMED Abstract]
Kube S, Vokuhl C, Dantonello T, et al.: Inflammatory myofibroblastic tumors-A retrospective analysis of the Cooperative Weichteilsarkom Studiengruppe. Pediatr Blood Cancer 65 (6): e27012, 2018. [PUBMED Abstract]
Dalton BG, Thomas PG, Sharp NE, et al.: Inflammatory myofibroblastic tumors in children. J Pediatr Surg 51 (4): 541-4, 2016. [PUBMED Abstract]
Favini F, Resti AG, Collini P, et al.: Inflammatory myofibroblastic tumor of the conjunctiva: response to chemotherapy with low-dose methotrexate and vinorelbine. Pediatr Blood Cancer 54 (3): 483-5, 2010. [PUBMED Abstract]
Schoot RA, Orbach D, Minard Colin V, et al.: Inflammatory Myofibroblastic Tumor With ROS1 Gene Fusions in Children and Young Adolescents. JCO Precis Oncol 7: e2300323, 2023. [PUBMED Abstract]
Doski JJ, Priebe CJ, Driessnack M, et al.: Corticosteroids in the management of unresected plasma cell granuloma (inflammatory pseudotumor) of the lung. J Pediatr Surg 26 (9): 1064-6, 1991. [PUBMED Abstract]
Diop B, Konate I, Ka S, et al.: Mesenteric myofibroblastic tumor: NSAID therapy after incomplete resection. J Visc Surg 148 (4): e311-4, 2011. [PUBMED Abstract]
Mossé YP, Lim MS, Voss SD, et al.: Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children’s Oncology Group phase 1 consortium study. Lancet Oncol 14 (6): 472-80, 2013. [PUBMED Abstract]
Gaudichon J, Jeanne-Pasquier C, Deparis M, et al.: Complete and Repeated Response of a Metastatic ALK-rearranged Inflammatory Myofibroblastic Tumor to Crizotinib in a Teenage Girl. J Pediatr Hematol Oncol 38 (4): 308-11, 2016. [PUBMED Abstract]
Mossé YP, Voss SD, Lim MS, et al.: Targeting ALK With Crizotinib in Pediatric Anaplastic Large Cell Lymphoma and Inflammatory Myofibroblastic Tumor: A Children’s Oncology Group Study. J Clin Oncol 35 (28): 3215-3221, 2017. [PUBMED Abstract]
Butrynski JE, D’Adamo DR, Hornick JL, et al.: Crizotinib in ALK-rearranged inflammatory myofibroblastic tumor. N Engl J Med 363 (18): 1727-33, 2010. [PUBMED Abstract]
Brivio E, Zwaan CM: ALK inhibition in two emblematic cases of pediatric inflammatory myofibroblastic tumor: Efficacy and side effects. Pediatr Blood Cancer 66 (5): e27645, 2019. [PUBMED Abstract]
Fischer M, Moreno L, Ziegler DS, et al.: Ceritinib in paediatric patients with anaplastic lymphoma kinase-positive malignancies: an open-label, multicentre, phase 1, dose-escalation and dose-expansion study. Lancet Oncol 22 (12): 1764-1776, 2021. [PUBMED Abstract]
Nishio M, Murakami H, Horiike A, et al.: Phase I Study of Ceritinib (LDK378) in Japanese Patients with Advanced, Anaplastic Lymphoma Kinase-Rearranged Non-Small-Cell Lung Cancer or Other Tumors. J Thorac Oncol 10 (7): 1058-66, 2015. [PUBMED Abstract]
Fujiki T, Sakai Y, Ikawa Y, et al.: Pediatric inflammatory myofibroblastic tumor of the bladder with ALK-FN1 fusion successfully treated by alectinib. Pediatr Blood Cancer 70 (4): e30172, 2023. [PUBMED Abstract]
Gounder MM, Agaram NP, Trabucco SE, et al.: Clinical genomic profiling in the management of patients with soft tissue and bone sarcoma. Nat Commun 13 (1): 3406, 2022. [PUBMED Abstract]
Sulkowski JP, Raval MV, Browne M: Margin status and multimodal therapy in infantile fibrosarcoma. Pediatr Surg Int 29 (8): 771-6, 2013. [PUBMED Abstract]
Hirschfeld R, Welch JJG, Harrison DJ, et al.: Two cases of humoral hypercalcemia of malignancy complicating infantile fibrosarcoma. Pediatr Blood Cancer 64 (10): , 2017. [PUBMED Abstract]
Kao YC, Fletcher CDM, Alaggio R, et al.: Recurrent BRAF Gene Fusions in a Subset of Pediatric Spindle Cell Sarcomas: Expanding the Genetic Spectrum of Tumors With Overlapping Features With Infantile Fibrosarcoma. Am J Surg Pathol 42 (1): 28-38, 2018. [PUBMED Abstract]
Wegert J, Vokuhl C, Collord G, et al.: Recurrent intragenic rearrangements of EGFR and BRAF in soft tissue tumors of infants. Nat Commun 9 (1): 2378, 2018. [PUBMED Abstract]
Tan SY, Al-Ibraheemi A, Ahrens WA, et al.: ALK rearrangements in infantile fibrosarcoma-like spindle cell tumours of soft tissue and kidney. Histopathology 80 (4): 698-707, 2022. [PUBMED Abstract]
Motta M, Barresi S, Pizzi S, et al.: RAF1 gene fusions are recurrent driver events in infantile fibrosarcoma-like mesenchymal tumors. J Pathol 263 (2): 166-177, 2024. [PUBMED Abstract]
Orbach D, Rey A, Cecchetto G, et al.: Infantile fibrosarcoma: management based on the European experience. J Clin Oncol 28 (2): 318-23, 2010. [PUBMED Abstract]
Orbach D, Brennan B, De Paoli A, et al.: Conservative strategy in infantile fibrosarcoma is possible: The European paediatric Soft tissue sarcoma Study Group experience. Eur J Cancer 57: 1-9, 2016. [PUBMED Abstract]
Hawkins DS, Black JO, Orbach D, et al.: Nonrhabdomyosarcoma soft-tissue sarcomas. In: Blaney SM, Helman LJ, Adamson PC, eds.: Pizzo and Poplack’s Pediatric Oncology. 8th ed. Wolters Kluwer, 2020, pp 721-46.
Loh ML, Ahn P, Perez-Atayde AR, et al.: Treatment of infantile fibrosarcoma with chemotherapy and surgery: results from the Dana-Farber Cancer Institute and Children’s Hospital, Boston. J Pediatr Hematol Oncol 24 (9): 722-6, 2002. [PUBMED Abstract]
Akyüz C, Küpeli S, Varan A, et al.: Infantile fibrosarcoma: retrospective analysis of eleven patients. Tumori 97 (2): 166-9, 2011 Mar-Apr. [PUBMED Abstract]
Gallego S, Pericas N, Barber I, et al.: Infantile fibrosarcoma of the retroperitoneum: a site of unfavorable prognosis? Pediatr Hematol Oncol 28 (5): 451-3, 2011. [PUBMED Abstract]
Parida L, Fernandez-Pineda I, Uffman JK, et al.: Clinical management of infantile fibrosarcoma: a retrospective single-institution review. Pediatr Surg Int 29 (7): 703-8, 2013. [PUBMED Abstract]
Laetsch TW, DuBois SG, Mascarenhas L, et al.: Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol 19 (5): 705-714, 2018. [PUBMED Abstract]
Kummar S, Lassen UN: TRK Inhibition: A New Tumor-Agnostic Treatment Strategy. Target Oncol 13 (5): 545-556, 2018. [PUBMED Abstract]
Drilon A, Laetsch TW, Kummar S, et al.: Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med 378 (8): 731-739, 2018. [PUBMED Abstract]
DuBois SG, Laetsch TW, Federman N, et al.: The use of neoadjuvant larotrectinib in the management of children with locally advanced TRK fusion sarcomas. Cancer 124 (21): 4241-4247, 2018. [PUBMED Abstract]
Hong DS, DuBois SG, Kummar S, et al.: Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 21 (4): 531-540, 2020. [PUBMED Abstract]
Laetsch TW, Voss S, Ludwig K, et al.: Larotrectinib for Newly Diagnosed Infantile Fibrosarcoma and Other Pediatric NTRK Fusion-Positive Solid Tumors (Children’s Oncology Group ADVL1823). J Clin Oncol 43 (10): 1188-1197, 2025. [PUBMED Abstract]
Drilon A, Nagasubramanian R, Blake JF, et al.: A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov 7 (9): 963-972, 2017. [PUBMED Abstract]
Yanagisawa R, Noguchi M, Fujita K, et al.: Preoperative Treatment With Pazopanib in a Case of Chemotherapy-Resistant Infantile Fibrosarcoma. Pediatr Blood Cancer 63 (2): 348-51, 2016. [PUBMED Abstract]
Evans HL: Low-grade fibromyxoid sarcoma: a clinicopathologic study of 33 cases with long-term follow-up. Am J Surg Pathol 35 (10): 1450-62, 2011. [PUBMED Abstract]
Scheer M, Vokuhl C, Veit-Friedrich I, et al.: Low-grade fibromyxoid sarcoma: A report of the Cooperative Weichteilsarkom Studiengruppe (CWS). Pediatr Blood Cancer 67 (2): e28009, 2020. [PUBMED Abstract]
Fletcher CDM, Bridge JA, Hogendoorn P, et al., eds.: WHO Classification of Tumours of Soft Tissue and Bone. 4th ed. IARC Press, 2013.
Sargar K, Kao SC, Spunt SL, et al.: MRI and CT of Low-Grade Fibromyxoid Sarcoma in Children: A Report From Children’s Oncology Group Study ARST0332. AJR Am J Roentgenol 205 (2): 414-20, 2015. [PUBMED Abstract]
Guillou L, Benhattar J, Gengler C, et al.: Translocation-positive low-grade fibromyxoid sarcoma: clinicopathologic and molecular analysis of a series expanding the morphologic spectrum and suggesting potential relationship to sclerosing epithelioid fibrosarcoma: a study from the French Sarcoma Group. Am J Surg Pathol 31 (9): 1387-402, 2007. [PUBMED Abstract]
Mohamed M, Fisher C, Thway K: Low-grade fibromyxoid sarcoma: Clinical, morphologic and genetic features. Ann Diagn Pathol 28: 60-67, 2017. [PUBMED Abstract]
O’Sullivan MJ, Sirgi KE, Dehner LP: Low-grade fibrosarcoma (hyalinizing spindle cell tumor with giant rosettes) with pulmonary metastases at presentation: case report and review of the literature. Int J Surg Pathol 10 (3): 211-6, 2002. [PUBMED Abstract]
Folpe AL, Lane KL, Paull G, et al.: Low-grade fibromyxoid sarcoma and hyalinizing spindle cell tumor with giant rosettes: a clinicopathologic study of 73 cases supporting their identity and assessing the impact of high-grade areas. Am J Surg Pathol 24 (10): 1353-60, 2000. [PUBMED Abstract]
Giani C, Denu RA, Ljevar S, et al.: Low-grade fibromyxoid sarcoma and sclerosing epithelioid fibrosarcoma, outcome of advanced disease: retrospective study from the Ultra-Rare Sarcoma Working Group. ESMO Open 9 (9): 103689, 2024. [PUBMED Abstract]
Maretty-Nielsen K, Baerentzen S, Keller J, et al.: Low-Grade Fibromyxoid Sarcoma: Incidence, Treatment Strategy of Metastases, and Clinical Significance of the FUS Gene. Sarcoma 2013: 256280, 2013. [PUBMED Abstract]
Prieto-Granada C, Zhang L, Chen HW, et al.: A genetic dichotomy between pure sclerosing epithelioid fibrosarcoma (SEF) and hybrid SEF/low-grade fibromyxoid sarcoma: a pathologic and molecular study of 18 cases. Genes Chromosomes Cancer 54 (1): 28-38, 2015. [PUBMED Abstract]
Arbajian E, Puls F, Antonescu CR, et al.: In-depth Genetic Analysis of Sclerosing Epithelioid Fibrosarcoma Reveals Recurrent Genomic Alterations and Potential Treatment Targets. Clin Cancer Res 23 (23): 7426-7434, 2017. [PUBMED Abstract]
Arbajian E, Puls F, Magnusson L, et al.: Recurrent EWSR1-CREB3L1 gene fusions in sclerosing epithelioid fibrosarcoma. Am J Surg Pathol 38 (6): 801-8, 2014. [PUBMED Abstract]
Dewaele B, Libbrecht L, Levy G, et al.: A novel EWS-CREB3L3 gene fusion in a mesenteric sclerosing epithelioid fibrosarcoma. Genes Chromosomes Cancer 56 (9): 695-699, 2017. [PUBMED Abstract]
Porteus C, Gan Q, Gong Y, et al.: Sclerosing epithelioid fibrosarcoma: cytologic characterization with histologic, immunohistologic, molecular, and clinical correlation of 8 cases. J Am Soc Cytopathol 9 (6): 513-519, 2020 Nov – Dec. [PUBMED Abstract]
Puls F, Agaimy A, Flucke U, et al.: Recurrent Fusions Between YAP1 and KMT2A in Morphologically Distinct Neoplasms Within the Spectrum of Low-grade Fibromyxoid Sarcoma and Sclerosing Epithelioid Fibrosarcoma. Am J Surg Pathol 44 (5): 594-606, 2020. [PUBMED Abstract]
Warmke LM, Meis JM: Sclerosing Epithelioid Fibrosarcoma: A Distinct Sarcoma With Aggressive Features. Am J Surg Pathol 45 (3): 317-328, 2021. [PUBMED Abstract]
Chew W, Benson C, Thway K, et al.: Clinical Characteristics and efficacy of chemotherapy in sclerosing epithelioid fibrosarcoma. Med Oncol 35 (11): 138, 2018. [PUBMED Abstract]
Treatment of Skeletal Muscle Tumors
Skeletal muscle tumors have several subtypes, including the following:
Ectomesenchymoma is a rare skeletal muscle tumor that mainly occurs in children. It is a biphenotypic soft tissue sarcoma with both mesenchymal and ectodermal components.
A single-institution retrospective review identified seven cases of malignant ectomesenchymoma.[1] All seven patients were male, with a mean age of 7.5 months (range, 0.6–17.0 months). Five of the seven patients in this series were healthy and free of disease at the time of reporting.
Histological features and genomic alterations
A retrospective review of six patients with malignant ectomesenchymoma from a single institution identified rhabdomyosarcoma as the mesenchymal element in five of six tumors.[2] Tumors with an alveolar rhabdomyosarcoma morphology exhibited the characteristic translocations, including translocation of the FOXO1 gene fusing with the PAX3 or PAX7 gene. No unifying molecular aberrations were identified.
A single-institution retrospective review identified seven cases of malignant ectomesenchymoma.[1] Most patients showed elements of embryonal rhabdomyosarcoma. The mixed neuroectodermal elements were scattered ganglion cells, ganglioneuroma, or ganglioneuroblastoma. Six of seven cases had HRAS variants. The trimethylation at lysine 27 of histone H3 (H3K27me3), typically lost in malignant peripheral nerve sheath tumor, was retained in all cases.
Treatment of ectomesenchymoma
Treatment options for ectomesenchymoma include the following:
Surgery.
Chemotherapy.
Radiation therapy.
The Cooperative Weichteilsarkom Studiengruppe (CWS) reported on six patients (aged 0.2–13.5 years) registered over 14 years.[3][Level of evidence C1] The tumors were located in various sites, including the extremities, abdomen, and orbit. All six patients were treated with surgery and chemotherapy directed at rhabdomyosarcoma. Two patients received radiation therapy. Three patients experienced tumor recurrences with rhabdomyosarcoma features. Although data are scant, it appears that the tumor may respond to chemotherapy.[3]
The European paediatric Soft Tissue Sarcoma Study Group (EpSSG) identified ten patients with ectomesenchymoma in a prospectively recorded database.[4] Of the ten cases, seven had an initial local diagnosis of rhabdomyosarcoma. All patients received chemotherapy according to rhabdomyosarcoma strategy, and four patients received radiation therapy. Overall, six patients were alive in first remission, two in second remission, and one after treatment for a second primary cancer. Only the patient with a metastatic tumor at diagnosis died of their disease.
References
Huang SC, Alaggio R, Sung YS, et al.: Frequent HRAS Mutations in Malignant Ectomesenchymoma: Overlapping Genetic Abnormalities With Embryonal Rhabdomyosarcoma. Am J Surg Pathol 40 (7): 876-85, 2016. [PUBMED Abstract]
Griffin BB, Chou PM, George D, et al.: Malignant Ectomesenchymoma: Series Analysis of a Histologically and Genetically Heterogeneous Tumor. Int J Surg Pathol 26 (3): 200-212, 2018. [PUBMED Abstract]
Dantonello TM, Leuschner I, Vokuhl C, et al.: Malignant ectomesenchymoma in children and adolescents: report from the Cooperative Weichteilsarkom Studiengruppe (CWS). Pediatr Blood Cancer 60 (2): 224-9, 2013. [PUBMED Abstract]
Milano GM, Orbach D, Casanova M, et al.: Malignant ectomesenchymoma in children: The European pediatric Soft tissue sarcoma Study Group experience. Pediatr Blood Cancer 70 (2): e30116, 2023. [PUBMED Abstract]
Treatment of Smooth Muscle Tumors
Leiomyosarcoma Not Otherwise Specified (NOS)
Leiomyosarcoma accounts for 2% of soft tissue sarcomas in patients younger than 20 years (see Table 1).
Risk factors
Among 43 children with HIV/AIDS who developed tumors, 8 developed Epstein-Barr virus–associated leiomyosarcoma.[1] Survivors of hereditary retinoblastoma have a statistically significant increased risk of developing leiomyosarcoma, and 78% of these patients were diagnosed 30 or more years after the initial diagnosis of retinoblastoma.[2]
Treatment of leiomyosarcoma
There are no standard treatment options for leiomyosarcoma in pediatric patients.
Trabectedin, an alkylating drug with multiple mechanisms of action that damage DNA, has been studied in adults with leiomyosarcoma. There are no studies using trabectedin to treat leiomyosarcoma in pediatric patients.
Results from studies in adult patients include the following:
In an open-label study of trabectedin in adult patients with recurrent sarcomas, the best overall response rate (complete remission and partial remission) was seen in patients with leiomyosarcoma (7.5%).[3] The clinical benefit rate (included stable disease) was 54% for patients with leiomyosarcoma.
In another adult study, patients with recurrent liposarcoma and leiomyosarcoma were randomly assigned to receive treatment with either trabectedin or dacarbazine. Patients treated with trabectedin had a 45% reduction in disease progression.[4]
The French Sarcoma group conducted a randomized trial for the treatment of adult patients with leiomyosarcoma (age range, 52–69 years).[5] Patients were randomly assigned to receive either single-agent doxorubicin (six cycles) or doxorubicin with trabectedin, with continued maintenance therapy using trabectedin for patients in the doxorubicin/trabectedin group who did not have disease progression. Surgery to resect residual disease was allowed in each group after six cycles of therapy. The median overall survival was longer in the doxorubicin/trabectedin group (33 months; 95% confidence interval [CI], 26–48) than in the doxorubicin group (24 months; 95% CI, 19–31). The adjusted hazard ratio for death was 0.65 (95% CI, 0.44–0.95).
References
Pollock BH, Jenson HB, Leach CT, et al.: Risk factors for pediatric human immunodeficiency virus-related malignancy. JAMA 289 (18): 2393-9, 2003. [PUBMED Abstract]
Kleinerman RA, Tucker MA, Abramson DH, et al.: Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst 99 (1): 24-31, 2007. [PUBMED Abstract]
Samuels BL, Chawla S, Patel S, et al.: Clinical outcomes and safety with trabectedin therapy in patients with advanced soft tissue sarcomas following failure of prior chemotherapy: results of a worldwide expanded access program study. Ann Oncol 24 (6): 1703-9, 2013. [PUBMED Abstract]
Demetri GD, von Mehren M, Jones RL, et al.: Efficacy and Safety of Trabectedin or Dacarbazine for Metastatic Liposarcoma or Leiomyosarcoma After Failure of Conventional Chemotherapy: Results of a Phase III Randomized Multicenter Clinical Trial. J Clin Oncol 34 (8): 786-93, 2016. [PUBMED Abstract]
Pautier P, Italiano A, Piperno-Neumann S, et al.: Doxorubicin-Trabectedin with Trabectedin Maintenance in Leiomyosarcoma. N Engl J Med 391 (9): 789-799, 2024. [PUBMED Abstract]
Treatment of So-Called Fibrohistiocytic Tumors
Plexiform Fibrohistiocytic Tumor
Plexiform fibrohistiocytic tumor is a rare, low- to intermediate-grade so-called fibrohistiocytic tumor that most commonly affects children and young adults. The median age at presentation ranges from 8 to 14.5 years. However, the tumor has been described in patients as young as 3 months.[1,2]
Clinical presentation
The tumor commonly arises as a painless mass in the skin or subcutaneous tissue and most often involves the upper extremities, including the fingers, hand, and wrist.[3–5] Plexiform fibrohistiocytic tumor is an intermediate-grade tumor that rarely metastasizes. However, there are rare reports of the tumor spreading to regional lymph nodes or the lungs.[1,5,6]
Genomic alterations
No consistent chromosomal anomalies have been detected, but a t(4;15)(q21;q15) translocation has been reported.[7]
Treatment of plexiform fibrohistiocytic tumor
Treatment options for plexiform fibrohistiocytic tumor include the following:
Surgery.
Surgery is the treatment of choice, but local recurrence has been reported in 12% to 50% of cases.[8]
References
Enzinger FM, Zhang RY: Plexiform fibrohistiocytic tumor presenting in children and young adults. An analysis of 65 cases. Am J Surg Pathol 12 (11): 818-26, 1988. [PUBMED Abstract]
Black J, Coffin CM, Dehner LP: Fibrohistiocytic tumors and related neoplasms in children and adolescents. Pediatr Dev Pathol 15 (1 Suppl): 181-210, 2012. [PUBMED Abstract]
Moosavi C, Jha P, Fanburg-Smith JC: An update on plexiform fibrohistiocytic tumor and addition of 66 new cases from the Armed Forces Institute of Pathology, in honor of Franz M. Enzinger, MD. Ann Diagn Pathol 11 (5): 313-9, 2007. [PUBMED Abstract]
Billings SD, Folpe AL: Cutaneous and subcutaneous fibrohistiocytic tumors of intermediate malignancy: an update. Am J Dermatopathol 26 (2): 141-55, 2004. [PUBMED Abstract]
Remstein ED, Arndt CA, Nascimento AG: Plexiform fibrohistiocytic tumor: clinicopathologic analysis of 22 cases. Am J Surg Pathol 23 (6): 662-70, 1999. [PUBMED Abstract]
Salomao DR, Nascimento AG: Plexiform fibrohistiocytic tumor with systemic metastases: a case report. Am J Surg Pathol 21 (4): 469-76, 1997. [PUBMED Abstract]
Redlich GC, Montgomery KD, Allgood GA, et al.: Plexiform fibrohistiocytic tumor with a clonal cytogenetic anomaly. Cancer Genet Cytogenet 108 (2): 141-3, 1999. [PUBMED Abstract]
Malignant Peripheral Nerve Sheath Tumor (MPNST) NOS
MPNSTs account for 5% of soft tissue sarcomas in patients younger than 20 years (see Table 1).
Risk factors
MPNST can arise sporadically and in children with neurofibromatosis type 1 (NF1).[1] Among patients with NF1, a family history of MPNST is associated with an increased risk of developing early-onset MPNST.[2]
A rare case of a child with documented neurofibromatosis type 2 (NF2) and a benign neurofibroma had five recurrences of disease. During this time, the lesions progressively lost markers (such as S-100) and acquired clear-cut signs of malignant transformation to MPNST, documented by multiple markers, including the first example of NOTCH2 in this disease.[3]
Histological features, diagnostic evaluation, and genomic alterations
The molecular pathogenesis of adult MPNSTs demonstrates inactivating variants in at least three pathways, including NF1, CDKN2A, CDKN2B, and PRC2 complex core components. Similar alterations have been reported in pediatric tumors.[4]
Inactivating variants of SUZ12 have been described in these tumors and are absent in neurofibromas.[5]
A DNA methylation array for methylome-based and profile-based chromosomal characterization was performed on 171 peripheral nerve sheath tumors.[6] Atypical neurofibromas and low-grade MPNSTs were indistinguishable, with a common methylation profile and loss of CDKN2A. Epigenomic analysis identified two groups of conventional high-grade MPNSTs sharing a frequent loss of neurofibromin. The larger group showed an additional loss of trimethylation of H3K27me3. The smaller group of patients with predominantly spinal primary sites showed retention of H3K27me3.
Genomic profiling was performed on 201 MPNSTs.[7] Thirteen of 201 tumors demonstrated BRAF alterations.
The Memorial Sloan Kettering Cancer Center studied archival and consultation material from 64 pediatric and young adult patients (aged 20 years or younger).[4] Fifty-eight percent of patients had a clinical history of NF1. All but one patient had high-grade MPNSTs. Overall, 89% of patients were classified as having high-grade MPNSTs, and 94% of patients had conventional histological features. There were 16 high-grade tumors available for molecular characterization using the MSK-IMPACT assay. These pediatric and adolescent tumors had genomic driver events that were similar to those in adult tumors. The study found genomic perturbations in PRC2 components (SUZ12 or EED; 9 cases), NF1 variants (8 cases), and CDKN2A and CDKN2B deletions (8 cases). Loss of HDK27me3 expression was noted in 82% of conventional high-grade MPNSTs. This finding is a potentially powerful immunohistochemical diagnostic marker for pediatric MPNSTs.
Prognostic factors and prognosis
Factors associated with a favorable prognosis include the following:[1,8–10]
Smaller tumor size. In a multivariate analysis, only tumor size and nuclear TP53 expression were found to be independent predictors of disease-specific survival.[9]
Factors associated with an unfavorable prognosis include the following:[12]
High grade.
Deep tumor location.
Locally advanced stage at diagnosis.
Presence of metastasis at diagnosis. A retrospective review of 140 patients with MPNST from the MD Anderson Cancer Center included children and adolescents. The disease-specific survival at 10 years was 32%. In this series, presence of metastatic disease was associated with a much worse prognosis.[9]
Macroscopically incomplete resection (R2).
Inactivation of TP53, either by variant or amplification of MDM2.[13]
High expression of TP53 and cyclin D1. These markers were identified as adverse prognostic factors using immunohistochemical staining of diagnostic biopsy tissue.[14][Level of evidence C2]
Presence of NF1 appears to be associated with an unfavorable prognosis, but the data are mixed.[4,15]
For patients with localized disease in the MD Anderson Cancer Center study, there was no significant difference in outcome between patients with and without NF1.[9] In other studies, it was not clear whether the absence of NF1 was a favorable prognostic factor as it has been associated with both favorable [8] and unfavorable outcomes.[1,8,10]
In the French Sarcoma Group study, NF1 was associated with other adverse prognostic features, but was not an independent predictor of poor outcome.[12] A retrospective analysis of cancer registry data from the Netherlands identified 784 patients with MPNST; 70 of the patients were aged 18 years or younger.[16][Level of evidence C1] In children with NF1, large tumor size was more common (>5 cm, 92.3% vs. 59.1%). Overall, the estimated 5-year survival rate was 52.4% (standard error [SE], 10.1%) for patients with localized MPNST and NF1, compared with 75.8% (SE, 7.1%) for patients without NF1.
The Cooperative Weichteilsarkom Studiengruppe (CWS) reported a retrospective review of patients with MPNST who were treated on five consecutive CWS trials.[17] A total of 159 patients were analyzed. NF1 was reported in 38 patients (24%). Nodal involvement was documented in 15 patients (9%) at diagnosis, and distant metastases was noted in 15 patients (9%) at diagnosis. Overall, the event-free survival (EFS) rate was 40.5% at 5 years and 36.3% at 10 years. The overall survival (OS) rate was 54.6% at 5 years and 47.1% at 10 years. Older age, positive NF1 status, primary tumor site other than extremity, larger tumor size, higher Intergroup Rhabdomyosarcoma Study (IRS) group, presence of metastatic disease, and failure to achieve first complete remission were identified as adverse prognostic factors for EFS and/or OS in the univariate analysis.
Treatment of MPNST
Treatment options for MPNST include the following:
Complete surgical removal of the tumor, whenever possible, is the mainstay of treatment.
The role of radiation therapy is difficult to assess, but durable local control of known postoperative microscopic residual tumor is not ensured after radiation therapy.
Chemotherapy
Chemotherapy has achieved objective responses in childhood MPNST.
Evidence (chemotherapy):
A large retrospective analysis of the German and Italian experience with this tumor reported the following results:[1]
Sixty-five percent of measurable tumors had objective responses to ifosfamide-containing chemotherapy regimens.
The analysis did not conclusively demonstrate improved survival with chemotherapy.
This retrospective analysis also noted a trend toward improved outcome with postoperative radiation therapy.
A series of 37 young patients with MPNST and NF1 showed that most patients had large invasive tumors that were poorly responsive to chemotherapy.[20]
The progression-free survival (PFS) rate was 19%, and the 5-year OS rate was 28%.
The European paediatric Soft Tissue Sarcoma Study Group (EpSSG) performed a prospective study in patients aged 21 years and younger with MPNST.[21] Surgical resection of primary tumors was classified as R0 if the resection was complete with negative microscopic margins, R1 if the margins were microscopically positive, and R2 if the resection left macroscopic residual tumor. Patients were nonrandomly assigned to one of the following four treatment groups:
Cohort 1: Patients with completely resected tumors (R0) who received surgery only (n = 13); the 5-year EFS rate was 92%.
Cohort 2: Patients with incompletely resected tumors (R1/R2) who received adjuvant radiation therapy (n = 4); the 5-year EFS rate was 33%.
Cohort 3: Patients with incompletely resected tumors (R1/R2) who received adjuvant chemotherapy (n = 7); the 5-year EFS rate was 29%.
Cohort 4: Patients who received either chemotherapy before surgical resection and/or who had nodal involvement (n = 27); the 5-year EFS rate was 52%.
For patients who received chemotherapy, treatment consisted of four courses of ifosfamide/doxorubicin and two courses of ifosfamide concomitant with radiation therapy (50.4–54 Gy).
The response rate to chemotherapy (partial response + complete response) in patients with measurable disease was 46%.
The presence of NF1 (51% of patients) was an independent poor prognostic factor for OS and EFS.
In a study of pediatric and adult patients with either sporadic (n = 14) or chemotherapy-naïve, NF1-associated (n = 34) MPNST, patients were treated with two cycles of ifosfamide and doxorubicin and two cycles of ifosfamide and etoposide.[22]
Response rates were lower in patients with NF1-associated tumors than in patients with sporadic tumors (17.9% vs. 44.4%). However, the premature closure of the study did not allow sufficient power to detect meaningful differences in objective responses between the two groups.
The rates of stable disease were similar between the two groups.
Recurrent MPNST
Of 120 patients enrolled in Italian pediatric protocols from 1979 to 2004, an analysis identified 73 patients younger than 21 years with relapsed MPNST. Treatment options included surgery, radiation therapy, and chemotherapy.[23]
The time to relapse from initial diagnosis ranged from 1 month to 204 months, with a median time to relapse of 7 months.
Median OS from first relapse was 11 months, with an OS rate of 39% at 1 year and 16% at 5 years.
The factors associated with a higher probability of survival after relapse were lower tumor invasiveness at initial presentation, longer time to relapse, and complete surgical resection of the tumor at relapse.
A retrospective study evaluated nine patients with unresectable or metastatic MPNST (seven patients were previously treated) who were treated with selinexor with or without doxorubicin. Three patients experienced a partial response that lasted for 3 months to longer than 8 months, and four patients had stable disease.[24]
Treatment options under clinical evaluation
Information about National Cancer Institute (NCI)-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
The following is an example of a national and/or institutional clinical trial that is currently being conducted:
NCT04465643 (Neoadjuvant Nivolumab Plus Ipilimumab for Newly Diagnosed Malignant Peripheral Nerve Sheath Tumor): The purpose of the study is to evaluate the safety and feasibility of neoadjuvant nivolumab plus ipilimumab before standard therapy (surgery, chemotherapy, or radiation therapy) in patients with NF1 and newly diagnosed premalignant and malignant peripheral nerve sheath tumors for whom surgery for resection of tumor is indicated.
Malignant Triton Tumor
Malignant triton tumors are now classified as a variant of MPNSTs. They occur most often in patients with NF1 and consist of neurogenic and rhabdomyoblastic components.[25] Most malignant triton tumors are reported in adults, although they may also arise in children and adolescents.[26]
Distinguishing between malignant triton tumors and NF1-altered rhabdomyosarcomas can be difficult. The genomic characteristics of malignant triton tumors can aid in differentiating between the two tumors. CDKN2A deep deletions and loss-of-function alterations in genes of the PRC2 complex (e.g., SUZ12 and EED1) are commonly observed in malignant triton tumors, while they are uncommon in NF1-altered rhabdomyosarcomas. The loss of PRC2 function leads to loss of H3K27me3 expression, a finding that is common in malignant triton tumors. H3K27me3 expression is generally maintained in rhabdomyosarcomas.[26–28]
References
Carli M, Ferrari A, Mattke A, et al.: Pediatric malignant peripheral nerve sheath tumor: the Italian and German soft tissue sarcoma cooperative group. J Clin Oncol 23 (33): 8422-30, 2005. [PUBMED Abstract]
Malbari F, Spira M, B Knight P, et al.: Malignant Peripheral Nerve Sheath Tumors in Neurofibromatosis: Impact of Family History. J Pediatr Hematol Oncol 40 (6): e359-e363, 2018. [PUBMED Abstract]
Agresta L, Salloum R, Hummel TR, et al.: Malignant peripheral nerve sheath tumor: Transformation in a patient with neurofibromatosis type 2. Pediatr Blood Cancer 66 (2): e27520, 2019. [PUBMED Abstract]
Agaram NP, Wexler LH, Chi P, et al.: Malignant peripheral nerve sheath tumor in children: A clinicopathologic and molecular study with parallels to the adult counterpart. Genes Chromosomes Cancer 62 (3): 131-138, 2023. [PUBMED Abstract]
Zhang M, Wang Y, Jones S, et al.: Somatic mutations of SUZ12 in malignant peripheral nerve sheath tumors. Nat Genet 46 (11): 1170-2, 2014. [PUBMED Abstract]
Röhrich M, Koelsche C, Schrimpf D, et al.: Methylation-based classification of benign and malignant peripheral nerve sheath tumors. Acta Neuropathol 131 (6): 877-87, 2016. [PUBMED Abstract]
Kaplan HG, Rostad S, Ross JS, et al.: Genomic Profiling in Patients With Malignant Peripheral Nerve Sheath Tumors Reveals Multiple Pathways With Targetable Mutations. J Natl Compr Canc Netw 16 (8): 967-974, 2018. [PUBMED Abstract]
Hagel C, Zils U, Peiper M, et al.: Histopathology and clinical outcome of NF1-associated vs. sporadic malignant peripheral nerve sheath tumors. J Neurooncol 82 (2): 187-92, 2007. [PUBMED Abstract]
Zou C, Smith KD, Liu J, et al.: Clinical, pathological, and molecular variables predictive of malignant peripheral nerve sheath tumor outcome. Ann Surg 249 (6): 1014-22, 2009. [PUBMED Abstract]
Okada K, Hasegawa T, Tajino T, et al.: Clinical relevance of pathological grades of malignant peripheral nerve sheath tumor: a multi-institution TMTS study of 56 cases in Northern Japan. Ann Surg Oncol 14 (2): 597-604, 2007. [PUBMED Abstract]
Amirian ES, Goodman JC, New P, et al.: Pediatric and adult malignant peripheral nerve sheath tumors: an analysis of data from the surveillance, epidemiology, and end results program. J Neurooncol 116 (3): 609-16, 2014. [PUBMED Abstract]
Valentin T, Le Cesne A, Ray-Coquard I, et al.: Management and prognosis of malignant peripheral nerve sheath tumors: The experience of the French Sarcoma Group (GSF-GETO). Eur J Cancer 56: 77-84, 2016. [PUBMED Abstract]
Høland M, Kolberg M, Danielsen SA, et al.: Inferior survival for patients with malignant peripheral nerve sheath tumors defined by aberrant TP53. Mod Pathol 31 (11): 1694-1707, 2018. [PUBMED Abstract]
Krawczyk MA, Karpinsky G, Izycka-Swieszewska E, et al.: Immunohistochemical assessment of cyclin D1 and p53 is associated with survival in childhood malignant peripheral nerve sheath tumor. Cancer Biomark 24 (3): 351-361, 2019. [PUBMED Abstract]
Akshintala S, Mallory NC, Lu Y, et al.: Outcome of Patients With Malignant Peripheral Nerve Sheath Tumors Enrolled on Sarcoma Alliance for Research Through Collaboration (SARC) Phase II Trials. Oncologist 28 (5): 453-459, 2023. [PUBMED Abstract]
Martin E, Coert JH, Flucke UE, et al.: Neurofibromatosis-associated malignant peripheral nerve sheath tumors in children have a worse prognosis: A nationwide cohort study. Pediatr Blood Cancer 67 (4): e28138, 2020. [PUBMED Abstract]
Meister MT, Scheer M, Hallmen E, et al.: Malignant peripheral nerve sheath tumors in children, adolescents, and young adults: Treatment results of five Cooperative Weichteilsarkom Studiengruppe (CWS) trials and one registry. J Surg Oncol 122 (7): 1337-1347, 2020. [PUBMED Abstract]
Bahig H, Roberge D, Bosch W, et al.: Agreement among RTOG sarcoma radiation oncologists in contouring suspicious peritumoral edema for preoperative radiation therapy of soft tissue sarcoma of the extremity. Int J Radiat Oncol Biol Phys 86 (2): 298-303, 2013. [PUBMED Abstract]
Baldini EH, Wang D, Haas RL, et al.: Treatment Guidelines for Preoperative Radiation Therapy for Retroperitoneal Sarcoma: Preliminary Consensus of an International Expert Panel. Int J Radiat Oncol Biol Phys 92 (3): 602-12, 2015. [PUBMED Abstract]
Ferrari A, Bisogno G, Macaluso A, et al.: Soft-tissue sarcomas in children and adolescents with neurofibromatosis type 1. Cancer 109 (7): 1406-12, 2007. [PUBMED Abstract]
van Noesel MM, Orbach D, Brennan B, et al.: Outcome and prognostic factors in pediatric malignant peripheral nerve sheath tumors: An analysis of the European Pediatric Soft Tissue Sarcoma Group (EpSSG) NRSTS-2005 prospective study. Pediatr Blood Cancer 66 (10): e27833, 2019. [PUBMED Abstract]
Higham CS, Steinberg SM, Dombi E, et al.: SARC006: Phase II Trial of Chemotherapy in Sporadic and Neurofibromatosis Type 1 Associated Chemotherapy-Naive Malignant Peripheral Nerve Sheath Tumors. Sarcoma 2017: 8685638, 2017. [PUBMED Abstract]
Bergamaschi L, Bisogno G, Manzitti C, et al.: Salvage rates and prognostic factors after relapse in children and adolescents with malignant peripheral nerve sheath tumors. Pediatr Blood Cancer 65 (2): , 2018. [PUBMED Abstract]
Al-Ezzi E, Gounder M, Watson G, et al.: Selinexor, a First in Class, Nuclear Export Inhibitor for the Treatment of Advanced Malignant Peripheral Nerve Sheath Tumor. Oncologist 26 (4): e710-e714, 2021. [PUBMED Abstract]
WHO Classification of Tumours Editorial Board: WHO Classification of Tumours. Volume 3: Soft Tissue and Bone Tumours. 5th ed., IARC Press, 2020.
de Traux de Wardin H, Dermawan JK, Vanoli F, et al.: NF1-Driven Rhabdomyosarcoma Phenotypes: A Comparative Clinical and Molecular Study of NF1-Mutant Rhabdomyosarcoma and NF1-Associated Malignant Triton Tumor. JCO Precis Oncol 8: e2300597, 2024. [PUBMED Abstract]
Schaefer IM, Fletcher CD, Hornick JL: Loss of H3K27 trimethylation distinguishes malignant peripheral nerve sheath tumors from histologic mimics. Mod Pathol 29 (1): 4-13, 2016. [PUBMED Abstract]
Infantile hemangiopericytoma, a subtype of myopericytoma, is a highly vascularized tumor of uncertain origin.
For children with hemangiopericytomas, those younger than 1 year seem to have a better prognosis than children older than 1 year.[1–3]
Histology
Histologically, hemangiopericytomas are composed of packed round or fusiform cells that are arranged around a complex vasculature, forming many branch-like structures. Hyalinization is often present. Infantile hemangiopericytomas have similar histology but many are multilobular with vasculature outside the tumor mass.[4]
Treatment and outcome of infantile hemangiopericytomas
Treatment options for infantile hemangiopericytomas include the following:
Chemotherapy.
Evidence (chemotherapy):
In a series of 17 children, the differences in metastatic potential and response to treatment were clearly demonstrated for adult and infantile hemangiopericytomas. Eleven children were older than 1 year. Several of these patients had disease in the lymph nodes or lungs.[5]
Three patients with stage I disease survived, although one patient had recurrence in the lungs.
Eight patients had stage II or stage III disease. Two of these patients survived and six patients had disease progression and died.
Six patients had infantile hemangiopericytoma, five of which were greater than stage I. All six patients survived, and three patients had good responses to vincristine, actinomycin, and cyclophosphamide.
Several studies have reported on soft tissue sarcomas in children that were more akin to infantile myofibromatosis or hemangiopericytoma.[6,7] Rather than the ETV6::NTRK3 fusion protein seen in congenital infantile fibrosarcoma, a LMNA::NTRK1 fusion protein was identified.[8] One patient carrying this fusion responded to crizotinib. For more information about infantile myofibromatosis, see the Infantile Myofibromatosis section.
Infantile Myofibromatosis
Infantile myofibromatosis is a fibrous tumor of infancy and childhood that most commonly presents in the first 2 years of life.[9]
The lesion can present as a single subcutaneous nodule (myofibroma) most commonly involving the head and neck region, or lesions can affect multiple skin areas, muscle, and bone (myofibromatosis).[10–13]
Genomic alterations and genetic testing
Somatic gain-of-function PDGFRB variants have been identified in sporadic cases of infantile myofibromatosis, including activating single nucleotide variants and in-frame indels and duplications.[14,15] PDGFRB variants are observed in most cases with multicentric nodules, but are less common in cases with solitary myofibroma.[15,16] Some PDGFRB variants that cause infantile myofibromatosis are sensitive to tyrosine kinase inhibitors like imatinib.[15,16]
An inherited autosomal dominant form of infantile myofibromatosis has been described. It is associated with germline pathogenic variants of the PDGFRB gene, with the R561C variant being most commonly observed.[17–19] The R561C variant is a relatively weak activator of PDGFRB, which may explain the presence of additional PDGFRB variants with stronger activity in some familial infantile myofibromatosis cases.[16,17]
The European Society for Paediatric Oncology Host Genome Working Group developed counseling and germline testing guidelines for these groups of children. This group recommends germline analysis for children with infantile myofibromatosis who have at least one of the following criteria:[20]
Multicentric disease.
First- or second-degree relatives with infantile myofibromatosis or soft tissue nodules during childhood.
A known PDGFRB germline pathogenic variant in the family.
Treatment and outcome of infantile myofibromatosis
Patients with these tumors usually have an excellent prognosis and the tumors can regress spontaneously. However, about one-third of cases with multicentric involvement will also have visceral involvement, and the prognosis for these patients is poor.[12,13,21]
Treatment options for infantile myofibromatosis include the following:
Observation.
Chemotherapy.
Tyrosine kinase inhibitors effective against PDGFRB.
Ninety-five patients were prospectively enrolled in five Cooperative Weichteilsarkom Studiengruppe (CWS) trials and one registry trial between 1981 and 2016.[22] Localized disease was diagnosed in 71 patients. Forty-two (59%) of these patients were infants younger than 12 months. The mainstay of treatment (applied to 55 children) was watch and wait after initial biopsy or resection. Systemic therapy was only recommended in cases of life-threatening progressive disease or in cases of compression of vital structures or organ dysfunction in the setting of progressive disease. Based on these criteria, chemotherapy was administered to 16 of 71 patients as an individual decision at the treating center: 8 patients received methotrexate/vinblastine, 5 patients received vincristine/dactinomycin/cyclophosphamide (VAC), and 3 patients received other therapies.
Of the patients who received chemotherapy, nine could be assessed for response. Two patients experienced a complete remission (CR) or partial remission (PR) (objective response rate, 22%).
Overall, 77 patients were alive in CR, and 10 patients were alive in PR. Three patients died of progressive disease.
The 5-year event-free survival (EFS) rate was 73% for patients with localized disease and 51% for patients with multifocal disease.
The 5-year overall survival (OS) rate was 95% for patients with localized or multifocal disease.
The use of combination chemotherapy with vincristine/dactinomycin and vinblastine/methotrexate have proven effective in cases of multicentric disease with visceral involvement and in cases in which the disease has progressed and has threatened the life of the patient (e.g., upper airway obstruction).[12,13,23]
Case reports have described prompt tumor regression in patients with infantile myofibromatosis that have PDGFRB variants when treated with tyrosine kinase inhibitors like imatinib and sunitinib, which inhibit the PDGFRB gain-of-function variant in the tumor.[24–27]
References
Rodriguez-Galindo C, Ramsey K, Jenkins JJ, et al.: Hemangiopericytoma in children and infants. Cancer 88 (1): 198-204, 2000. [PUBMED Abstract]
Ferrari A, Casanova M, Bisogno G, et al.: Hemangiopericytoma in pediatric ages: a report from the Italian and German Soft Tissue Sarcoma Cooperative Group. Cancer 92 (10): 2692-8, 2001. [PUBMED Abstract]
Bien E, Stachowicz-Stencel T, Godzinski J, et al.: Retrospective multi-institutional study on hemangiopericytoma in Polish children. Pediatr Int 51 (1): 19-24, 2009. [PUBMED Abstract]
Fernandez-Pineda I, Parida L, Jenkins JJ, et al.: Childhood hemangiopericytoma: review of St Jude Children’s Research Hospital. J Pediatr Hematol Oncol 33 (5): 356-9, 2011. [PUBMED Abstract]
Haller F, Knopf J, Ackermann A, et al.: Paediatric and adult soft tissue sarcomas with NTRK1 gene fusions: a subset of spindle cell sarcomas unified by a prominent myopericytic/haemangiopericytic pattern. J Pathol 238 (5): 700-10, 2016. [PUBMED Abstract]
Wong V, Pavlick D, Brennan T, et al.: Evaluation of a Congenital Infantile Fibrosarcoma by Comprehensive Genomic Profiling Reveals an LMNA-NTRK1 Gene Fusion Responsive to Crizotinib. J Natl Cancer Inst 108 (1): , 2016. [PUBMED Abstract]
Doebele RC, Davis LE, Vaishnavi A, et al.: An Oncogenic NTRK Fusion in a Patient with Soft-Tissue Sarcoma with Response to the Tropomyosin-Related Kinase Inhibitor LOXO-101. Cancer Discov 5 (10): 1049-57, 2015. [PUBMED Abstract]
Wiswell TE, Davis J, Cunningham BE, et al.: Infantile myofibromatosis: the most common fibrous tumor of infancy. J Pediatr Surg 23 (4): 315-8, 1988. [PUBMED Abstract]
Levine E, Fréneaux P, Schleiermacher G, et al.: Risk-adapted therapy for infantile myofibromatosis in children. Pediatr Blood Cancer 59 (1): 115-20, 2012. [PUBMED Abstract]
Larralde M, Hoffner MV, Boggio P, et al.: Infantile myofibromatosis: report of nine patients. Pediatr Dermatol 27 (1): 29-33, 2010 Jan-Feb. [PUBMED Abstract]
Agaimy A, Bieg M, Michal M, et al.: Recurrent Somatic PDGFRB Mutations in Sporadic Infantile/Solitary Adult Myofibromas But Not in Angioleiomyomas and Myopericytomas. Am J Surg Pathol 41 (2): 195-203, 2017. [PUBMED Abstract]
Arts FA, Sciot R, Brichard B, et al.: PDGFRB gain-of-function mutations in sporadic infantile myofibromatosis. Hum Mol Genet 26 (10): 1801-1810, 2017. [PUBMED Abstract]
Dachy G, de Krijger RR, Fraitag S, et al.: Association of PDGFRB Mutations With Pediatric Myofibroma and Myofibromatosis. JAMA Dermatol 155 (8): 946-950, 2019. [PUBMED Abstract]
Cheung YH, Gayden T, Campeau PM, et al.: A recurrent PDGFRB mutation causes familial infantile myofibromatosis. Am J Hum Genet 92 (6): 996-1000, 2013. [PUBMED Abstract]
Martignetti JA, Tian L, Li D, et al.: Mutations in PDGFRB cause autosomal-dominant infantile myofibromatosis. Am J Hum Genet 92 (6): 1001-7, 2013. [PUBMED Abstract]
Murray N, Hanna B, Graf N, et al.: The spectrum of infantile myofibromatosis includes both non-penetrance and adult recurrence. Eur J Med Genet 60 (7): 353-358, 2017. [PUBMED Abstract]
Hettmer S, Dachy G, Seitz G, et al.: Genetic testing and surveillance in infantile myofibromatosis: a report from the SIOPE Host Genome Working Group. Fam Cancer 20 (4): 327-336, 2021. [PUBMED Abstract]
Gopal M, Chahal G, Al-Rifai Z, et al.: Infantile myofibromatosis. Pediatr Surg Int 24 (3): 287-91, 2008. [PUBMED Abstract]
Sparber-Sauer M, Vokuhl C, Seitz G, et al.: Infantile myofibromatosis: Excellent prognosis but also rare fatal progressive disease. Treatment results of five Cooperative Weichteilsarkom Studiengruppe (CWS) trials and one registry. Pediatr Blood Cancer 69 (3): e29403, 2022. [PUBMED Abstract]
Weaver MS, Navid F, Huppmann A, et al.: Vincristine and Dactinomycin in Infantile Myofibromatosis With a Review of Treatment Options. J Pediatr Hematol Oncol 37 (3): 237-41, 2015. [PUBMED Abstract]
Weller JM, Keil VC, Gielen GH, et al.: PDGRFB mutation-associated myofibromatosis: Response to targeted therapy with imatinib. Am J Med Genet A 179 (9): 1895-1897, 2019. [PUBMED Abstract]
Wenger TL, Bly RA, Wu N, et al.: Activating variants in PDGFRB result in a spectrum of disorders responsive to imatinib monotherapy. Am J Med Genet A 182 (7): 1576-1591, 2020. [PUBMED Abstract]
Mudry P, Slaby O, Neradil J, et al.: Case report: rapid and durable response to PDGFR targeted therapy in a child with refractory multiple infantile myofibromatosis and a heterozygous germline mutation of the PDGFRB gene. BMC Cancer 17 (1): 119, 2017. [PUBMED Abstract]
Pond D, Arts FA, Mendelsohn NJ, et al.: A patient with germ-line gain-of-function PDGFRB p.N666H mutation and marked clinical response to imatinib. Genet Med 20 (1): 142-150, 2018. [PUBMED Abstract]
Treatment of Tumors of Uncertain Differentiation
Tumors of uncertain differentiation have many subtypes, including the following:
Carney complex is an autosomal dominant syndrome caused by variants in the PRKAR1A gene, located on chromosome 17.[1] The syndrome is characterized by cardiac and cutaneous myxomas, pale brown to brown lentigines, blue nevi, primary pigmented nodular adrenocortical disease causing Cushing syndrome, and a variety of endocrine and nonendocrine tumors, including pituitary adenomas, thyroid tumors, and large cell calcifying Sertoli cell tumor of the testis.[1–3] There are published surveillance guidelines for patients with Carney complex that include cardiac, testicular, and thyroid ultrasonography.
For patients with the Carney complex, prognosis depends on the frequency of recurrences of cardiac and skin myxomas and other tumors.
For more information about the treatment of conditions related to Carney complex, see the following summaries:
Synovial Sarcoma NOS (Poorly Differentiated, Spindle Cell, and Biphasic Varieties)
Synovial sarcoma accounts for 9% of soft tissue sarcomas in patients younger than 20 years (see Table 1).
Synovial sarcoma is one of the most common nonrhabdomyosarcomatous soft tissue sarcoma (NRSTS) in children and adolescents. In a review of the Surveillance, Epidemiology, and End Results (SEER) Program database from 1973 to 2005, 1,268 patients with synovial sarcoma were identified. Approximately 17% of these patients were children and adolescents, and the median age at diagnosis was 34 years.[4] In addition, in the Children’s Oncology Group (COG) ARST0332 (NCT00346164) and European paediatric Soft Tissue Sarcoma Study Group (EpSSG) 2005 protocols, synovial sarcoma was the single most common histological subtype.[5]
Clinical presentation
The most common primary tumor location is the extremities, followed by trunk and head and neck.[4] Rarely, a synovial sarcoma may arise in the heart or pericardium or appear with a pleuropulmonary presentation.[6–9]
The most common site of metastasis is the lung.[10,11] The risk of metastases is highly influenced by tumor size. Patients with tumors that are larger than 5 cm have an estimated 32-fold higher risk of developing metastases compared with patients who have tumors smaller than 5 cm.
The Cooperative Weichteilsarkom Studiengruppe (CWS) reported on 432 patients younger than 21 years diagnosed with synovial sarcoma between 1981 and 2018.[12] The study compared three age groups of patients: children (aged 0–12 years; n = 176), adolescents (aged 13–16 years; n = 178), and young adults (aged 17–21 years; n = 78).
The proportion of invasive tumors was significantly higher in older patients (children, 33%; adolescents, 39%; and young adults, 54%; P = .009).
The proportion of tumors larger than 10 cm (children, 13%; adolescents, 21%; and young adults, 31%; P = .006) and the presence of metastasis at first diagnosis were also higher in older patients (children, 6%; adolescents, 10%; and young adults, 21%; P = .001).
Histological features, diagnostic evaluation, and genomic alterations
Synovial sarcoma can be subclassified as the following types:
Synovial sarcoma, spindle cell.
Synovial sarcoma, biphasic.
Synovial sarcoma, poorly differentiated.
The diagnosis of synovial sarcoma is made by immunohistochemical analysis, ultrastructural findings, and demonstration of the specific chromosomal translocation t(x;18)(p11.2;q11.2). This abnormality is specific for synovial sarcoma and is found in all morphological subtypes. Synovial sarcoma results in rearrangement of the SS18 gene on chromosome 18 with one of the subtypes (1, 2, or 4) of the SSX gene on chromosome X.[13,14] It is thought that the SS18::SSX fusion transcript promotes epigenetic silencing of key tumor suppressor genes.[15]
In one report, reduced SMARCB1 nuclear reactivity on immunohistochemical staining was seen in 49 cases of synovial sarcoma, suggesting that this pattern may help distinguish synovial sarcoma from other histologies.[16]
Prognostic factors
Favorable prognostic factors
Patients younger than 10 years have more favorable outcomes and clinical features than older patients.
Favorable clinical features include the following:[4,17–19]
Extremity primary tumors.
Smaller tumors.
Localized disease.
Response to chemotherapy was correlated with improved survival in one meta-analysis.
Unfavorable prognostic factors
The following studies have reported multiple factors associated with unfavorable outcomes:
In a retrospective analysis of synovial sarcoma in children and adolescents who were treated in Germany and Italy, tumor size (>5 cm or ≤5 cm in greatest dimension) was an important predictor of event-free survival (EFS).[20] In this analysis, local invasiveness conferred an inferior probability of EFS, but surgical margins were not associated with clinical outcome.
In a single-institution retrospective analysis of 111 patients with synovial sarcoma who were younger than 22 years at diagnosis, larger tumor size, greater depth in tissue, greater local invasiveness, and more proximal tumor location were associated with poorer overall survival (OS).[21][Level of evidence C1]
A multicenter analysis included 219 children from various treating centers, including Germany, St. Jude Children’s Research Hospital (SJCRH), Instituto Tumori, and MD Anderson Cancer Center. The study reported an estimated 5-year OS rate of 80% and an EFS rate of 72%.[19] In this analysis, an interaction between tumor size and invasiveness was observed. In multivariate analysis, patients with large or invasive tumors or with Intergroup Rhabdomyosarcoma Study (IRS) group III disease (localized, incompletely resected or with biopsy only) and group IV disease (metastases at diagnosis) had decreased OS. Treatment with radiation therapy was related to improved OS (hazard ratio [HR], 0.4; 95% confidence interval [CI], 0.2–0.7). In patients with IRS group III disease, objective response to chemotherapy (18 of 30 [60%]) correlated with improved survival.
Expression and genomic index prognostic signatures have been studied in synovial sarcoma. Complex genomic profiles, with greater rearrangement of the genome, are more common in adults than in younger patients with synovial sarcoma and are associated with a higher risk of metastasis.[22]
A review of 84 patients with localized synovial sarcoma who had information on fusion status (SS18::SSX) and histological grading found no difference in OS according to these criteria. However, for tumor size at diagnosis, the study showed that patients with tumors between 5 cm and 10 cm had a worse prognosis than those with smaller tumors (P = .02). Patients with tumors larger than 10 cm had an even worse OS (P = .0003).[23][Level of evidence C1]
The German CWS group reviewed 27 evaluable patients younger than 21 years with pulmonary metastases among 296 patients with synovial sarcoma. All patients had metastasis to the lungs. The 5-year EFS rate was 26%, and the OS rate was 30%. The most important prognostic factor at presentation was that the metastases were limited to one lesion in one lung or one lesion in both lungs (a group they termed oligometastatic). Treatment elements associated with superior survival were adequate local therapy of the primary tumor and, if feasible, for the metastases. The use of whole-lung irradiation did not correlate with better outcomes.[24][Level of evidence C1]
The EpSSG designed a genomic index for synovial sarcoma.[25][Level of evidence C2] Genomic index was defined as A2/C, where A is the total number of alterations (segmental gains and losses) and C is the number of involved chromosomes on array comparative genomic hybridization results. In a multivariate analysis of 61 pediatric, adolescent, and young adult patients (aged <25 years), high genomic index was an independent predictor of decreased EFS and OS.
In adults, factors such as International Union Against Cancer/American Joint Committee on Cancer stage III and stage IVA, poor tumor necrosis, truncal location, elevated mitotic rate, older age, and higher histological grade have been associated with a worse prognosis.[26–28]
Treatment of synovial sarcoma
Treatment options for synovial sarcoma include the following:
The COG and the EpSSG reported a combined analysis of 60 patients younger than 21 years with localized synovial sarcoma prospectively assigned to surgery without adjuvant radiation therapy or chemotherapy.[31] Enrollment was limited to patients with initial complete resection with histologically free margins, with a grade 2 tumor of any size or a grade 3 tumor 5 cm or smaller.
The 3-year EFS rate was 90% (median follow-up, 5.2 years; range, 1.9–9.1 years).
All eight events were local tumor recurrence; no metastatic recurrences were seen.
All patients with recurrent disease were effectively treated with second-line therapy, resulting in an OS rate of 100%.
Therefore, the authors concluded that a surgery-only approach was optimal for patients who achieved an R0 resection (complete resection with negative microscopic margins) and had tumors smaller than 5 cm, regardless of grade.
Surgery and chemotherapy, with or without radiation therapy
Synovial sarcoma appears to be more sensitive to chemotherapy than many other soft tissue sarcomas. Children with synovial sarcoma seem to have a better prognosis than adults with synovial sarcoma.[11,28,32–37]
The most commonly used chemotherapy regimens for the treatment of synovial sarcoma incorporate ifosfamide and doxorubicin.[19,35,38] Response rates to the ifosfamide and doxorubicin regimen are higher than in other NRSTS.[39]
Evidence (surgery and chemotherapy with or without radiation therapy):
Several treatment centers advocate chemotherapy after resection and radiation therapy for children and young adults with synovial sarcoma.[19,20,40–42]
The International Society of Pediatric Oncology-Malignant Mesenchymal Tumors studies showed that select patients (young age, <5 cm resected tumors) with nonmetastatic synovial sarcoma treated with chemotherapy can have excellent outcomes in the absence of radiation therapy. However, it is still unclear whether that approach obviates an advantage of radiation for local or regional control.[41]
A German trial suggested a benefit for postoperative chemotherapy in children with synovial sarcoma.[42]
A meta-analysis also suggested that chemotherapy may provide benefit.[19]
The COG reported an analysis of the subset of patients with synovial sarcoma treated on the ARST0332 (NCT00346164) trial. This was a prospective treatment assignment trial for patients younger than 30 years with NRSTS.[43] They analyzed the outcomes of 138 eligible patients.
Overall, R0 resection or R1 resection (microscopically positive margins) of the primary tumor was achieved in 129 patients (93.5%): 69 patients (53.5%) at study entry and 60 patients (46.5%) after neoadjuvant chemotherapy. Of these, 104 patients (80.6%) had an R0 resection: 55 patients (53%) at study entry and 49 patients (47%) after neoadjuvant chemotherapy.
In the 60 patients who received neoadjuvant chemotherapy, response was evaluable in 55 patients. Two patients (3.6%) had complete responses, 9 (16.4%) had partial responses, 41 (74.6%) had stable disease, and 3 (5.5%) had progressive disease. The tissue from 57 tumors was centrally reviewed after definitive resection. Forty-one tumors (72%) had less than 90% necrosis, and 16 tumors (28%) had 90% necrosis or more.
The study prospectively defined three risk groups:
Low risk (about 50% of population): Patients with nonmetastatic, grossly resected tumors, except patients who had tumors that were both high grade and >5 cm in maximal diameter.
Intermediate risk (about 35% of population): Patients with nonmetastatic tumors that were both high grade and >5 cm in maximal diameter and patients with nonmetastatic, nonresectable tumors regardless of grade and size.
High risk (about 15% of population): Patients with metastatic tumors, including those with metastases restricted to regional lymph nodes.
For the 46 patients in the low-risk group, the 5-year EFS rate was 81.9% (95% CI, 69%–94.8%), and the OS rate was 97.7% (95% CI, 92.7%–100%).
For the 23 patients in the intermediate-risk group (treatment arm C), the 5-year EFS rate was 64% (95% CI, 42.4%–85.8%), and the OS rate was 89.5% (95% CI, 75.3%–100%).
For the 49 patients in the intermediate-risk group (treatment arm D), the 5-year EFS rate was 71.2% (95% CI, 56.5%–85.9%), and the OS rate was 86.5% (95% CI, 75.6%–97.3%).
For the 21 patients in the high-risk group, the 5-year EFS rate was 7.6% (95% CI, 0%–22%), and the OS rate was 12.5% (95% CI, 0%–28.7%).
The EpSSG performed a prospective study of patients younger than 21 years with synovial sarcoma (CCLG-EPSSG-NRSTS-2005 [NCT00334854]).[44][Level of evidence C1] Patients were stratified into the following three risk groups and nonrandomly assigned to treatment by risk group:
Low-risk patients had IRS group I tumors less than 5 cm in size and nonaxial primary tumors.
Intermediate-risk patients had no axial primary tumors and IRS group I tumors greater than 5 cm or IRS group II tumors.
High-risk patients included all patients with axial primary sites (head and neck, lung and pleura, trunk, retroperitoneal), IRS group III tumors, or N1 tumors.
Outcomes for patients treated on the CCLG-EPSSG-NRSTS-2005 trial are described in Table 12.
Table 12. Event-Free Survival (EFS) and Overall Survival (OS) in Patients With Low-, Intermediate-, and High-Risk Synovial Sarcoma Treated on the CCLG-EPSSG-NRSTS-2005 Trial
aChemotherapy was ifosfamide/doxorubicin, with doxorubicin omitted during radiation therapy.
b59.4 Gy in cases without the option of secondary resection; 50.4 Gy as preoperative radiation therapy; 50.4, 54, and 59.4 Gy as postoperative radiation therapy, in the case of R0, R1, and R2 resections, respectively (no additional radiation therapy in the case of secondary complete resections with free margins, in children younger than 6 years).
Low
Surgery alone
92
100
Intermediate
Surgery, 3–6 cycles chemotherapya, ± RTb
91
100
High (IRS group III)
3 cycles of chemotherapya, surgery, 3 additional cycles of chemotherapy, ± RTb
77
94
High (axial primary sites)
Surgery, 6 cycles of chemotherapya, RTb
78
100
The CWS reported results from a prospective trial for the treatment of patients with synovial sarcoma. Eligibility was restricted to patients with localized tumors with macroscopic residual disease after first surgery, before the initiation of systemic therapy (IRS III) and no clinically detectable metastatic disease. There were 145 patients in the study with a median age of 14.5 years (range, 0.2–33.2 years). The protocols recommended but did not require radiation therapy to be given before definitive tumor resection. Radiation therapy was administered to 115 patients (79%), and 23 patients did not receive radiation therapy (no information documented for 7 patients). Of the 115 irradiated patients, 57 were irradiated before tumor excision and 52 after tumor excision.[45]
In this nonrandomized comparison, the sequencing of radiation therapy before definitive resection was associated with a statistically significant improvement in local recurrence-free survival rates, compared with definitive surgery before radiation therapy.
Omission of radiation therapy was associated with an inferior outcome.
Table 13. Effects of Radiation Therapy Timing on Outcomes of Patients With Synovial Sarcoma
Radiation Therapy
Patients (No.)
5-Year EFS Rate
5-Year OS Rate
5-Year Local Recurrence-Free Survival Rate
EFS = event-free survival; OS = overall survival.
No radiation therapy
23
44%
57%
76%
Radiation therapy before surgery
57
70%
83%
98%
Radiation therapy after surgery
52
73%
82%
86%
Recurrent synovial sarcoma NOS
For patients with recurrent synovial sarcoma, the survival rate after relapse is poor (30%–40% at 5 years). Factors associated with outcome after relapse include duration of first remission (> or ≤ 18 months) and lack of a second remission.[46,47]
In a German experience, surgical resection of metastatic disease was the most common way to achieve a second complete remission.[47] Maintenance chemotherapy with oral trofosfamide, idarubicin, and etoposide or oral cyclophosphamide and intravenous vinblastine was administered on an individual basis.
A consortium of six European referral centers reported a retrospective review of patients younger than 21 years with recurrent synovial sarcoma. Among 41 patients, the first relapses occurred within 3 to 132 months (median, 18 months) after first diagnoses. The relapses were local in 34% of patients, metastatic in 54%, and both in 12%. Treatments at first relapse included surgery in 56% of patients, radiation therapy in 34%, and systemic therapy in 88%. In all, 36 patients received second-line medical treatment, which included chemotherapy in 32 patients (with 10 different regimens) and targeted therapy in 4 patients. No patient was included in early-phase clinical trials as second-line therapy. The overall response rate was 42%. The median EFS was 12 months, and the postrelapse 5-year EFS rate was 15.8%. The median OS was 30 months, and the postrelapse 5-year OS rate was 22.2%. In a Cox multivariable regression analysis, OS was significantly associated with time and type of relapse.[48]
Radiation therapy (stereotactic body radiation therapy) can be used to target select pulmonary metastases. This is usually considered after a minimum of one resection to confirm metastatic disease. Radiation therapy is particularly appropriate for patients with lesions that threaten air exchange because of their location adjacent to bronchi or cause pain by invading the chest wall.[49]
Between 70% to 80% of synovial sarcomas express NY-ESO-1, an immunogenic cancer testis antigen.[50] NY-ESO-1 can be targeted with adoptive transfer of T cells engineered to express NY-ESO-1c259, an affinity-enhanced T-cell receptor (TCR) targeting NY-ESO-1/LAGE1a.[51] The procedure to produce the genetically engineered T cells restricts their reactivity to a single HLA type. All clinical trials of this technology chose HLA-A*02 as the initial target and limited eligibility to patients whose tumors expressed NY-ESO-1 and who had HLA-A*02. In a multi-institutional trial, confirmed antitumor responses occurred in 50% of patients (6 of 12) and were characterized by tumor shrinkage over several months. Circulating NY-ESO-1c259 T cells were present postinfusion in all patients, and the cells persisted for at least 6 months in all responders.[52]
An open-label, international, phase II study enrolled patients with previously treated metastatic or unresectable synovial sarcoma and myxoid round cell liposarcoma.[53] Fifty-two patients with HLA-A*02 and tumors that expressed melanoma-associated antigen A4 (MAGE-A4) received a single intravenous dose of afamitresgene autoleucel (afami-cel) after lymphodepletion. Afami-cel is a MAGE-A4–directed, genetically modified, autologous T-cell immunotherapy. The overall response rate (the primary end point of the study) was 37% (19 of 52; 95% CI, 24%–51%), 39% (17 of 44; 95% CI, 24%–55%) for patients with synovial sarcoma, and 25% (2 of 8; 95% CI, 3%–65%) for patients with myxoid round cell liposarcoma. Cytokine release syndrome and cytopenias were the most frequent side effects. The authors concluded that afami-cel treatment resulted in durable responses in heavily pretreated eligible patients with synovial sarcoma.[53] These data led to FDA approval of afami-cel in adults with unresectable or metastatic synovial sarcoma who have received prior chemotherapy, are HLA-A*02 positive, and whose tumors express the MAGE-A4 antigen.
Treatment options under clinical evaluation
Information about National Cancer Institute (NCI)-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Epithelioid Sarcoma
Epithelioid sarcoma is a rare mesenchymal tumor of uncertain histogenesis that displays multilineage differentiation.[54]
Clinical presentation
Epithelioid sarcoma commonly presents as a slowly growing firm nodule based in the deep soft tissue. The proximal type predominantly affects adults and involves the axial skeleton and proximal sites. The tumor is highly aggressive and has a propensity for lymph node metastases.
Genomic alterations
Epithelioid sarcoma is characterized by inactivation of the SMARCB1 gene, which is present in both conventional and proximal types of epithelioid sarcoma.[55] This abnormality leads to increased dependence on EZH2 and tumor formation.[56]
Treatment of epithelioid sarcoma
Treatment options for epithelioid sarcoma include the following:
Surgery with or without chemotherapy and/or radiation therapy
Surgical removal of primary and recurrent tumor(s) is the most effective treatment.[57][Level of evidence C1] Because of the propensity of this disease to have occult metastasis to the lymph nodes, sentinel lymph node biopsy is recommended for epithelioid sarcoma of the extremities or buttocks in the absence of clinically (by imaging or physical examination) enlarged lymph nodes.[58]
Evidence (surgery with or without chemotherapy and/or radiation therapy):
In a German CWS retrospective analysis of 67 children, adolescents, and young adults (median age, 14 years) with epithelioid sarcoma, 53 patients presented with localized disease and 14 patients presented with metastatic disease.[59][Level of evidence C1] Fifty-eight of 67 patients were treated with primary resections. Resections were microscopically complete in 35 patients, microscopically incomplete in 12 patients, and macroscopically incomplete in 20 patients. Forty-nine patients received chemotherapy, and 33 patients received radiation therapy.
Complete remission was achieved in 45 of 53 patients (85%) with localized disease.
Twenty-seven patients with localized disease had local (n = 16), metastatic (n = 6), or combined (n = 4) relapses after a median time of 0.9 years (range, 0.1–2.3 years) after complete response of disease (45 of 63).
Patients with localized disease had a 5-year EFS rate of 35% (95% CI, ±12%) and an OS rate of 48% (95% CI, ±14%).
Patients with metastatic disease had a 5-year EFS rate of 7% (95% CI, ±14%) and an OS rate of 9% (95% CI, ±16%).
Smaller tumor size, lower IRS group, less tumor invasiveness, negative nodal status, and microscopically complete resection correlated with a favorable prognosis in patients with localized disease.
A retrospective analysis reviewed COG and EpSSG prospective clinical trials that enrolled patients younger than 30 years with epithelioid sarcoma.[60][Level of evidence B4] The analysis identified 63 patients who were treated between July 2005 and November 2015. Patients were stratified into three risk groups using a combination of clinical features and treatment received. Low-risk patients (n = 34) underwent surgery with or without radiation therapy and included predominantly patients with nonmetastatic widely or marginally resected tumors 5 cm or smaller. The intermediate-risk group included patients (n = 16) with nonmetastatic, high-grade, and larger than 5 cm tumors or unresectable tumors. Patients with nodal or distant metastatic disease were at high risk (n = 13) , regardless of tumor grade or size.
Partial responses were observed in 11 of 22 patients (50%) who received neoadjuvant therapy.
Events were local recurrence (n = 10) and distant recurrence (n = 15).
The estimated 5-year OS rates were 86.4% for low-risk patients, 63.5% for intermediate-risk patients, and 0% for high-risk patients.
Locoregional nodal involvement, invasive tumor, high grade, and lesser extent of resection predicted poorer EFS in patients without metastases.
A review of 30 pediatric patients with epithelioid sarcoma (median age at presentation, 12 years) reported the following results:[61]
Responses to chemotherapy were reported in 40% of patients using sarcoma-based treatment regimens.
Sixty percent of patients were alive at 5 years after initial diagnosis.
A single-institution retrospective review of 20 patients, which included children and adults (median age, 27.3 years), reported the following:[57]
There was no difference in the probability of recurrence between patients who received chemotherapy and those who did not receive chemotherapy.
The authors suggested that radiation therapy may be useful.
Targeted therapy
Evidence (tazemetostat):
In a phase II trial of 62 adult patients with epithelioid sarcoma and documented loss of INI1 by immunohistochemistry or biallelic SMARCB1 (the gene that encodes INI1) alterations, tazemetostat showed clinical activity.[62]
There were 9 of 62 confirmed partial responses, with an objective response rate of 15% and a disease control rate of 26%.
In January 2020, the U.S. Food and Drug Administration (FDA) granted accelerated approval to tazemetostat for adult and pediatric patients aged 16 years and older with metastatic or locally advanced epithelioid sarcoma who were not eligible for complete resection.
Treatment options under clinical evaluation for epithelioid sarcoma
Information about NCI-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
The following is an example of a national and/or institutional clinical trial that is currently being conducted:
PEPN2121 (NCT05286801) (Tiragolumab and Atezolizumab for the Treatment of Relapsed or Refractory SMARCB1– or SMARCA4-Deficient Tumors): This study is evaluating the combination of a PD-L1 targeting antibody (atezolizumab) with a TIGIT targeting antibody (tiragolumab) for patients with SMARCB1– or SMARCA4-deficient tumors. Patients with epithelioid sarcoma may be eligible for this study.
Alveolar Soft Part Sarcoma
Alveolar soft part sarcomas account for 1.4% of soft tissue sarcomas in patients younger than 20 years (see Table 1).
Clinical presentation
The median age at presentation is 25 years for patients with alveolar soft part sarcoma. This tumor most commonly arises in the extremities but can occur in the oral and maxillofacial region.[63–65] Alveolar soft part sarcoma in children can present with evidence of metastatic disease.[66] Delayed metastases to the brain and lung are uncommon.[63]
In a series of 61 patients with alveolar soft part sarcoma who were treated in four consecutive CWS trials and the Soft Tissue Sarcoma Registry (SoTiSaR), 46 patients presented with localized disease and 15 patients had evidence of metastasis at diagnosis.[67]
Sixty-nine patients younger than 30 years with alveolar soft part sarcoma were treated between 1980 and 2014 at four major institutions. The median age at diagnosis was 17 years, and 64% of patients were female. The most common site of disease was the lower extremity, and 26 patients had an ASPSCR1::TFE3 gene translocation.[68]
Genomic alterations
This tumor of uncertain histogenesis is characterized by a consistent chromosomal translocation t(X;17)(p11.2;q25) that fuses the ASPSCR1 gene with the TFE3 gene.[69,70]
Prognosis
Alveolar soft part sarcoma in children may have an indolent course.[66] Patients with alveolar soft part sarcoma may relapse several years after a prolonged period of apparent remission.[67,71]
In a series of 19 treated patients with alveolar soft part sarcoma, one study reported a 5-year OS rate of 80%. The OS rate was 91% for patients with localized disease, 100% for patients with tumors 5 cm or smaller, and 31% for patients with tumors larger than 5 cm.[72]
In another series of 33 patients, the OS rate was 68% at 5 years from diagnosis and 53% at 10 years from diagnosis. Survival was better for patients with smaller tumors (≤5 cm) and completely resected tumors.[73][Level of evidence C1]
A retrospective review of children and young adults younger than 30 years (median age, 17 years; range, 1.5–30 years) from four institutions identified 69 patients treated primarily with surgery between 1980 and 2014.[68][Level of evidence C1] The ASPSCR1::TFE3 translocation was present in all 26 patients tested. There were 19 patients with IRS group I tumors (28%), 7 patients with IRS group II tumors (10%), 5 patients with IRS group III tumors (7%), and 38 patients with IRS group IV tumors (55%). The 5-year EFS rate was 80%, and the OS rate was 87% for the 31 patients with localized tumors (IRS postsurgical groups I, II, and III). The 5-year EFS rate was 7%, and the OS rate was 61% for the 38 patients with metastatic tumors (IRS group IV).
In a series of patients treated on consecutive studies from Germany, 15 of 61 patients (25%) presented with metastases, often miliary in nature. Despite lack of response to chemotherapy, the 5-year OS rate was 61%, with an EFS rate of 20%.[67]
Treatment of alveolar soft part sarcoma
Treatment options for alveolar soft part sarcoma include the following:
Targeted therapy (tyrosine kinase inhibitors and checkpoint inhibitors).[74]
Surgery with or without radiation therapy and chemotherapy
The standard treatment approach is complete resection of the primary lesion.[72] If complete excision is not feasible, radiation therapy is administered.
Evidence (surgery with or without chemotherapy):
A study from China reported on 18 patients with alveolar soft part sarcoma of the oral and maxillofacial region. Fifteen patients were younger than 30 years. Surgical removal with negative margins was the primary treatment.[65][Level of evidence C2]
All patients survived, and only one patient had metastatic disease recurrence.
In a series of patients treated on consecutive studies from Germany, the following was reported:[67]
Progression-free survival (PFS) for patients without metastases on presentation appeared to improve with complete resection of the primary tumor.
The 5-year EFS rate was 100% for patients with completely resected tumors, compared with 50% for patients with microscopic or gross residual disease.
In a series of 51 pediatric patients aged 0 to 21 years with alveolar soft part sarcoma, the following was reported:[63][Level of evidence C1]
The OS rate was 78% at 10 years, and the EFS rate was about 63%.
Patients with localized disease (n = 37) had a 10-year OS rate of 87%.
The 14 patients with metastases at diagnosis had a 10-year OS rate of 44%, partly resulting from the surgical removal of the primary tumor and lung metastases in some patients.
Only 3 of 18 patients (17%) with measurable disease had a response to conventional antisarcoma chemotherapy, but two of four patients treated with sunitinib had a partial response.
Targeted therapy
Studies of targeted therapy (tyrosine kinase inhibitors and checkpoint inhibitors) have been done.
Sunitinib
Evidence (sunitinib):
A small retrospective study of nine adult patients with metastatic alveolar soft part sarcoma treated with sunitinib reported partial responses in five patients and stable disease in two patients.[75][Level of evidence C3]
In another study, 15 adult patients with alveolar soft part sarcoma were treated with sunitinib. Five patients were treated with sunitinib for longer than 2 years.[76][Level of evidence C1]
Six patients experienced partial responses.
The median PFS was 19 months, and the median OS was 56 months.
The 5-year OS rate was 49%.
Cediranib
Cediranib is an inhibitor of all three known vascular epidermal growth factor receptors.
Evidence (cediranib):
In a pediatric phase II trial of cediranib, using 70% of the adult maximum tolerated dose in patients younger than 16 years, the following was reported:[77][Level of evidence B4]
Five of seven patients had stable disease for 14 months or longer.
An international group performed a double-blind, placebo-controlled, randomized, phase II trial of cediranib in adolescent and adult patients with alveolar soft part sarcoma.[78][Level of evidence A1]
Median percentage change in sum of target marker lesion diameters for the evaluable population was -8.3% (interquartile range [IQR], -26.5 to 5.9) for patients who received cediranib therapy, compared with 13.4% (IQR, 1.1–21.3) for patients who received the placebo (one-sided P = .0010).
The authors concluded that cediranib is an active agent in patients with alveolar soft part sarcoma.
In a phase II trial of cediranib, 15 of 43 adult patients (35%) with metastatic alveolar soft part sarcoma had partial responses.[79][Level of evidence C3]
Pazopanib
Evidence (pazopanib):
In an open-label trial that evaluated the efficacy of pazopanib in six adult patients, one patient achieved a partial response and five patients had stable disease.[80]
Another trial included 30 adult patients who were treated with pazopanib.[81]
One patient experienced a complete response, seven patients experienced partial responses, and 17 patients had stable disease.
The median PFS was 13.6 months.
Axitinib and pembrolizumab
Axitinib is a vascular endothelial growth factor receptor tyrosine kinase inhibitor. Pembrolizumab is an anti–PD-1 immune checkpoint inhibitor.
Evidence (axitinib and pembrolizumab):
In one trial, adult patients with advanced sarcomas were treated with a combination of axitinib and pembrolizumab.[74]
For the 12 patients with alveolar soft part sarcoma, the 3-month PFS rate was 73%.
Six of eleven patients with evaluable disease had partial responses to axitinib.
Atezolizumab
Atezolizumab is a monoclonal antibody directed against PD-1 and PD-L1.
Evidence (atezolizumab):
In a phase II trial, 52 patients older than 2 years with advanced alveolar soft part sarcoma were treated with atezolizumab.[82]
Nineteen patients experienced a response, 18 had partial responses and 1 had a complete response.
Based on these data, the FDA approved the use of atezolizumab in children older than 2 years with unresectable or metastatic alveolar soft part sarcoma.
Other therapies
There have been sporadic reports of objective responses to treatment with interferon-alpha and bevacizumab.[63,83,84]
Because these tumors are rare, all children with alveolar soft part sarcoma should be considered for enrollment in prospective clinical trials. Information about ongoing clinical trials is available from the NCI website.
Treatment options under clinical evaluation for alveolar soft part sarcoma
Information about NCI-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
Clear Cell Sarcoma NOS
Clear cell sarcoma (formerly called malignant melanoma of soft parts) is a rare soft tissue sarcoma that typically involves the deep soft tissues of the extremities. It is also called clear cell sarcoma of tendons and aponeuroses. The tumor often affects adolescents and young adults.
Clinical presentation
The tumor most commonly affects the lower extremity, particularly the foot, heel, and ankle.[85,86] It has a high propensity for nodal dissemination, especially metastases to regional lymph nodes (12%–43%).[86,87]
The tumor typically has an indolent clinical course. Patients who have small, localized tumors with low mitotic rate and intermediate histological grade have the best outcomes.[88]
Genomic alterations
Clear cell sarcoma of soft tissue is characterized by EWSR1::ATF1 or EWSR1::CREB1 gene fusions.[89,90]
Treatment of clear cell sarcoma of soft tissue
Treatment options for clear cell sarcoma of soft tissue include the following:
Surgery with or without radiation therapy is the treatment of choice and offers the best chance for cure.
Evidence (surgery with or without radiation therapy):
In a series of 28 pediatric patients reported by the Italian and German Soft Tissue Cooperative Studies, the median age at diagnosis was 14 years and the lower extremity was the most common primary site (50%).[91]; [92][Level of evidence C2]
In this series, 12 of 13 patients with completely resected tumors were cured.
For patients with more advanced disease, the outcome is poor and chemotherapy is rarely effective.
Targeted therapy
Evidence (targeted therapy):
In a study by the European Organization for Research and Treatment of Cancer, 26 patients with clear cell sarcoma who had metastatic disease and documented EWSR1 rearrangements were treated with crizotinib.[93]
One patient had a partial response, and 17 patients had stable disease.
Extraskeletal Myxoid Chondrosarcoma
Extraskeletal myxoid chondrosarcoma is relatively rare among soft tissue sarcomas, representing only 2.3% of all soft tissue sarcomas.[94] It has been reported in children and adolescents.[95]
The tumor has traditionally been considered to have low-grade malignant potential.[96] However, reports from large institutions showed that extraskeletal myxoid chondrosarcoma has significant malignant potential, especially if patients are monitored for a long time.[97,98] Patients tend to have slow protracted courses. Nodal involvement has been well described. Local recurrence (57%) and metastatic spread to lungs (26%) have been reported.[98]
Genomic alterations
Extraskeletal myxoid chondrosarcoma is a multinodular neoplasm. The rounded cells are arranged in cords and strands in a chondroitin sulfate myxoid background. Several cytogenetic abnormalities have been identified (see Table 2), with the most frequent being the EWSR1::NR4A3 gene fusion.[99]
Treatment of extraskeletal myxoid chondrosarcoma
Treatment options for extraskeletal myxoid chondrosarcoma include the following:
Surgery.
Radiation therapy.
Aggressive local control and resection of metastases led to OS rates of 87% at 5 years and 63% at 10 years. Tumors were relatively resistant to radiation therapy.[97] The therapeutic benefit of chemotherapy has not been established.
There may be potential genetic targets for small molecules, but these need to be studied as part of a clinical trial. In an adult study, six of ten patients who received sunitinib achieved partial responses.[100]
Desmoplastic small round cell tumor is a rare primitive sarcoma.
Clinical presentation
Desmoplastic small round cell tumor most frequently involves the peritoneum in the abdomen, pelvis, and/or peritoneum into the scrotal sac, but it may occur in the kidney or other solid organs.[102–106] Dozens to hundreds of intraperitoneal implants are often found. The tumor occurs predominantly in males (85%) and may spread to the lungs and elsewhere.[106,107]
Diagnostic evaluation
A large single-institution series of 65 patients compared computed tomography (CT) scans (n = 54) with positron emission tomography (PET)-CT scans (n = 11). PET-CT scans produced very few false-negative results and detected metastatic sites missed on conventional CT scans.[107]
Genomic alterations
Cytogenetic studies of these tumors have demonstrated the recurrent translocation t(11;22)(p13;q12), which has been characterized as a fusion of the WT1 and EWSR1 genes.[105,108] The EWSR1::WT1 fusion confirms the diagnosis of desmoplastic small round cell tumor. The average tumor variant burden is low for desmoplastic small round cell tumor (<1 variant per megabase), and recurring gene alterations other than the EWSR1::WT1 fusion are uncommon.[109] A small percentage of cases (approximately 3%) have activating variants in FGFR4, with amplification of FGFR4 observed at similar frequency.[109,110] Inactivating variants in TP53 and ARID1A are observed in a small percentage of desmoplastic small round cell tumor cases.[109,110]
Prognosis
The overall prognosis for patients with desmoplastic small round cell tumor remains extremely poor, with reported rates of death at 90%. Greater than 90% tumor resection either at presentation or after preoperative chemotherapy may be a favorable prognostic factor for OS.[111,112]; [113][Level of evidence C1] Response to neoadjuvant chemotherapy and complete resection (near 100%) is associated with improved outcome.[106,114]
Treatment of desmoplastic small round cell tumor
There is no standard approach to the treatment of desmoplastic small round cell tumor.
Treatment options for desmoplastic small round cell tumor include the following:
Complete surgical resections are rare and usually performed in highly specialized centers, but are critical for any improved survival. Successful treatment modalities include neoadjuvant Ewing-type chemotherapy, followed by complete surgical resection of the extensive intra-abdominal tumors, followed by total abdominal radiation therapy. With this multimodality approach, survival can be achieved in 30% to 40% of patients at 5 years.[102,103,111,115–118]
Surgery with HIPEC
HIPEC is a local treatment method that may control more of the microscopic intra-abdominal disease. The theory is that the heated chemotherapy that is instilled in the abdominal cavity after surgical resection (at the time of surgery) provides synergistic cytotoxicity to any microscopic cells remaining in the abdomen.[119]
The addition of HIPEC to complete surgical resection (cytoreductive surgery) is a new technique first applied to children in 2006 in a phase I clinical trial. Cytoreductive surgery and HIPEC for desmoplastic small round cell tumors is part of a multidisciplinary approach and is only being done in highly specialized centers. Surgeries can last more than 12 hours, and technical aspects of this unique tumor resection should be considered.[119]
Evidence (surgery with HIPEC):
A single-institution phase II study showed HIPEC to be a potentially promising addition to complete surgical resection. Fourteen patients with desmoplastic small round cell tumor and five patients with other sarcomas were enrolled. These highly selected patients had tumor limited to the abdominal cavity. They demonstrated a partial response to neoadjuvant Ewing-type chemotherapy, had complete surgical resections and received HIPEC using cisplatin. They also received adjuvant total-abdominal radiation therapy followed by adjuvant chemotherapy.[119]
With this standardized approach, patients with desmoplastic small round cell tumors had an OS rate of 80% at 30 months and 40% at 50 months.
Patients with desmoplastic small round cell tumors without liver metastasis had no intra-abdominal recurrences, whereas 87% of patients with liver metastasis or portal disease had a recurrence.
In a retrospective study from centers in France, patients were treated with cytoreductive surgery and HIPEC. Twenty-two patients were selected, and the median age at diagnosis was 14.8 years (range, 4.2–17.6 years). Seven patients had peritoneal mesotheliomas, seven patients had desmoplastic small round cells tumors, and eight patients had other histological tumor types. A complete macroscopic resection (CC-0, where CC is completeness of cytoreduction) was achieved in 16 cases (73%). Four of the seven patients with desmoplastic small round cell tumors had complete resections.[120][Level of evidence C1]
Sixteen patients (72%) experienced relapses after a median time of 9.6 months (range, 1.4–86.4 months).
Nine patients (41%) died of relapsed disease after a median time of 5.3 months (range, 0.1–36.1 months).
Another study from France reviewed the use of cytoreductive surgery and HIPEC for the treatment of patients with desmoplastic small round cell tumors who had disease limited to the abdomen. In 107 patients with desmoplastic small round cell tumors, 48 had no extraperitoneal metastasis and underwent cytoreductive surgery. Of 48 patients (mean age, 22 years), 38 (79%) received preoperative and/or postoperative chemotherapy, and 23 (48%) received postoperative whole-abdominopelvic radiation therapy. Intraperitoneal chemotherapy was administered to 11 patients (23%), 2 of whom received early postoperative intraperitoneal chemotherapy (EPIC) and 9 of whom received HIPEC.[121]
After a median follow-up of 30 months, the median OS of the entire cohort was 42 months.
The 2-year OS rate was 72%, and the 5-year OS rate was 19%.
The 2-year disease-free survival (DFS) rate was 30%, and the 5-year DFS rate was 12%.
Whole-abdominopelvic radiation therapy was the only variable associated with longer peritoneal recurrence-free survival and DFS after cytoreductive surgery.
Of 11 patients who received intraperitoneal chemotherapy (HIPEC or EPIC), six different chemotherapy regimens were used. The survival or outcome of this group is not reported in the manuscript.
The influence of HIPEC/EPIC on OS and DFS was not statistically significant, but standardized regimens were not used in all patients, making results difficult to determine.
A single-institutional retrospective study reported on nine patients (median age, 19 years) with desmoplastic small round cell tumor. Most patients had widespread disease, including four patients with extra-abdominal disease and five patients with liver involvement. These nine patients underwent ten cytoreductive and HIPEC treatments. Additionally, seven patients also received radiation therapy, and three patients underwent stem cell transplant.[122]
The 3-year relapse-free survival rate was 13%, and the OS rate was 55%.
Therapy was often associated with prolonged hospitalizations.
Long-term parenteral nutrition was required in eight patients for a median of 261 days.
Other long-term complications included gastroparesis (n = 1), small bowel obstruction (n = 3), and hemorrhagic cystitis (n = 2).
Other treatment options
The Center for International Blood and Marrow Transplant Research analyzed patients with desmoplastic small round cell tumor in their registry who received consolidation with high-dose chemotherapy and autologous stem cell reconstitution.[123] While this retrospective registry analysis suggested some benefit to this approach, other investigators have abandoned the approach because of excessive toxicity and lack of efficacy.[111]
A single-institution study reported that five of five patients with recurrent desmoplastic small round cell tumor had partial responses to treatment with the combination of vinorelbine, cyclophosphamide, and temsirolimus.[124]
Rhabdoid Tumor NOS (Extrarenal)
Malignant rhabdoid tumors were first described in children with renal tumors in 1981. These tumors were later found in a variety of extrarenal sites. These tumors are uncommon and highly malignant, especially in children younger than 2 years. For more information, see the Rhabdoid Tumors of the Kidney section in Wilms Tumor and Other Childhood Kidney Tumors Treatment.
Extrarenal (extracranial) rhabdoid tumors account for 2% of soft tissue sarcomas in patients younger than 20 years (see Table 1).
Genetic and genomic alterations
The first sizeable series of children with extrarenal extracranial malignant rhabdoid tumor of soft tissues came from 26 patients enrolled on the IRS I through III studies during a review of pathology material. Only five patients (19%) were alive without disease beyond 2 years.[125]
Investigation of children with atypical teratoid/rhabdoid tumors of the brain, as well as those with renal and extrarenal malignant rhabdoid tumors, found germline pathogenic variants and acquired variants of the SMARCB1 gene in all 29 tumors tested.[126] Rhabdoid tumors may be associated with germline pathogenic variants of the SMARCB1 gene and may be inherited from an apparently unaffected parent.[127] This observation was extended to 32 malignant rhabdoid tumors at all sites in patients whose mean age at diagnosis was 12 months.[128]
Genetic testing and surveillance
Germline analysis should be considered for individuals of all ages with rhabdoid tumors. Genetic counseling is also part of the treatment plan, given the low-but-actual risk of familial recurrence. In cases of variants, parental screening should be considered, although such screening carries a low probability of positivity. Prenatal diagnosis can be performed in situations where a specific SMARCB1 variant or deletion has been documented in the family.[127]
To date, there is little evidence regarding the effectiveness of surveillance for patients with rhabdoid tumor predisposition syndrome type 1 caused by loss-of-function germline SMARCB1 pathogenic variants. However, because of the aggressive nature of the tumors with significant lethality and young age of onset in SMARCB1 carriers with truncating variants, consensus recommendations have been developed. These recommendations were developed by a group of pediatric cancer genetic experts (including oncologists, radiologists, and geneticists). They have not been formally studied to confirm the benefit of monitoring patients with germline SMARCB1 pathogenic variants. Given the potential survival benefit of surgically resectable disease, it is postulated that early detection might improve OS.[129–131]
Surveillance for patients with germline SMARCB1 pathogenic variants includes the following:
Brain magnetic resonance imaging (MRI) every 3 months from birth (or diagnosis) until age 5 years.
Abdominal ultrasonography with a focus on the kidneys every 3 months.
Young age and metastatic disease at presentation are associated with poor outcomes in children with extracranial rhabdoid tumors.
One study that used data from the National Cancer Database identified 202 patients (aged younger than 18 years) with non–central nervous system (CNS) malignant rhabdoid tumors. The primary site of the malignant rhabdoid tumor was soft tissue (46%), kidney (45%), and liver (9%).[132]
The 1-year OS rate was 48.8%, and the 5-year OS rate was 35.9%.
The multivariate analysis demonstrated that age younger than 1 year and presence of metastasis were negative prognostic indications (P = .058).
In the cohort of surgical patients (n = 143), there was a trend for an association between the presence of residual disease and a clinically significant worse outcome (HR, 1.54; 95% CI, 0.88–2.69; P = .13).
A SEER study examined 229 patients with renal, CNS, and extrarenal malignant rhabdoid tumor. Patients aged 2 to 18 years, patients with a limited extent of tumor, and patients who received radiation therapy had favorable outcomes compared with other patients (P < .002 for each comparison). Site of the primary tumor was not prognostically significant. The OS rate was 33% at 5 years.[133]
A European registry for extracranial rhabdoid tumors identified 100 patients from 14 countries between 2009 and 2018.[134] Half of the patients were younger than 1 year at diagnosis. In 30 patients (30%), the tumor was located in the kidneys. Extracranial, extrarenal malignant rhabdoid tumor was found in 70% of patients (70 of 100), and the most common locations were in the cervical region, thoracic region, and liver. Nine patients demonstrated synchronous tumors. Distant metastases at diagnosis were present in 35% of patients (35 of 100). SMARCB1 germline pathogenic variants were detected in 21% of patients (17 of 81 evaluable). The 5-year OS rate was 45.8% (± 5.4%), and the EFS rate was 35.2% (± 5.1%). In an adjusted multivariate model, presence of a germline pathogenic variant, metastasis, and lack of a gross-total resection were the strongest significant negative predictors of outcome.
Treatment of extrarenal (extracranial) rhabdoid tumor
Treatment options for extrarenal (extracranial) rhabdoid tumor include the following:[135–137][Level of evidence C1]
Surgical removal when possible.
Chemotherapy as used for soft tissue sarcomas (but no single regimen is currently accepted as best).
Information about NCI-supported clinical trials can be found on the NCI website. For information about clinical trials sponsored by other organizations, see the ClinicalTrials.gov website.
The following is an example of a national and/or institutional clinical trial that is currently being conducted:
PEPN2121 (NCT05286801) (Tiragolumab and Atezolizumab for the Treatment of Relapsed or Refractory SMARCB1– or SMARCA4-Deficient Tumors): This study is evaluating the combination of a PD-L1 targeting antibody (atezolizumab) with a TIGIT targeting antibody (tiragolumab) for patients with SMARCB1– or SMARCA4-deficient tumors. Patients with extrarenal (extracranial) rhabdoid tumors may be eligible for this study.
PEComa, Malignant
Clinical presentation
PEComas occur in various rare gastrointestinal, pulmonary, gynecological, and genitourinary sites. Soft tissue, visceral, and gynecological PEComas are more commonly seen in middle-aged female patients and are usually not associated with the tuberous sclerosis complex.[139] The disease course may be indolent.
Risk factors and molecular features
Benign PEComas are common in patients with tuberous sclerosis, an autosomal dominant syndrome that also predisposes to renal cell cancer and brain tumors. Tuberous sclerosis is caused by germline pathogenic inactivation of either TSC1 (9q34) or TSC2 (16p13.3), and the same tumor suppressor genes are inactivated somatically in sporadic PEComas.[140] Inactivation of either gene results in stimulation of the mTOR pathway, providing the basis for the treatment of nonsurgically curable tumors with similar genetic inactivation (lymphangioleiomyomatosis and angiomyolipoma) with mTOR inhibitors.[141,142] A small proportion of PEComas have TFE3 rearrangements with fusions involving various genes, including SFPQ and RAD51B.[143]
Prognosis
Most PEComas have a benign clinical course, but malignant behavior has been reported and can be predicted based on the size of the tumor, mitotic rate, and presence of necrosis.[144]
Treatment of PEComas
There are no standard treatment options. Treatment may include surgery or observation followed by surgery when the tumor is large.[145]
In tumors with evidence of mTORC1 activation and TSC1 or TSC2 loss, including lymphangioleiomyomatosis and angiomyolipoma,[141] clinical activity using mTOR inhibitors, such as sirolimus, has been well documented. In a small case series, three adult patients with PEComas responded to sirolimus.[146]
In a phase II trial, 34 patients with metastatic or locally advanced malignant PEComas were treated with sirolimus protein-bound particles for injectable suspension (albumin-bound) (nab-sirolimus). Of the 31 patients eligible for efficacy analysis, 12 (39%) had a response (1 complete response and 11 partial responses), 16 (52%) had stable disease, and 3 (10%) had progressive disease. Responses were rapid and durable. The median duration of response was not reached after a median follow-up of 2.5 years. Treatment was ongoing for 7 of 12 patients who responded to treatment (range, 5.6 months to longer than 47.2 months). Tumor variant profiling was completed for 25 specimens. Eight of nine patients with TSC2 variants responded to treatment, while only 2 of 16 patients without TSC2 variants responded. In addition, responses were noted in 10 of 17 patients with phospho-S6 (pS6) expression. No response was noted in eight patients without pS6 expression. The absence of pS6 expression reflects the lack of mTORC1 activation.[147][Level of evidence C1] In 2021, the FDA approved nab-sirolimus for adult patients with PEComas.
Undifferentiated Sarcoma
From 1972 to 2006, patients with undifferentiated soft tissue sarcoma were eligible for participation in rhabdomyosarcoma trials coordinated by the IRS group and the COG. The rationale was that patients with undifferentiated soft tissue sarcoma had sites of disease and outcomes that were similar to those in patients with alveolar rhabdomyosarcoma. Therapeutic trials for adults with soft tissue sarcoma include patients with undifferentiated soft tissue sarcoma and other histologies, which are treated similarly, using ifosfamide and doxorubicin, and sometimes with other chemotherapy agents, surgery, and radiation therapy.
In the COG ARST0332 (NCT00346164) trial, patients with high-grade undifferentiated sarcoma were treated with an ifosfamide- and doxorubicin-based regimen. Results for the patients with high-grade undifferentiated sarcoma were reported together with all high-grade soft tissue sarcomas in the trial. The estimated 5-year EFS rate was 64% and the OS rate was 77% for sarcomas classified as high grade by the Fédération Nationale des Centres de Lutte Contre Le Cancer (FNCLC).[5][Level of evidence C1]
In a report of 32 patients with undifferentiated soft tissue sarcomas who were enrolled on the ARST0332 (NCT00346164) trial, the median age at enrollment was 13.6 years, and two-thirds of the patients were male. The most common primary sites were the paraspinal region and extremities. Five patients presented with metastatic disease.[148]
The 5-year EFS rate was 71%, and the OS rate was 83%.
Of the nine children with low-risk disease (localized low-grade resected disease or localized high-grade disease <5 cm resected with negative margins) who were treated with surgery or radiation therapy only, the 5-year EFS rate was 65% and the OS rate was 100%, suggesting that patients with low-risk disease can be salvaged if the disease recurs.
The remaining 23 patients had either intermediate-risk disease (resected high-grade tumor >5 cm, unresected high-grade tumor >5 cm) or high-risk disease (metastasis to lymph nodes or distant sites) and were treated with chemoradiation therapy and delayed surgery when feasible. The 5-year EFS rate was 73%, and the OS estimate was 77%.
Copy number aberrations were common, most frequently involving loss of 1p (25%), gain of 1q (25%), gain of chromosome 8 (25%), and gain of chromosome 2 (16%). These alterations were more commonly seen in patients with intermediate-risk or high-risk tumors, and there was a strong association between loss of chromosome 1p or gain of chromosome 1q and inferior clinical outcomes. Co-occurrence of 1q gain and 1p loss was associated with a particularly poor clinical outcome (5-year EFS and OS rates of 20%). Next-generation sequencing identified oncogenic fusions in eight of ten tumor samples, which included BCOR and CIC rearrangements, as well as COL1A1::PDGFB, KIAA1549::BRAF, and SAMD5::SASH1 gene fusions.
At one time, malignant fibrous histiocytoma was the single most common histotype among adults with soft tissue sarcomas. Since it was first recognized in the early 1960s, malignant fibrous histiocytoma has been controversial, in terms of both its histogenesis and its validity as a clinico-pathological entity. The World Health Organization (WHO) classification no longer includes malignant fibrous histiocytoma as a distinct diagnostic category but rather as a subtype of an undifferentiated pleomorphic sarcoma.[149,150]
This entity accounts for 2% to 6% of all childhood soft tissue sarcomas.[151]
Clinical presentation
These tumors occur mainly in the second decade of life. In a series of ten patients, the median age was 10 years, and the tumor was most commonly located in the extremities. In this series, all tumors were localized, and five of nine patients (for whom follow-up was available) were alive and in first remission.[151]
In another series of 17 pediatric patients with malignant fibrous histiocytoma (now classified as undifferentiated pleomorphic sarcoma), the median age at diagnosis was 5 years and the extremities were involved in eight cases.[152] All patients with metastatic disease died, and two patients experienced a clinical response to a doxorubicin-based regimen.
These tumors can arise in previously irradiated sites or as a second malignancy in patients with retinoblastoma.[153]
Molecular features
An analysis of 70 patients who were diagnosed with malignant fibrous histiocytosis of no specific type, storiform or pleomorphic malignant fibrous histiocytoma, pleomorphic sarcoma, or undifferentiated pleomorphic sarcoma showed a highly complex karyotype with no specific recurrent aberrations.[154]
Undifferentiated sarcomas with 12q13–15 amplification, including MDM2 and CDK4, are best classified as dedifferentiated liposarcomas.[154] The relationship between this tumor and the family of undifferentiated/unclassified tumors with spindle cell morphology remains relatively undefined.
Treatment of recurrent or refractory pleomorphic sarcoma
Treatment options for recurrent or refractory pleomorphic sarcoma include the following:
Pembrolizumab.
The Sarcoma Alliance for Research through Collaboration conducted a phase II trial of the checkpoint inhibitor pembrolizumab in patients aged 18 years and older with recurrent soft tissue sarcoma.[155][Level of evidence C3]
Seven of 40 patients (18%) with soft tissue sarcoma had an objective response.
Four of ten patients (40%) with undifferentiated pleomorphic sarcoma, two of ten patients (20%) with liposarcoma, and one of ten patients (10%) with synovial sarcoma had objective responses.
No patients with leiomyosarcoma (n = 10) had an objective response.
Intracranial Mesenchymal Tumor
Intracranial mesenchymal tumor, with the FET::CREB gene fusion, has previously been called angiomatoid fibrous histiocytoma (AFP) or intracranial myxoid mesenchymal tumor. The molecular findings suggest that these tumors are histological variants of intracranial mesenchymal tumor.[156] In one study, the tumors of 20 patients were separated into two epigenetic subgroups. Group A tumors clustered nearest to but independent of solitary fibrous tumor and occurred in adolescents and young adults. Group B tumors clustered nearest to but independent of clear cell sarcoma and occurred in children. Patients with group B tumors had an inferior survival compared with patients with group A tumors (4.5 vs. 49 months; P = .001).[157]
Round Cell Sarcoma, Undifferentiated
Undifferentiated small round cell sarcomas with BCOR genetic alterations
Carney JA, Young WF: Primary pigmented nodular adrenocortical disease and its associated conditions. Endocrinologist 2: 6-21, 1992.
Ryan MW, Cunningham S, Xiao SY: Maxillary sinus melanoma as the presenting feature of Carney complex. Int J Pediatr Otorhinolaryngol 72 (3): 405-8, 2008. [PUBMED Abstract]
Sultan I, Rodriguez-Galindo C, Saab R, et al.: Comparing children and adults with synovial sarcoma in the Surveillance, Epidemiology, and End Results program, 1983 to 2005: an analysis of 1268 patients. Cancer 115 (15): 3537-47, 2009. [PUBMED Abstract]
Spunt SL, Million L, Chi YY, et al.: A risk-based treatment strategy for non-rhabdomyosarcoma soft-tissue sarcomas in patients younger than 30 years (ARST0332): a Children’s Oncology Group prospective study. Lancet Oncol 21 (1): 145-161, 2020. [PUBMED Abstract]
Wang JG, Li NN: Primary cardiac synovial sarcoma. Ann Thorac Surg 95 (6): 2202-9, 2013. [PUBMED Abstract]
Chirmade PC, Parikh S, Anand A, et al.: Primary pleuropulmonary synovial sarcoma with brain metastases in a paediatric patient: an unusual presentation. Adv Respir Med 85 (4): 206-210, 2017. [PUBMED Abstract]
Frazier AA, Franks TJ, Pugatch RD, et al.: From the archives of the AFIP: Pleuropulmonary synovial sarcoma. Radiographics 26 (3): 923-40, 2006 May-Jun. [PUBMED Abstract]
Essary LR, Vargas SO, Fletcher CD: Primary pleuropulmonary synovial sarcoma: reappraisal of a recently described anatomic subset. Cancer 94 (2): 459-69, 2002. [PUBMED Abstract]
Pappo AS, Fontanesi J, Luo X, et al.: Synovial sarcoma in children and adolescents: the St Jude Children’s Research Hospital experience. J Clin Oncol 12 (11): 2360-6, 1994. [PUBMED Abstract]
Ferrari A, De Salvo GL, Oberlin O, et al.: Synovial sarcoma in children and adolescents: a critical reappraisal of staging investigations in relation to the rate of metastatic involvement at diagnosis. Eur J Cancer 48 (9): 1370-5, 2012. [PUBMED Abstract]
Scheer M, Blank B, Bauer S, et al.: Synovial sarcoma disease characteristics and primary tumor sites differ between patient age groups: a report of the Cooperative Weichteilsarkom Studiengruppe (CWS). J Cancer Res Clin Oncol 146 (4): 953-960, 2020. [PUBMED Abstract]
van de Rijn M, Barr FG, Collins MH, et al.: Absence of SYT-SSX fusion products in soft tissue tumors other than synovial sarcoma. Am J Clin Pathol 112 (1): 43-9, 1999. [PUBMED Abstract]
Krsková L, Sumerauer D, Stejskalová E, et al.: A novel variant of SYT-SSX1 fusion gene in a case of spindle cell synovial sarcoma. Diagn Mol Pathol 16 (3): 179-83, 2007. [PUBMED Abstract]
Su L, Sampaio AV, Jones KB, et al.: Deconstruction of the SS18-SSX fusion oncoprotein complex: insights into disease etiology and therapeutics. Cancer Cell 21 (3): 333-47, 2012. [PUBMED Abstract]
Arnold MA, Arnold CA, Li G, et al.: A unique pattern of INI1 immunohistochemistry distinguishes synovial sarcoma from its histologic mimics. Hum Pathol 44 (5): 881-7, 2013. [PUBMED Abstract]
Vlenterie M, Ho VK, Kaal SE, et al.: Age as an independent prognostic factor for survival of localised synovial sarcoma patients. Br J Cancer 113 (11): 1602-6, 2015. [PUBMED Abstract]
Smolle MA, Parry M, Jeys L, et al.: Synovial sarcoma: Do children do better? Eur J Surg Oncol 45 (2): 254-260, 2019. [PUBMED Abstract]
Okcu MF, Munsell M, Treuner J, et al.: Synovial sarcoma of childhood and adolescence: a multicenter, multivariate analysis of outcome. J Clin Oncol 21 (8): 1602-11, 2003. [PUBMED Abstract]
Brecht IB, Ferrari A, Int-Veen C, et al.: Grossly-resected synovial sarcoma treated by the German and Italian Pediatric Soft Tissue Sarcoma Cooperative Groups: discussion on the role of adjuvant therapies. Pediatr Blood Cancer 46 (1): 11-7, 2006. [PUBMED Abstract]
Stanelle EJ, Christison-Lagay ER, Healey JH, et al.: Pediatric and adolescent synovial sarcoma: multivariate analysis of prognostic factors and survival outcomes. Ann Surg Oncol 20 (1): 73-9, 2013. [PUBMED Abstract]
Lagarde P, Przybyl J, Brulard C, et al.: Chromosome instability accounts for reverse metastatic outcomes of pediatric and adult synovial sarcomas. J Clin Oncol 31 (5): 608-15, 2013. [PUBMED Abstract]
Stegmaier S, Leuschner I, Poremba C, et al.: The prognostic impact of SYT-SSX fusion type and histological grade in pediatric patients with synovial sarcoma treated according to the CWS (Cooperative Weichteilsarkom Studie) trials. Pediatr Blood Cancer 64 (1): 89-95, 2017. [PUBMED Abstract]
Scheer M, Dantonello T, Hallmen E, et al.: Primary Metastatic Synovial Sarcoma: Experience of the CWS Study Group. Pediatr Blood Cancer 63 (7): 1198-206, 2016. [PUBMED Abstract]
Orbach D, Mosseri V, Pissaloux D, et al.: Genomic complexity in pediatric synovial sarcomas (Synobio study): the European pediatric soft tissue sarcoma group (EpSSG) experience. Cancer Med 7 (4): 1384-1393, 2018. [PUBMED Abstract]
Trassard M, Le Doussal V, Hacène K, et al.: Prognostic factors in localized primary synovial sarcoma: a multicenter study of 128 adult patients. J Clin Oncol 19 (2): 525-34, 2001. [PUBMED Abstract]
Guillou L, Benhattar J, Bonichon F, et al.: Histologic grade, but not SYT-SSX fusion type, is an important prognostic factor in patients with synovial sarcoma: a multicenter, retrospective analysis. J Clin Oncol 22 (20): 4040-50, 2004. [PUBMED Abstract]
Ferrari A, Gronchi A, Casanova M, et al.: Synovial sarcoma: a retrospective analysis of 271 patients of all ages treated at a single institution. Cancer 101 (3): 627-34, 2004. [PUBMED Abstract]
Bahig H, Roberge D, Bosch W, et al.: Agreement among RTOG sarcoma radiation oncologists in contouring suspicious peritumoral edema for preoperative radiation therapy of soft tissue sarcoma of the extremity. Int J Radiat Oncol Biol Phys 86 (2): 298-303, 2013. [PUBMED Abstract]
Baldini EH, Wang D, Haas RL, et al.: Treatment Guidelines for Preoperative Radiation Therapy for Retroperitoneal Sarcoma: Preliminary Consensus of an International Expert Panel. Int J Radiat Oncol Biol Phys 92 (3): 602-12, 2015. [PUBMED Abstract]
Ferrari A, Chi YY, De Salvo GL, et al.: Surgery alone is sufficient therapy for children and adolescents with low-risk synovial sarcoma: A joint analysis from the European paediatric soft tissue sarcoma Study Group and the Children’s Oncology Group. Eur J Cancer 78: 1-6, 2017. [PUBMED Abstract]
McGrory JE, Pritchard DJ, Arndt CA, et al.: Nonrhabdomyosarcoma soft tissue sarcomas in children. The Mayo Clinic experience. Clin Orthop (374): 247-58, 2000. [PUBMED Abstract]
Van Glabbeke M, van Oosterom AT, Oosterhuis JW, et al.: Prognostic factors for the outcome of chemotherapy in advanced soft tissue sarcoma: an analysis of 2,185 patients treated with anthracycline-containing first-line regimens–a European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group Study. J Clin Oncol 17 (1): 150-7, 1999. [PUBMED Abstract]
Koscielniak E, Harms D, Henze G, et al.: Results of treatment for soft tissue sarcoma in childhood and adolescence: a final report of the German Cooperative Soft Tissue Sarcoma Study CWS-86. J Clin Oncol 17 (12): 3706-19, 1999. [PUBMED Abstract]
Pappo AS, Devidas M, Jenkins J, et al.: Phase II trial of neoadjuvant vincristine, ifosfamide, and doxorubicin with granulocyte colony-stimulating factor support in children and adolescents with advanced-stage nonrhabdomyosarcomatous soft tissue sarcomas: a Pediatric Oncology Group Study. J Clin Oncol 23 (18): 4031-8, 2005. [PUBMED Abstract]
Cecchetto G, Alaggio R, Dall’Igna P, et al.: Localized unresectable non-rhabdo soft tissue sarcomas of the extremities in pediatric age: results from the Italian studies. Cancer 104 (9): 2006-12, 2005. [PUBMED Abstract]
Pappo AS, Rao BN, Jenkins JJ, et al.: Metastatic nonrhabdomyosarcomatous soft-tissue sarcomas in children and adolescents: the St. Jude Children’s Research Hospital experience. Med Pediatr Oncol 33 (2): 76-82, 1999. [PUBMED Abstract]
Brennan B, Stevens M, Kelsey A, et al.: Synovial sarcoma in childhood and adolescence: a retrospective series of 77 patients registered by the Children’s Cancer and Leukaemia Group between 1991 and 2006. Pediatr Blood Cancer 55 (1): 85-90, 2010. [PUBMED Abstract]
Ferrari A, Miceli R, Rey A, et al.: Non-metastatic unresected paediatric non-rhabdomyosarcoma soft tissue sarcomas: results of a pooled analysis from United States and European groups. Eur J Cancer 47 (5): 724-31, 2011. [PUBMED Abstract]
Raney RB: Synovial sarcoma in young people: background, prognostic factors, and therapeutic questions. J Pediatr Hematol Oncol 27 (4): 207-11, 2005. [PUBMED Abstract]
Orbach D, Mc Dowell H, Rey A, et al.: Sparing strategy does not compromise prognosis in pediatric localized synovial sarcoma: experience of the International Society of Pediatric Oncology, Malignant Mesenchymal Tumors (SIOP-MMT) Working Group. Pediatr Blood Cancer 57 (7): 1130-6, 2011. [PUBMED Abstract]
Ladenstein R, Treuner J, Koscielniak E, et al.: Synovial sarcoma of childhood and adolescence. Report of the German CWS-81 study. Cancer 71 (11): 3647-55, 1993. [PUBMED Abstract]
Venkatramani R, Xue W, Randall RL, et al.: Synovial Sarcoma in Children, Adolescents, and Young Adults: A Report From the Children’s Oncology Group ARST0332 Study. J Clin Oncol 39 (35): 3927-3937, 2021. [PUBMED Abstract]
Ferrari A, De Salvo GL, Brennan B, et al.: Synovial sarcoma in children and adolescents: the European Pediatric Soft Tissue Sarcoma Study Group prospective trial (EpSSG NRSTS 2005). Ann Oncol 26 (3): 567-72, 2015. [PUBMED Abstract]
Scheer M, Hallmen E, Vokuhl C, et al.: Pre-operative radiotherapy is associated with superior local relapse-free survival in advanced synovial sarcoma. J Cancer Res Clin Oncol 149 (5): 1717-1731, 2023. [PUBMED Abstract]
Ferrari A, De Salvo GL, Dall’Igna P, et al.: Salvage rates and prognostic factors after relapse in children and adolescents with initially localised synovial sarcoma. Eur J Cancer 48 (18): 3448-55, 2012. [PUBMED Abstract]
Scheer M, Dantonello T, Hallmen E, et al.: Synovial Sarcoma Recurrence in Children and Young Adults. Ann Surg Oncol 23 (Suppl 5): 618-626, 2016. [PUBMED Abstract]
Ferrari A, Orbach D, Bergamaschi L, et al.: Treatment at relapse for synovial sarcoma of children and adolescents: A multi-institutional European retrospective analysis. Pediatr Blood Cancer 71 (7): e31038, 2024. [PUBMED Abstract]
Dhakal S, Corbin KS, Milano MT, et al.: Stereotactic body radiotherapy for pulmonary metastases from soft-tissue sarcomas: excellent local lesion control and improved patient survival. Int J Radiat Oncol Biol Phys 82 (2): 940-5, 2012. [PUBMED Abstract]
Lai JP, Robbins PF, Raffeld M, et al.: NY-ESO-1 expression in synovial sarcoma and other mesenchymal tumors: significance for NY-ESO-1-based targeted therapy and differential diagnosis. Mod Pathol 25 (6): 854-8, 2012. [PUBMED Abstract]
Robbins PF, Kassim SH, Tran TL, et al.: A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 21 (5): 1019-27, 2015. [PUBMED Abstract]
D’Angelo SP, Melchiori L, Merchant MS, et al.: Antitumor Activity Associated with Prolonged Persistence of Adoptively Transferred NY-ESO-1 c259T Cells in Synovial
Kaposi sarcoma (KS) was first described in 1872 by the Hungarian dermatologist, Moritz Kaposi. From that time until the HIV and AIDS epidemic, KS remained a rare tumor. Classic KS is most commonly seen in Europe and North America in older men of Italian or Eastern European Jewish ancestry,[1] and endemic KS is most commonly seen in sub-Saharan Africa. The disseminated, fulminant form of KS associated with HIV disease is referred to as AIDS-associated KS to distinguish it from classic and endemic KS. Transplant-related KS (also sometimes called iatrogenic KS) is seen in patients receiving chronic immunosuppression therapy, such as after organ transplant.[2,3]
Histopathology
Although the histopathology of the different types of KS is essentially identical, the clinical manifestations and course of the disease differ dramatically.[2] Human herpesvirus 8 (HHV8), also known as Kaposi sarcoma-associated herpesvirus, was identified in KS tissue biopsies from almost all patients with classic, endemic, AIDS-associated, and transplant-related KS but was absent from noninvolved tissue.[2]
Classic Kaposi Sarcoma
Classic KS is considered a rare disease. It occurs more often in men, at a ratio of approximately 10 to 15 men to 1 woman. In North American and European populations, the usual age at onset is between 50 and 70 years. Classic KS tumors usually present with one or more asymptomatic red, purple, or brown patches, plaques, or nodular skin lesions. The disease is often limited to single or multiple lesions usually localized to one or both lower extremities, especially involving the ankles and soles.
Classic KS most commonly runs a relatively benign, indolent course for 10 to 15 years or more, with slow enlargement of the original tumors and the gradual development of additional lesions. Venous stasis and lymphedema of the involved lower extremity are frequent complications. In long-standing cases, systemic lesions can develop along the gastrointestinal tract, in lymph nodes, and in other organs. The visceral lesions are generally asymptomatic and are most often discovered only at autopsy, though clinically, gastrointestinal bleeding can occur. As many as 33% of patients with classic KS develop a second primary malignancy, which is most often non-Hodgkin lymphoma.[4]
Endemic Kaposi Sarcoma
Endemic KS refers to KS diagnosed in patients, typically children and younger adults, living in sub-Saharan Africa. This classification was based on several reports from the 1950s of KS in this younger HIV-negative cohort in human herpesvirus–endemic African countries. Prior to the AIDS epidemic, the estimated incidence for endemic KS was highest (>6 per 1,000 person-years) in Uganda, Tanzania, Cameroon, and Congo. The etiology behind endemic KS is unclear but may possibly be related to saliva-sharing practices, chronic infection, and malnutrition.[3]
The clinical presentation of endemic KS varies and differs between children and adults. Whereas adults present with disease that resembles classic KS, children can have more aggressive disease, including diffuse lymphadenopathy, significant lymphedema, and visceral dissemination.[3]
AIDS-Associated Kaposi Sarcoma
The use of antiretroviral therapy for patients with AIDS-associated KS has been associated with a sustained and substantial decline in KS incidence in multiple large cohorts.[5–7] Antiretroviral therapy has delayed or prevented the emergence of drug-resistant HIV strains, profoundly decreased viral load, led to increased survival, and lessened the risk of opportunistic infections.[8] KS can still appear during antiretroviral therapy with complete suppression of HIV; most cases in the United States occur in patients with high CD4 counts receiving ongoing antiretroviral therapy.[9]
The disease often progresses in an orderly fashion from a few localized or widespread mucocutaneous lesions that may involve the skin, oral mucosa, and lymph nodes to more numerous lesions and generalized skin disease that involves visceral organs, such as the gastrointestinal tract, lung, liver, and spleen. Most patients with HIV disease who present with mucocutaneous KS lesions feel healthy and are usually free of systemic symptoms, as compared with HIV patients who first develop an opportunistic infection. AIDS-associated KS presents at sites that are much more varied than those seen in other types of this neoplasm. While most patients present with skin disease, KS involvement of lymph nodes or the gastrointestinal tract may occasionally precede the appearance of the cutaneous lesions.
Transplant-Related Kaposi Sarcoma
Transplant-related KS (also called iatrogenic KS) is diagnosed in patients who are therapeutically immunosuppressed, such as after an organ transplant. In fact, solid-organ transplant recipients are 200-fold more likely to develop KS than the general population. Risk factors include male sex, older age, higher levels of immune suppression, and living in HHV8-endemic areas.[3]
Transplant-related KS typically yields cutaneous lesions, though mucosal and visceral disease can occur. The lesions commonly occur within the first several months of immunosuppression therapy and regress with changes or reductions in immunosuppression.[3]
References
Ruocco E, Ruocco V, Tornesello ML, et al.: Kaposi’s sarcoma: etiology and pathogenesis, inducing factors, causal associations, and treatments: facts and controversies. Clin Dermatol 31 (4): 413-422, 2013 Jul-Aug. [PUBMED Abstract]
Uldrick TS, Whitby D: Update on KSHV epidemiology, Kaposi Sarcoma pathogenesis, and treatment of Kaposi Sarcoma. Cancer Lett 305 (2): 150-62, 2011. [PUBMED Abstract]
Cesarman E, Damania B, Krown SE, et al.: Kaposi sarcoma. Nat Rev Dis Primers 5 (1): 9, 2019. [PUBMED Abstract]
Safai B, Good RA: Kaposi’s sarcoma: a review and recent developments. Clin Bull 10 (2): 62-9, 1980. [PUBMED Abstract]
Portsmouth S, Stebbing J, Gill J, et al.: A comparison of regimens based on non-nucleoside reverse transcriptase inhibitors or protease inhibitors in preventing Kaposi’s sarcoma. AIDS 17 (11): F17-22, 2003. [PUBMED Abstract]
Carrieri MP, Pradier C, Piselli P, et al.: Reduced incidence of Kaposi’s sarcoma and of systemic non-hodgkin’s lymphoma in HIV-infected individuals treated with highly active antiretroviral therapy. Int J Cancer 103 (1): 142-4, 2003. [PUBMED Abstract]
Grabar S, Abraham B, Mahamat A, et al.: Differential impact of combination antiretroviral therapy in preventing Kaposi’s sarcoma with and without visceral involvement. J Clin Oncol 24 (21): 3408-14, 2006. [PUBMED Abstract]
Lodi S, Guiguet M, Costagliola D, et al.: Kaposi sarcoma incidence and survival among HIV-infected homosexual men after HIV seroconversion. J Natl Cancer Inst 102 (11): 784-92, 2010. [PUBMED Abstract]
Yanik EL, Achenbach CJ, Gopal S, et al.: Changes in Clinical Context for Kaposi’s Sarcoma and Non-Hodgkin Lymphoma Among People With HIV Infection in the United States. J Clin Oncol 34 (27): 3276-83, 2016. [PUBMED Abstract]
Stage Information and Response Evaluation for Kaposi Sarcoma
Staging
The staging evaluation of patients with classic Kaposi sarcoma (KS) should be individualized. The advanced age of most patients, localized nature of the tumor, rarity of visceral involvement, and usually indolent course of the disease should temper the extent of the evaluation. A careful examination of the skin and lymph nodes is sufficient in most cases.
For the rare patient with a rapidly progressive tumor or signs or symptoms of visceral involvement, appropriate evaluation is indicated. No universally accepted classification is available for AIDS-associated KS. Staging schemes that incorporate laboratory parameters as well as clinical features have been proposed. Since most patients with AIDS-associated KS do not die of the disease, factors besides tumor burden are apparently involved in survival.
The conventions used to stage KS and the methods used to evaluate the benefits of KS treatment continue to evolve because of changes in the treatment of HIV and in recognition of deficiencies in standard tumor assessment. The clinical course of KS, the selection of treatment, and the response to treatment are heavily influenced by the degree of underlying immune dysfunction and opportunistic infections.
The AIDS Clinical Trials Group (ACTG) Oncology Committee has published criteria for the evaluation of AIDS-associated KS.[1] The staging system incorporates measures of extent of disease, severity of immunodeficiency, and presence of systemic symptoms. As shown in Table 1 below, the ACTG criteria categorize the extent of the tumor as localized or disseminated, the CD4 cell number as high or low, and systemic illness as absent or present.
A subsequent prospective analysis of 294 patients entered on ACTG trials for KS between 1989 and 1995 showed that each of the tumor (T), immune system (I), and systemic illness (S) variables was independently associated with survival.[2] Multivariate analysis showed that immune system impairment was the most important single predictor of survival. In patients with relatively high CD4 counts, tumor stage was predictive. A CD4 count of 150 cells/µL may be a better discriminator than the published cutoff of 200 cells/µL. A study is under way to determine if quantifying viral load adds predictive value. None of the previous studies were conducted at a time when antiretroviral therapy was readily available. The impact of antiretroviral therapy on survival in KS requires continued assessment.
Table 1. AIDS Clinical Trials Group (ACTG) Staging Classification
Variable
Good Risk (0)
Poor Risk (1)
KS = Kaposi sarcoma; OI = opportunistic infection.
aMinimal oral disease is non-nodular KS confined to the palate.
b“B” symptoms are unexplained fever, night sweats, >10% involuntary weight loss, or diarrhea persisting >2 weeks.
(Any of the following)
(Any of the following)
Tumor (T)
Confined to skin and/or lymph nodes and/or minimal oral diseasea
Tumor-associated edema or ulceration
Extensive oral KS
Gastrointestinal KS
KS in other non-nodal viscera
Immune system (I)
CD4 cells ≥200/µL
CD4 cells <200/µL
Systemic illness (S)
No history of OIs or thrush
History of OIs and/or thrush
No “B” symptomsb
“B” symptoms present
Performance status ≥70 (Karnofsky)
Performance status <70
Other HIV-related illness (e.g., neurological disease or lymphoma)
Response Evaluation
The ACTG proposed a unified treatment response evaluation system for AIDS-related KS for clinical practice and research.[1] After appropriate clinical examination and relevant interval imaging or endoscopy, patients are characterized as having complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD), based on the following criteria:
CR: No detectable residual disease and no tumor-related edema for at least 4 weeks.
PR: No new mucocutaneous lesions, visceral disease, or worsening tumor-related edema. Existing sites show a 50% reduction in (1) the number of lesions, (2) the form of lesions (i.e. flattening of raised lesions), and/or (3) the sum of the products of the largest perpendicular diameters of five measurable lesions. If residual tumor-related edema is present despite meeting CR criteria, the response is still characterized as a PR.
PD: Increase of more than 25% in (1) the size of existing lesions and/or (2) the number of existing lesions that have more nodular or plaque-like form, or the development of new sites of disease.
SD: No PR, CR, or PD.
References
Krown SE, Metroka C, Wernz JC: Kaposi’s sarcoma in the acquired immune deficiency syndrome: a proposal for uniform evaluation, response, and staging criteria. AIDS Clinical Trials Group Oncology Committee. J Clin Oncol 7 (9): 1201-7, 1989. [PUBMED Abstract]
Krown SE, Testa MA, Huang J: AIDS-related Kaposi’s sarcoma: prospective validation of the AIDS Clinical Trials Group staging classification. AIDS Clinical Trials Group Oncology Committee. J Clin Oncol 15 (9): 3085-92, 1997. [PUBMED Abstract]
Treatment of Classic and Endemic Kaposi Sarcoma
Classic Kaposi sarcoma (KS), as well as endemic KS in adult patients, is usually limited to the skin and has an indolent course. Thus, management for both is typically similar. Patients are predisposed to develop a second primary malignancy, and the treating physician should consider this factor when arranging a schedule of follow-up treatment for the patient.
Treatment Options for Localized Classic and Endemic Kaposi Sarcoma
Treatment options for localized skin disease include (options are equivalent):
For solitary lesions or lesions of limited extent, modest doses of radiation applied with a limited margin provide excellent control of disease in the treated area. Usually, superficial radiation beams, such as electron beams, are used. Some authors believe disease recurrence in adjacent untreated skin is common if only involved-field radiation therapy is used and claim better cure rates when extended-field radiation therapy is used.[1]
For low-voltage (100 kv) photon radiation therapy, 8 Gy to 10 Gy is given as a single dose or 15 Gy to 20 Gy is given over 1 week because solitary lesions control nearly 100% of local disease, but recurrence in adjacent areas is common.
For electron-beam radiation therapy (EBRT), 4 Gy is given once weekly for 6 to 8 consecutive weeks with a 4-MeV to 6-MeV electron beam. Ports should include the entire skin surface 15 cm above the lesion.
Surgery
Surgical excision may benefit patients with small superficial lesions, but local recurrence is likely to occur. However, multiple small excisions can continue to be performed for good disease control.
Other options
Based on extent and accessibility of lesions, alternate modalities such as cryo-, laser, intralesional, and topical therapy can be used. Use of these modalities is based on evidence extrapolated from treatment of AIDS-associated KS.[2,3]
Treatment Options for Advanced Classic and Endemic Kaposi Sarcoma
Treatment options for advanced skin disease include:
Modest doses can be effective in controlling widespread skin disease. The type of radiation (i.e., photon vs. electron) and fields used must be tailored to suit the distribution of disease in the individual patient.[1] Radiation therapy options include:
Extended-field EBRT.
For disease limited to areas distal to the knee, subtotal-skin EBRT directed to skin below the umbilicus.
For disease that extends above the knee, total-skin EBRT.
EBRT used in this manner gave long-term results that were superior to those obtained with radiation therapy administered to successive individual lesions as they appeared.[4]
For extensive disease, EBRT 4 Gy given once weekly for 6 to 8 consecutive weeks, and subtotal- or total-skin radiation therapy.
Chemotherapy
Because classic KS is such a rare disease in the United States, and is usually treated initially with radiation therapy, few patients have been treated with chemotherapy. Its use in classic KS is based on data extrapolated from treatment of AIDS-associated KS, and no randomized prospective trials have compared one agent with another in classic KS. The agents listed below have potential benefit.
Pegylated liposomal doxorubicin (PLD)
PLD has shown activity in several case series and single-institution analyses.[5–8]
Evidence (PLD):
A multicenter trial included 55 patients with classic KS who were treated over a decade.[5]
A 71% overall response rate was seen using PLD, with a median response duration of 25 months.[5][Level of evidence C3]
Taxanes
Paclitaxel has shown activity in both AIDS-associated and classic KS in small case series.[9–12]
Evidence (taxanes):
A small trial included 73 patients with AIDS-associated KS (32% had an undetectable HIV viral load). Patients were randomly assigned to receive either PLD or paclitaxel.[9]
Response rates were 46% for patients who received PLD and 56% for patients who received paclitaxel. The median progression-free survival (PFS) was 12 months for patients who received PLD and 18 months for patients who received paclitaxel. The 2-year overall survival rates were 78% for patients who received PLD and 79% for patients who received paclitaxel.[9][Level of evidence B3]
Other chemotherapy agents
Single-agent vinblastine [13–16], oral etoposide [17–19], and gemcitabine [20–22] have all shown good activity in classic and AIDS-associated KS.
Evidence (other chemotherapy agents):
A phase III trial included 65 patients with classic KS. Patients were randomly assigned to receive either oral etoposide or vinblastine.[18]
Response rates were relatively high and did not significantly differ (58% for patients who received PLD and 74% for patients who received paclitaxel).[18][Level of evidence B3]
Biological and targeted therapy
Agents that modulate the immune system, such as imide drugs and interferon alfa-2b, have shown efficacy in both classic and AIDS-associated KS.
Pomalidomide
The U.S. Food and Drug Administration (FDA) approved pomalidomide for the treatment of KS in patients with and without HIV.
Evidence (pomalidomide):
A phase I/II study of pomalidomide included 28 patients with KS. Ten patients were HIV-positive and 18 patients were HIV-negative.[23]
The overall response rate was 71%, and 80% among patients without HIV. The median PFS was 10 months.[23]
Pomalidomide was generally well tolerated. Common adverse events included neutropenia, anemia, fatigue, constipation, and rash. There were few grade 3 events (neutropenia, infection, and edema).[24]
Pomalidomide is teratogenic, prescribed through a Risk Evaluation and Mitigation Strategy (REMS) program, and it should be given with aspirin to mitigate venous thromboembolism risk.
Interferon alfa-2b
The FDA approved interferon alfa-2b for the treatment of AIDS-associated KS. It is sometimes used off-label for classic KS.
Evidence (interferon alfa-2b):
A small case series included 16 patients without HIV.[25]
Treatment with interferon alfa-2b led to a response in ten patients (one complete response [CR], nine partial responses [PRs]).
Immunotherapy
Immune checkpoint inhibitor therapy has been tested in classic KS and yielded promising results.
Pembrolizumab monotherapy
Evidence (pembrolizumab monotherapy):
Pembrolizumab monotherapy (given every 3 weeks for up to 6 months) was evaluated in a multicenter, single-arm, phase II study of 17 patients. Eight patients had classic KS and nine patients had endemic KS.[26][Level of evidence C3]
At a median follow-up of 20.4 months, with 88% of patients completing 6 months of treatment, the best overall response rate was 71% (12% with CR, 59% with PR). An additional 29% of patients had stable disease based on AIDS Clinical Trials Group response evaluation.
The median time to response was 5 months (interquartile range, 3.4–12), the estimated median duration of response was 23 months (95% confidence interval [CI], 21.2–not reached [NR]), and the median time to progression (TTP) was 24 months (95% CI, 15–NR).
Sixty-four percent (7 of 11) of pretreated patients and 83% (5 of 6) of chemotherapy-naïve patients had a CR or PR.
Pembrolizumab was generally well tolerated. Two patients (12%) had grade 3 events: acute cardiac decompensation and granulomatous reaction in the lung. Two patients discontinued treatment due to grade 2 pancreatitis and grade 3 acute cardiac decompensation, respectively.
Ipilimumab and nivolumab
Evidence (ipilimumab and nivolumab):
Ipilimumab and nivolumab combination therapy was evaluated in a phase II study of 18 patients with refractory classic KS. Patients received nivolumab 240 mg every 2 weeks and ipilimumab 1 mg/kg every 6 weeks until disease progression, for up to 24 months.[27]
At a median follow-up of 24.4 months, the overall response rate was 87% by Response Evaluation Criteria In Solid Tumors (RECIST) criteria. The 6- and 12-month PFS rates were 77% and 59%, respectively.[27][Level of evidence B4]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Tsao MN, Sinclair E, Assaad D, et al.: Radiation therapy for the treatment of skin Kaposi sarcoma. Ann Palliat Med 5 (4): 298-302, 2016. [PUBMED Abstract]
Epstein JB, Lozada-Nur F, McLeod WA, et al.: Oral Kaposi’s sarcoma in acquired immunodeficiency syndrome. Review of management and report of the efficacy of intralesional vinblastine. Cancer 64 (12): 2424-30, 1989. [PUBMED Abstract]
Bodsworth NJ, Bloch M, Bower M, et al.: Phase III vehicle-controlled, multi-centered study of topical alitretinoin gel 0.1% in cutaneous AIDS-related Kaposi’s sarcoma. Am J Clin Dermatol 2 (2): 77-87, 2001. [PUBMED Abstract]
Nisce LZ, Safai B, Poussin-Rosillo H: Once weekly total and subtotal skin electron beam therapy for Kaposi’s sarcoma. Cancer 47 (4): 640-4, 1981. [PUBMED Abstract]
Di Lorenzo G, Kreuter A, Di Trolio R, et al.: Activity and safety of pegylated liposomal doxorubicin as first-line therapy in the treatment of non-visceral classic Kaposi’s sarcoma: a multicenter study. J Invest Dermatol 128 (6): 1578-80, 2008. [PUBMED Abstract]
Castiñeiras I, Almagro M, Rodríguez-Lozano J, et al.: Disseminated classic Kaposi’s sarcoma. Two cases with excellent response to pegylated liposomal doxorubicin. J Dermatolog Treat 17 (6): 377-80, 2006. [PUBMED Abstract]
Di Lorenzo G, Di Trolio R, Montesarchio V, et al.: Pegylated liposomal doxorubicin as second-line therapy in the treatment of patients with advanced classic Kaposi sarcoma: a retrospective study. Cancer 112 (5): 1147-52, 2008. [PUBMED Abstract]
Potouridou I, Korfitis C, Ioannidou D, et al.: Low to moderate cumulative doses of pegylated liposomal doxorubicin in the treatment of classic Kaposi sarcoma in elderly patients with comorbidities. Br J Dermatol 158 (2): 431-2, 2008. [PUBMED Abstract]
Cianfrocca M, Lee S, Von Roenn J, et al.: Randomized trial of paclitaxel versus pegylated liposomal doxorubicin for advanced human immunodeficiency virus-associated Kaposi sarcoma: evidence of symptom palliation from chemotherapy. Cancer 116 (16): 3969-77, 2010. [PUBMED Abstract]
Brambilla L, Romanelli A, Bellinvia M, et al.: Weekly paclitaxel for advanced aggressive classic Kaposi sarcoma: experience in 17 cases. Br J Dermatol 158 (6): 1339-44, 2008. [PUBMED Abstract]
Chao SC, Lee JY, Tsao CJ: Treatment of classical type Kaposi’s sarcoma with paclitaxel. Anticancer Res 21 (1B): 571-3, 2001. [PUBMED Abstract]
Fardet L, Stoebner PE, Bachelez H, et al.: Treatment with taxanes of refractory or life-threatening Kaposi sarcoma not associated with human immunodeficiency virus infection. Cancer 106 (8): 1785-9, 2006. [PUBMED Abstract]
Solan AJ, Greenwald ES, Silvay O: Long-term complete remissions of Kaposi’s sarcoma with vinblastine therapy. Cancer 47 (4): 637-9, 1981. [PUBMED Abstract]
Tucker SB, Winkelmann RK: Treatment of Kaposi sarcoma with vinblastine. Arch Dermatol 112 (7): 958-61, 1976. [PUBMED Abstract]
Scott WP, Voight JA: Kaposi’s sarcoma. Management with vincaleucoblastine. Cancer 19 (4): 557-64, 1966. [PUBMED Abstract]
Klein E, Schwartz RA, Laor Y, et al.: Treatment of Kaposi’s sarcoma with vinblastine. Cancer 45 (3): 427-31, 1980. [PUBMED Abstract]
Tas F, Sen F, Keskin S, et al.: Oral etoposide as first-line therapy in the treatment of patients with advanced classic Kaposi’s sarcoma (CKS): a single-arm trial (oral etoposide in CKS). J Eur Acad Dermatol Venereol 27 (6): 789-92, 2013. [PUBMED Abstract]
Brambilla L, Labianca R, Boneschi V, et al.: Mediterranean Kaposi’s sarcoma in the elderly. A randomized study of oral etoposide versus vinblastine. Cancer 74 (10): 2873-8, 1994. [PUBMED Abstract]
Brambilla L, Labianca R, Ferrucci SM, et al.: Treatment of classical Kaposi’s sarcoma with gemcitabine. Dermatology 202 (2): 119-22, 2001. [PUBMED Abstract]
Zustovich F, Lombardi G, Pastorelli D: Important role of gemcitabine in the treatment of classic Kaposi’s sarcoma. Tumori 95 (4): 562-3, 2009. [PUBMED Abstract]
Zustovich F, Ferro A, Toso S: Gemcitabine for the treatment of classic Kaposi’s Sarcoma: a case series. Anticancer Res 33 (12): 5531-4, 2013. [PUBMED Abstract]
Ramaswami R, Polizzotto MN, Lurain K, et al.: Safety, Activity, and Long-term Outcomes of Pomalidomide in the Treatment of Kaposi Sarcoma among Individuals with or without HIV Infection. Clin Cancer Res 28 (5): 840-850, 2022. [PUBMED Abstract]
Polizzotto MN, Uldrick TS, Wyvill KM, et al.: Pomalidomide for Symptomatic Kaposi’s Sarcoma in People With and Without HIV Infection: A Phase I/II Study. J Clin Oncol 34 (34): 4125-4131, 2016. [PUBMED Abstract]
Costa da Cunha CS, Lebbe C, Rybojad M, et al.: Long-term follow-up of non-HIV Kaposi’s sarcoma treated with low-dose recombinant interferon alfa-2b. Arch Dermatol 132 (3): 285-90, 1996. [PUBMED Abstract]
Delyon J, Biard L, Renaud M, et al.: PD-1 blockade with pembrolizumab in classic or endemic Kaposi’s sarcoma: a multicentre, single-arm, phase 2 study. Lancet Oncol 23 (4): 491-500, 2022. [PUBMED Abstract]
Zer A, Icht O, Yosef L, et al.: Phase II single-arm study of nivolumab and ipilimumab (Nivo/Ipi) in previously treated classical Kaposi sarcoma (cKS). Ann Oncol 33 (7): 720-727, 2022. [PUBMED Abstract]
Treatment of AIDS-Associated Kaposi Sarcoma
Treatment of AIDS-associated Kaposi sarcoma (KS) may result in the following:
The disappearance or reduction in size of specific skin lesions, thereby alleviating the discomfort associated with the chronic edema and ulcerations that often accompany multiple skin tumors seen on the lower extremities.
Control of symptoms associated with mucosal or visceral lesions.
No data are available to show that treatment improves survival.[1] In addition to antitumor treatment, essential components of an optimal KS treatment strategy in this population include antiretroviral treatment, prophylaxis for opportunistic infections, and rapid recognition and treatment of intercurrent infections. Therefore, close collaboration between oncologists and HIV specialists is vital.
Most patients with good-risk disease, defined by the AIDS Clinical Trials Group as T0, show tumor regression with antiretroviral therapy alone.[2–4] Patients with poor-risk disease, defined as T1, usually require a combination of antiretroviral therapy and chemotherapy with discontinuation of the chemotherapy after disappearance of the skin lesion.[2–4]
Treatment Options for AIDS-Associated Kaposi Sarcoma
Small localized lesions of KS may be treated by electrodesiccation and curettage, cryotherapy, or by surgical excision. KS tumors are also generally very responsive to local radiation therapy, and excellent palliation has been obtained with doses at 20 Gy or slightly higher.[5,6] Radiation therapy is generally reserved to treat localized areas of the skin and oral cavity. It is used less often to control pulmonary, gastrointestinal tract, or other sites of KS lesions. Localized KS lesions have also been effectively treated with intralesional injections of vinblastine.[7] Alitretinoin 0.1% gel provided local control in a randomized, prospective, multicenter trial.[8][Level of evidence B3]
Chemotherapy
In AIDS-associated KS, the already profoundly depressed immunologic status of the patient limits the therapeutic usefulness of systemic chemotherapy. Systemic chemotherapy studies in patients with AIDS-associated KS have used doxorubicin, bleomycin, vinblastine, vincristine, etoposide, paclitaxel, and docetaxel alone or in combination.[9–13][Level of evidence C3] The combination of antiretroviral therapy and liposomal doxorubicin resulted in a 5-year overall survival rate of 85% in 140 patients with T1 disease.[3][Level of evidence C3]
Randomized multicenter trials showed an improvement in response rate (45%–60% vs. 20%–25%) and a more favorable toxic effects profile for pegylated liposomal doxorubicin (PLD) or liposomal daunorubicin, compared with the combination of doxorubicin, bleomycin, and vincristine or bleomycin and vincristine.[14–16][Level of evidence B3] During antiretroviral therapy, both PLD and paclitaxel are active single agents with response rates close to 50%.[17][Level of evidence B3]
Biological and targeted therapy
Interferon alfa
The interferon alfas have also been widely studied and show a 40% objective response rate in patients with AIDS-associated KS.[18,19] In these reports, the responses differed significantly according to the following prognostic factors:
Extent of disease.
Prior or coexistent opportunistic infections.
Prior treatment with chemotherapy.
CD4 lymphocyte counts lower than 200 cells/µL.
Presence of circulating acid-labile interferon alfa.
Increase in beta-2-microglobulin.
Several treatment studies have combined interferon alfa with other chemotherapeutic agents. Overall, these trials have shown no benefit with the interferon-chemotherapy combinations as compared with the single-agent activities.
Recombinant interferon alfa-2a and recombinant interferon alfa-2b were the first agents approved for the treatment of KS. Approval was based on single-agent studies performed in the 1980s before the advent of antiretroviral therapy. The early studies demonstrated improved efficacy at relatively high doses.
High-dose monotherapy is rarely used today, and instead, interferon is given in combination with other anti-HIV drugs in doses of 4 to 18 million units. Neutropenia is dose limiting, and trials of doses of 1 to 10 million units combined with less myelosuppressive antiretroviral agents are in progress. Response to interferon is slow, and the maximum effect is seen after 6 or more months. Interferon should probably not be used to treat patients with rapidly progressive, symptomatic KS.
Imatinib
Imatinib is a c-kit/platelet-derived growth factor receptor inhibitor.
Evidence (imatinib):
A phase II trial included 30 patients with AIDS-associated KS.[20]
A partial response (PR) was seen in 10 of 30 previously treated patients . Previous treatment included antiretroviral therapy and chemotherapy.
Bevacizumab
Bevacizumab is a humanized, anti–vascular endothelial growth factor monoclonal antibody.
Evidence (bevacizumab):
A phase II study included 16 assessable patients with KS and HIV. Patients received bevacizumab intravenously (IV) on days 1 and 8 and then every 3 weeks.[21]
There was a response in 5 of 16 patients. These patients had not improved after prior antiretroviral therapy and chemotherapy.[21][Level of evidence C3]
Interleukin-12
Evidence (interleukin-12):
A phase I and phase II trial included 24 evaluable patients. Patients received interleukin-12 subcutaneously twice weekly.[22]
Treatment resulted in a response rate of 71% (95% confidence interval, 48%–89%).[22][Level of evidence C3]
Pomalidomide
The U.S. Food and Drug Administration approved pomalidomide for patients with AIDS-associated KS.
Evidence (pomalidomide):
A phase I/II study of pomalidomide included 28 patients with KS. Ten patients were HIV-positive and 18 patients were HIV-negative.[23]
The overall response rate was 71%, and 80% among patients without HIV. The median progression-free survival was 10 months.[23]
Pomalidomide was generally well tolerated. Common adverse events included neutropenia, anemia, fatigue, constipation, and rash. There were few grade 3 events (neutropenia, infection, and edema).[24]
Pomalidomide is teratogenic, prescribed through a Risk Evaluation and Mitigation Strategy (REMS) program, and it should be given with aspirin to mitigate venous thromboembolism risk.
Bortezomib
Evidence (bortezomib):
Bortezomib was evaluated in a small phase I dose-escalation study of 17 patients with AIDS-associated KS. Patients received bortezomib IV on days 1, 8, and 15 of 28-day cycles.[25]
Bortezomib was relatively well tolerated and led to a PR in 60% of all evaluable patients. Among those receiving the highest bortezomib dose, 83% of patients had a PR and 17% had stable disease.
Management of Immune Reconstitution Inflammatory Syndrome in AIDS-Associated Kaposi Sarcoma
Immune reconstitution inflammatory syndrome (IRIS) is a hyperimmune response in patients with HIV/AIDS within the first 6 months of starting antiretroviral therapy. Kaposi sarcoma (KS)-associated IRIS (KS-IRIS) is not well-defined, but is considered to be the sudden clinical worsening of previous KS (“paradoxical”) or the new presentation of KS (“unmasked”) in close proximity to starting or modifying antiretroviral therapy.[26]
Estimates for KS-IRIS incidence vary from 2% to 39%, with the highest risk in patients with any of the following characteristics:[26,27]
High HIV viral loads (>10,000 copies/mL).
CD4 lymphocyte count less than 200 cells/µL.
Detectable plasma human herpesvirus 8 DNA.
Pulmonary KS involvement.
Recent steroid use.
KS-IRIS typically presents with increased swelling/tenderness of lesions, new or worsening edema, and visceral or pulmonary involvement.
Management of KS-IRIS typically includes continuing antiretroviral therapy and initiating systemic treatment, such as liposomal doxorubicin or paclitaxel, for KS. The evidence for use of chemotherapy to prevent KS-IRIS is mixed, but can be considered on an individual basis.[26] Glucocorticoids are avoided due to the risk of dramatic worsening of KS.[27,28] Thalidomide has also been used for steroid-refractory IRIS.[29]
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.
Krown SE: Highly active antiretroviral therapy in AIDS-associated Kaposi’s sarcoma: implications for the design of therapeutic trials in patients with advanced, symptomatic Kaposi’s sarcoma. J Clin Oncol 22 (3): 399-402, 2004. [PUBMED Abstract]
Bower M, Dalla Pria A, Coyle C, et al.: Prospective stage-stratified approach to AIDS-related Kaposi’s sarcoma. J Clin Oncol 32 (5): 409-14, 2014. [PUBMED Abstract]
Krell J, Stebbing J: Broader implications of a stage-guided stratified therapeutic approach for AIDS-related Kaposi’s sarcoma. J Clin Oncol 32 (5): 373-5, 2014. [PUBMED Abstract]
Singh NB, Lakier RH, Donde B: Hypofractionated radiation therapy in the treatment of epidemic Kaposi sarcoma–a prospective randomized trial. Radiother Oncol 88 (2): 211-6, 2008. [PUBMED Abstract]
Tsao MN, Sinclair E, Assaad D, et al.: Radiation therapy for the treatment of skin Kaposi sarcoma. Ann Palliat Med 5 (4): 298-302, 2016. [PUBMED Abstract]
Epstein JB, Lozada-Nur F, McLeod WA, et al.: Oral Kaposi’s sarcoma in acquired immunodeficiency syndrome. Review of management and report of the efficacy of intralesional vinblastine. Cancer 64 (12): 2424-30, 1989. [PUBMED Abstract]
Bodsworth NJ, Bloch M, Bower M, et al.: Phase III vehicle-controlled, multi-centered study of topical alitretinoin gel 0.1% in cutaneous AIDS-related Kaposi’s sarcoma. Am J Clin Dermatol 2 (2): 77-87, 2001. [PUBMED Abstract]
Evans SR, Krown SE, Testa MA, et al.: Phase II evaluation of low-dose oral etoposide for the treatment of relapsed or progressive AIDS-related Kaposi’s sarcoma: an AIDS Clinical Trials Group clinical study. J Clin Oncol 20 (15): 3236-41, 2002. [PUBMED Abstract]
Saville MW, Lietzau J, Pluda JM, et al.: Treatment of HIV-associated Kaposi’s sarcoma with paclitaxel. Lancet 346 (8966): 26-8, 1995. [PUBMED Abstract]
Lim ST, Tupule A, Espina BM, et al.: Weekly docetaxel is safe and effective in the treatment of advanced-stage acquired immunodeficiency syndrome-related Kaposi sarcoma. Cancer 103 (2): 417-21, 2005. [PUBMED Abstract]
Gill PS, Tulpule A, Espina BM, et al.: Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi’s sarcoma. J Clin Oncol 17 (6): 1876-83, 1999. [PUBMED Abstract]
Di Lorenzo G, Konstantinopoulos PA, Pantanowitz L, et al.: Management of AIDS-related Kaposi’s sarcoma. Lancet Oncol 8 (2): 167-76, 2007. [PUBMED Abstract]
Stewart S, Jablonowski H, Goebel FD, et al.: Randomized comparative trial of pegylated liposomal doxorubicin versus bleomycin and vincristine in the treatment of AIDS-related Kaposi’s sarcoma. International Pegylated Liposomal Doxorubicin Study Group. J Clin Oncol 16 (2): 683-91, 1998. [PUBMED Abstract]
Northfelt DW, Dezube BJ, Thommes JA, et al.: Pegylated-liposomal doxorubicin versus doxorubicin, bleomycin, and vincristine in the treatment of AIDS-related Kaposi’s sarcoma: results of a randomized phase III clinical trial. J Clin Oncol 16 (7): 2445-51, 1998. [PUBMED Abstract]
Gill PS, Wernz J, Scadden DT, et al.: Randomized phase III trial of liposomal daunorubicin versus doxorubicin, bleomycin, and vincristine in AIDS-related Kaposi’s sarcoma. J Clin Oncol 14 (8): 2353-64, 1996. [PUBMED Abstract]
Cianfrocca M, Lee S, Von Roenn J, et al.: Randomized trial of paclitaxel versus pegylated liposomal doxorubicin for advanced human immunodeficiency virus-associated Kaposi sarcoma: evidence of symptom palliation from chemotherapy. Cancer 116 (16): 3969-77, 2010. [PUBMED Abstract]
Real FX, Oettgen HF, Krown SE: Kaposi’s sarcoma and the acquired immunodeficiency syndrome: treatment with high and low doses of recombinant leukocyte A interferon. J Clin Oncol 4 (4): 544-51, 1986. [PUBMED Abstract]
Groopman JE, Gottlieb MS, Goodman J, et al.: Recombinant alpha-2 interferon therapy for Kaposi’s sarcoma associated with the acquired immunodeficiency syndrome. Ann Intern Med 100 (5): 671-6, 1984. [PUBMED Abstract]
Koon HB, Krown SE, Lee JY, et al.: Phase II trial of imatinib in AIDS-associated Kaposi’s sarcoma: AIDS Malignancy Consortium Protocol 042. J Clin Oncol 32 (5): 402-8, 2014. [PUBMED Abstract]
Uldrick TS, Wyvill KM, Kumar P, et al.: Phase II study of bevacizumab in patients with HIV-associated Kaposi’s sarcoma receiving antiretroviral therapy. J Clin Oncol 30 (13): 1476-83, 2012. [PUBMED Abstract]
Little RF, Pluda JM, Wyvill KM, et al.: Activity of subcutaneous interleukin-12 in AIDS-related Kaposi sarcoma. Blood 107 (12): 4650-7, 2006. [PUBMED Abstract]
Ramaswami R, Polizzotto MN, Lurain K, et al.: Safety, Activity, and Long-term Outcomes of Pomalidomide in the Treatment of Kaposi Sarcoma among Individuals with or without HIV Infection. Clin Cancer Res 28 (5): 840-850, 2022. [PUBMED Abstract]
Polizzotto MN, Uldrick TS, Wyvill KM, et al.: Pomalidomide for Symptomatic Kaposi’s Sarcoma in People With and Without HIV Infection: A Phase I/II Study. J Clin Oncol 34 (34): 4125-4131, 2016. [PUBMED Abstract]
Reid EG, Suazo A, Lensing SY, et al.: Pilot Trial AMC-063: Safety and Efficacy of Bortezomib in AIDS-associated Kaposi Sarcoma. Clin Cancer Res 26 (3): 558-565, 2020. [PUBMED Abstract]
Fernández-Sánchez M, Iglesias MC, Ablanedo-Terrazas Y, et al.: Steroids are a risk factor for Kaposi’s sarcoma-immune reconstitution inflammatory syndrome and mortality in HIV infection. AIDS 30 (6): 909-14, 2016. [PUBMED Abstract]
Volkow PF, Cornejo P, Zinser JW, et al.: Life-threatening exacerbation of Kaposi’s sarcoma after prednisone treatment for immune reconstitution inflammatory syndrome. AIDS 22 (5): 663-5, 2008. [PUBMED Abstract]
Brunel AS, Reynes J, Tuaillon E, et al.: Thalidomide for steroid-dependent immune reconstitution inflammatory syndromes during AIDS. AIDS 26 (16): 2110-2, 2012. [PUBMED Abstract]
Treatment of Transplant-Related Kaposi Sarcoma
Treatment Options for Transplant-Related Kaposi Sarcoma
In general, transplant-related Kaposi sarcoma is effectively managed by reduction in immunosuppression and does not require systemic treatment. Transitioning immunosuppression therapy to an mTOR inhibitor, such as sirolimus, has demonstrated efficacy in small studies and can be considered.[1]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Stallone G, Schena A, Infante B, et al.: Sirolimus for Kaposi’s sarcoma in renal-transplant recipients. N Engl J Med 352 (13): 1317-23, 2005. [PUBMED Abstract]
Latest Updates to This Summary (05/07/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Editorial changes were made to this summary.
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of Kaposi sarcoma. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Kaposi Sarcoma Treatment are:
Eric J. Seifter, MD (Johns Hopkins University)
Minh Tam Truong, MD (Boston University Medical Center)
Vinayak Venkataraman, MD (Dana Farber Cancer Institute)
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Kaposi Sarcoma Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/soft-tissue-sarcoma/hp/kaposi-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389335]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
We offer evidence-based supportive and palliative care information for health professionals on the assessment and management of cancer-related symptoms and conditions.
Screening With the Papanicolaou (Pap) Test: Benefits
Based on solid evidence, regular screening for cervical cancer with the Pap test in an appropriate population of women reduces mortality from cervical cancer. The benefits of screening women younger than 21 years are small because of the low prevalence of lesions that will progress to invasive cancer. Screening is not beneficial in women older than 65 years if they have had a recent history of negative test results.[1–3]
Magnitude of Effect: Regular Pap screening decreases cervix cancer incidence and mortality by at least 80%.
Study Design: Population-based and cohort studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Screening With the Pap Test: Harms
Based on solid evidence, regular screening with the Pap test leads to additional diagnostic procedures (e.g., colposcopy) and possible overtreatment for low-grade squamous intraepithelial lesions (LSILs). These harms are greatest for younger women, who have a higher prevalence of LSILs, lesions that often regress without treatment. Harms are also increased in younger women because they have a higher rate of false-positive results. Excisional procedures to treat preinvasive disease has been associated with increased risk of long-term consequences for fertility and pregnancy.[4]
Magnitude of Effect: Additional diagnostic procedures were performed in 50% of women undergoing regular Pap testing. Approximately 5% were treated for LSILs. The number of women with impaired fertility and pregnancy complications is unknown.
Study Design: Evidence obtained from cohort or case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Screening With the Human Papillomavirus (HPV) DNA Test: Benefits
Based on solid evidence, screening with an HPV DNA or HPV RNA test detects high-grade cervical dysplasia, a precursor lesion for cervical cancer. Additional clinical trials show that HPV testing is superior to other cervical cancer screening strategies. In April 2014, the U.S. Food and Drug Administration approved an HPV DNA test that can be used alone for the primary screening of cervical cancer risk in women aged 25 years and older.[5]
Magnitude of Effect: In one prospective, clustered, randomized trial, HPV testing was superior to other strategies for preventing cervical cancer mortality.[6,7]
Study Design: Clustered randomized controlled trial (RCT).
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Screening With the HPV DNA Test: Harms
Based on solid evidence, HPV testing identifies numerous infections that will not lead to cervical dysplasia or cervical cancer. This is especially true in women younger than 30 years, in whom rates of HPV infection may be higher.
Magnitude of Effect: In one study, 86.7% of women with a positive HPV test did not develop cervical cancer or related premalignant disease after more than a decade of follow-up.[8]
Study Design: Long-term observational trials.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Screening With the Pap Test and the HPV DNA Test (Cotesting): Benefits
Based on solid evidence, screening every 5 years with the Pap test and the HPV DNA test (cotesting) in women aged 30 years and older is more sensitive in detecting cervical abnormalities, compared with the Pap test alone. Screening with the Pap test and HPV DNA test reduces the incidence of cervical cancer.[3]
Magnitude of Effect: HPV-based screening provides 60% to 70% greater protection against invasive cervical carcinoma, compared with cytology.[9]
Study Design: RCTs.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Screening With the Pap Test and the HPV DNA Test (Cotesting): Harms
Based on solid evidence, HPV and Pap cotesting is associated with more false-positives than is the Pap test alone. Abnormal test results can lead to more frequent testing and invasive diagnostic procedures.[3]
Magnitude of Effect: The percentage of U.S. women undergoing cotesting who will have a normal cytology test result and a positive HPV test result (and who will therefore require additional testing) ranges from 11% among women aged 30 to 34 years to 2.6% among women aged 60 to 65 years.[3]
Study Design: RCTs.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Screening Women Without a Cervix
Based on solid evidence, screening is not helpful in women who do not have a cervix as a result of a hysterectomy for a benign condition.
Magnitude of Effect: Among women without cervices, fewer than 1 per 1,000 had abnormal Pap test results.
Study Design: Evidence obtained from a single cohort study.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
References
Sasieni P, Castanon A, Cuzick J: Effectiveness of cervical screening with age: population based case-control study of prospectively recorded data. BMJ 339: b2968, 2009. [PUBMED Abstract]
Sawaya GF, McConnell KJ, Kulasingam SL, et al.: Risk of cervical cancer associated with extending the interval between cervical-cancer screenings. N Engl J Med 349 (16): 1501-9, 2003. [PUBMED Abstract]
Moyer VA; U.S. Preventive Services Task Force: Screening for cervical cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 156 (12): 880-91, W312, 2012. [PUBMED Abstract]
Kyrgiou M, Athanasiou A, Paraskevaidi M, et al.: Adverse obstetric outcomes after local treatment for cervical preinvasive and early invasive disease according to cone depth: systematic review and meta-analysis. BMJ 354: i3633, 2016. [PUBMED Abstract]
Wright TC, Stoler MH, Behrens CM, et al.: Primary cervical cancer screening with human papillomavirus: end of study results from the ATHENA study using HPV as the first-line screening test. Gynecol Oncol 136 (2): 189-97, 2015. [PUBMED Abstract]
Sankaranarayanan R, Nene BM, Shastri SS, et al.: HPV screening for cervical cancer in rural India. N Engl J Med 360 (14): 1385-94, 2009. [PUBMED Abstract]
Szarewski A: Cervical screening by visual inspection with acetic acid. Lancet 370 (9585): 365-6, 2007. [PUBMED Abstract]
Chen HC, Schiffman M, Lin CY, et al.: Persistence of type-specific human papillomavirus infection and increased long-term risk of cervical cancer. J Natl Cancer Inst 103 (18): 1387-96, 2011. [PUBMED Abstract]
Ronco G, Dillner J, Elfström KM, et al.: Efficacy of HPV-based screening for prevention of invasive cervical cancer: follow-up of four European randomised controlled trials. Lancet 383 (9916): 524-32, 2014. [PUBMED Abstract]
Natural History, Incidence, and Mortality
It is estimated that 13,360 cases of invasive cervical cancer will be diagnosed and that 4,320 women will die of the disease in the United States in 2025.[1] Cervical cancer incidence and mortality rates have steadily improved over time and have stabilized in recent years, attributed largely to screening with Papanicolaou (Pap) test. However, the mortality rate in Black women is 50% higher and in Native American women is 70% higher than in White women.[1] When corrected for the prevalence of hysterectomy, the mortality rate for Black women is nearly twice the mortality rate for White women.[2]
Invasive squamous carcinoma of the cervix results from the progression of preinvasive precursor lesions called cervical intraepithelial neoplasia (CIN), or dysplasia. CIN is histologically graded into mild dysplasia (CIN 1), moderate dysplasia (CIN 2), or severe dysplasia (CIN 3). Not all of these lesions progress to invasive cancer; many mild and moderate lesions regress. A further categorization, the Bethesda System, is based on cytologic findings: atypical squamous cells of undetermined significance (ASCUS) or cannot rule out low-grade squamous intraepithelial lesions (LSILs), LSILs (consisting of cytologic atypia and CIN 1), and high-grade squamous intraepithelial lesions (HSILs), primarily CIN 2–3 plus carcinoma in situ.[3]
The rate at which invasive cancer develops from CIN is usually slow, measured in years and perhaps decades.[4] This long natural history provides the opportunity for screening to effectively detect this process during the preinvasive phase, thus allowing early treatment and cure. Because many of these preinvasive lesions (especially LSILs) may never progress to invasive cancer,[5–7] screening also runs the risk of leading to treatment for women who may not need it.
Human papillomavirus (HPV) is an oncogenic virus and the etiologic agent of cervical cancer and related premalignant disease. HPV is transmitted by sexual contact. Sexually inactive women rarely develop cervical cancer, while sexual activity at an early age with multiple sexual partners is a strong risk factor.[8] Nearly all women with invasive cervical cancer have evidence of HPV infection.[9–12] Most women with HPV infection, however, never develop cervical cancer; thus, this infection is necessary but not sufficient for the development of cancer.[13]
Although cervical cancer mortality increases with age,[14] the prevalence of CIN is highest among women in their 20s and 30s. Mortality is rare among women younger than 30 years; HSILs are rare among women older than 65 years who have been previously screened. About 70% of atypical squamous cells of undetermined significance and CIN 1 lesions regress within 6 years, while about 6% of CIN 1 lesions progress to CIN 3 or worse. In about 10% to 20% of women with CIN 3 lesions, the lesions progress to invasive cancer.[4,7,15]
Historically, cervical cancer mortality rates were substantially higher (twice as high or more) in Black women than in White women younger than 50 years. However, recent rates (2018–2022) have been only modestly higher (17%) in Black women than in White women in this age group. Among women older than 60 years, cervical cancer mortality rates have historically been up to three times as high in Black women than in White women; recent rates (2018–2022) are still almost twice as high in Black women than in White women.[14] In either case, mortality is rare among women of any age who have regular screenings.
References
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
Beavis AL, Gravitt PE, Rositch AF: Hysterectomy-corrected cervical cancer mortality rates reveal a larger racial disparity in the United States. Cancer 123 (6): 1044-1050, 2017. [PUBMED Abstract]
Solomon D, Davey D, Kurman R, et al.: The 2001 Bethesda System: terminology for reporting results of cervical cytology. JAMA 287 (16): 2114-9, 2002. [PUBMED Abstract]
Holowaty P, Miller AB, Rohan T, et al.: Natural history of dysplasia of the uterine cervix. J Natl Cancer Inst 91 (3): 252-8, 1999. [PUBMED Abstract]
Nasiell K, Roger V, Nasiell M: Behavior of mild cervical dysplasia during long-term follow-up. Obstet Gynecol 67 (5): 665-9, 1986. [PUBMED Abstract]
Melnikow J, Nuovo J, Willan AR, et al.: Natural history of cervical squamous intraepithelial lesions: a meta-analysis. Obstet Gynecol 92 (4 Pt 2): 727-35, 1998. [PUBMED Abstract]
Berrington de González A, Green J; International Collaboration of Epidemiological Studies of Cervical Cancer: Comparison of risk factors for invasive squamous cell carcinoma and adenocarcinoma of the cervix: collaborative reanalysis of individual data on 8,097 women with squamous cell carcinoma and 1,374 women with adenocarcinoma from 12 epidemiological studies. Int J Cancer 120 (4): 885-91, 2007. [PUBMED Abstract]
Bosch FX, Manos MM, Muñoz N, et al.: Prevalence of human papillomavirus in cervical cancer: a worldwide perspective. International biological study on cervical cancer (IBSCC) Study Group. J Natl Cancer Inst 87 (11): 796-802, 1995. [PUBMED Abstract]
Wallin KL, Wiklund F, Angström T, et al.: Type-specific persistence of human papillomavirus DNA before the development of invasive cervical cancer. N Engl J Med 341 (22): 1633-8, 1999. [PUBMED Abstract]
Alani RM, Münger K: Human papillomaviruses and associated malignancies. J Clin Oncol 16 (1): 330-7, 1998. [PUBMED Abstract]
Walboomers JM, Jacobs MV, Manos MM, et al.: Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 189 (1): 12-9, 1999. [PUBMED Abstract]
Ho GY, Bierman R, Beardsley L, et al.: Natural history of cervicovaginal papillomavirus infection in young women. N Engl J Med 338 (7): 423-8, 1998. [PUBMED Abstract]
Surveillance Research Program, National Cancer Institute: SEER*Explorer: An interactive website for SEER cancer statistics. Bethesda, MD: National Cancer Institute. Available online. Last accessed December 30, 2024.
The Papanicolaou (Pap) test has never been examined in a randomized controlled trial. A large body of consistent observational data, however, supports its effectiveness in reducing mortality from cervical cancer. Both incidence and mortality from cervical cancer have sharply decreased in a number of large populations after the introduction of well-run screening programs.[1–4] In Iceland, the mortality rate declined by 80% for more than 20 years, and in Finland and Sweden by 50% and 34%, respectively.[1,5] Similar reductions have been observed in large populations in the United States and Canada. Reductions in cervical cancer incidence and mortality were proportional to the intensity of screening.[1,5] Mortality in the Canadian provinces was reduced most remarkably in British Columbia, which had screening rates two to five times those of the other provinces.[6]
Case-control studies have found that the risk of developing invasive cervical cancer is three to ten times higher in women who have not been screened.[7–10] Risk also increases with long duration after the last normal Pap test, or similarly, with decreasing frequency of screening.[11,12] Screening every 2 to 3 years, however, has not been found to significantly increase the risk of finding invasive cervical cancer above the risk expected with annual screening.[12–14]
References
Lăără E, Day NE, Hakama M: Trends in mortality from cervical cancer in the Nordic countries: association with organised screening programmes. Lancet 1 (8544): 1247-9, 1987. [PUBMED Abstract]
Christopherson WM, Lundin FE, Mendez WM, et al.: Cervical cancer control: a study of morbidity and mortality trends over a twenty-one-year period. Cancer 38 (3): 1357-66, 1976. [PUBMED Abstract]
Miller AB, Lindsay J, Hill GB: Mortality from cancer of the uterus in Canada and its relationship to screening for cancer of the cervix. Int J Cancer 17 (5): 602-12, 1976. [PUBMED Abstract]
Johannesson G, Geirsson G, Day N: The effect of mass screening in Iceland, 1965-74, on the incidence and mortality of cervical carcinoma. Int J Cancer 21 (4): 418-25, 1978. [PUBMED Abstract]
Sigurdsson K: Effect of organized screening on the risk of cervical cancer. Evaluation of screening activity in Iceland, 1964-1991. Int J Cancer 54 (4): 563-70, 1993. [PUBMED Abstract]
Benedet JL, Anderson GH, Matisic JP: A comprehensive program for cervical cancer detection and management. Am J Obstet Gynecol 166 (4): 1254-9, 1992. [PUBMED Abstract]
Aristizabal N, Cuello C, Correa P, et al.: The impact of vaginal cytology on cervical cancer risks in Cali, Colombia. Int J Cancer 34 (1): 5-9, 1984. [PUBMED Abstract]
Clarke EA, Anderson TW: Does screening by “Pap” smears help prevent cervical cancer? A case-control study. Lancet 2 (8132): 1-4, 1979. [PUBMED Abstract]
La Vecchia C, Franceschi S, Decarli A, et al.: “Pap” smear and the risk of cervical neoplasia: quantitative estimates from a case-control study. Lancet 2 (8406): 779-82, 1984. [PUBMED Abstract]
Herrero R, Brinton LA, Reeves WC, et al.: Screening for cervical cancer in Latin America: a case-control study. Int J Epidemiol 21 (6): 1050-6, 1992. [PUBMED Abstract]
Celentano DD, Klassen AC, Weisman CS, et al.: Duration of relative protection of screening for cervical cancer. Prev Med 18 (4): 411-22, 1989. [PUBMED Abstract]
Screening for squamous cervical cancer: duration of low risk after negative results of cervical cytology and its implication for screening policies. IARC Working Group on evaluation of cervical cancer screening programmes. Br Med J (Clin Res Ed) 293 (6548): 659-64, 1986. [PUBMED Abstract]
Kleinman JC, Kopstein A: Who is being screened for cervical cancer? Am J Public Health 71 (1): 73-6, 1981. [PUBMED Abstract]
Qin J, Saraiya M, Martinez G, et al.: Prevalence of Potentially Unnecessary Bimanual Pelvic Examinations and Papanicolaou Tests Among Adolescent Girls and Young Women Aged 15-20 Years in the United States. JAMA Intern Med 180 (2): 274-280, 2020. [PUBMED Abstract]
Accuracy of the Pap Test
Ideally, determining the sensitivity and specificity of a screening test would involve a study that applies a gold standard test (such as colposcopy with appropriate biopsy) to all participants (whether the screening test results are positive or negative). Sensitivity (the percentage of true-positive cases that are detected by the screening test) and specificity (the percentage of true-negative cases that are negative by the screening test) could be calculated. Such studies have rarely been done for any screening test for cervical cancer. Studies that compare the Pap test with repeat Pap testing have found that the sensitivity of any abnormality on a single test for detecting high-grade lesions is 55% to 80%.[1,2] Because of the usual slow-growing nature of cervical cancer, the sensitivity of a program of regular Pap testing is likely higher.
To determine the sensitivity and specificity of the Pap smear, both a test threshold (i.e., the point at which the test will be considered to be positive) and a reference-standard threshold (i.e., the point at which the reference standard is considered to be positive) must be defined. In practice, atypical squamous cells of undetermined significance (ASCUS) are often used as the test threshold, and CIN 1 is often used as the reference threshold. This combination gives a sensitivity of about 68% and a specificity of about 75%. A more appropriate test threshold may be LSIL, with a reference threshold of CIN 2–3. This combination gives a sensitivity of 70% to 80%, with a specificity of about 95%.[3]
One important factor in the accuracy of the Pap test is the adequacy of the specimen obtained. Adequate training and using techniques such as the cytobrush may improve sensitivity.[4]
References
Soost HJ, Lange HJ, Lehmacher W, et al.: The validation of cervical cytology. Sensitivity, specificity and predictive values. Acta Cytol 35 (1): 8-14, 1991 Jan-Feb. [PUBMED Abstract]
Benoit AG, Krepart GV, Lotocki RJ: Results of prior cytologic screening in patients with a diagnosis of Stage I carcinoma of the cervix. Am J Obstet Gynecol 148 (5): 690-4, 1984. [PUBMED Abstract]
Nanda K, McCrory DC, Myers ER, et al.: Accuracy of the Papanicolaou test in screening for and follow-up of cervical cytologic abnormalities: a systematic review. Ann Intern Med 132 (10): 810-9, 2000. [PUBMED Abstract]
Martin-Hirsch P, Lilford R, Jarvis G, et al.: Efficacy of cervical-smear collection devices: a systematic review and meta-analysis. Lancet 354 (9192): 1763-70, 1999. [PUBMED Abstract]
Newer Screening Technologies
Newer techniques that employ liquid-based cytology (LBC) (e.g., ThinPrep) have been developed to improve the sensitivity of screening. In 1996, the ThinPrep® Papanicolaou (Pap) test became the first LBC approved by the U.S. Food and Drug Administration.[1] As with the Pap test, the optimal studies to determine the sensitivity and specificity of these technologies have not been conducted. Some less-than-optimal studies show that sensitivity is modestly higher for detecting any degree of cervical intraepithelial neoplasia, with modestly lower specificity.[2,3] One study, however, showed that conventional Pap testing was slightly more sensitive and specific than LBC.[4]
The evidence is also mixed about whether liquid-based techniques improve rates of test adequacy.[2,3] One advantage of LBC is that HPV testing can be performed on the same preparation; one disadvantage is that liquid-based approaches are more expensive than conventional Pap testing. No study has examined whether LBC actually reduces the number of women dying of cervical cancer compared with conventional Pap testing.
References
Gibb RK, Martens MG: The impact of liquid-based cytology in decreasing the incidence of cervical cancer. Rev Obstet Gynecol 4 (Suppl 1): S2-S11, 2011. [PUBMED Abstract]
Hartmann KE, Hall SA, Nanda K, et al.: Screening for Cervical Cancer. Rockville, Md: Agency for Health Research and Quality, 2002. Available online. Last accessed December 18, 2024.
McCrory DC, Matchar DB, Bastian L, et al.: Evaluation of Cervical Cytology. Rockville, Md: Agency for Health Research and Quality, 1999. Evidence Report/Technology Assessment No. 5. AHCPR Publication No. 99-E010. Also available online. Last accessed December 18, 2024.
Coste J, Cochand-Priollet B, de Cremoux P, et al.: Cross sectional study of conventional cervical smear, monolayer cytology, and human papillomavirus DNA testing for cervical cancer screening. BMJ 326 (7392): 733, 2003. [PUBMED Abstract]
Screening Women Who Have Had a Hysterectomy
Women who have had a hysterectomy with removal of the cervix for benign disease rarely have important abnormalities found on Pap testing. Several studies have shown that the rate of high-grade vaginal lesions or vaginal cancer is less than 1 in 1,000 tests;[1,2] no study has shown that screening for vaginal cancer reduces mortality from this rare condition.
References
Fox J, Remington P, Layde P, et al.: The effect of hysterectomy on the risk of an abnormal screening Papanicolaou test result. Am J Obstet Gynecol 180 (5): 1104-9, 1999. [PUBMED Abstract]
Pearce KF, Haefner HK, Sarwar SF, et al.: Cytopathological findings on vaginal Papanicolaou smears after hysterectomy for benign gynecologic disease. N Engl J Med 335 (21): 1559-62, 1996. [PUBMED Abstract]
Screening Interval
Because cervical cancer is slow growing, there is considerable uncertainty about the optimal screening interval. The most direct evidence about this issue comes from a prospective cohort analysis of a randomized controlled trial.[1] Among 2,561 women (mean age, 66.7 years) with normal Pap tests at baseline, 110 had an abnormal Pap test within the next 2 years. No woman was found to have cervical intraepithelial neoplasia (CIN) 2–3 or invasive cancer, and only one woman had CIN 1–2. Thus, the positive predictive value (PPV) of screening 1 year after a negative Pap test was 0%; after 2 years, the PPV was 0.9%. The authors concluded that Pap tests should not be repeated within 2 years of a negative test. A large (N = 332,000) prospective cohort study of cervical cytology and human papillomavirus DNA cotesting in U.S. women aged 30 years and older found that a negative Pap smear was associated with a low risk of developing CIN 3 or cancer (CIN 3+) for up to 5 years after the test (cumulative incidence of CIN 3+ at 3 and 5 years was 0.17% and 0.36%, respectively).[2]
A large study that included data from the National Breast and Cervical Cancer Early Detection Program together with modeling found little further mortality reduction from cervical cancer for screening every year as compared with screening every 3 years.[3] A similar modeling study from Australia found no differences between screening every 2 years and screening every 3 years.[4]
References
Sawaya GF, Grady D, Kerlikowske K, et al.: The positive predictive value of cervical smears in previously screened postmenopausal women: the Heart and Estrogen/progestin Replacement Study (HERS). Ann Intern Med 133 (12): 942-50, 2000. [PUBMED Abstract]
Katki HA, Kinney WK, Fetterman B, et al.: Cervical cancer risk for women undergoing concurrent testing for human papillomavirus and cervical cytology: a population-based study in routine clinical practice. Lancet Oncol 12 (7): 663-72, 2011. [PUBMED Abstract]
Sawaya GF, McConnell KJ, Kulasingam SL, et al.: Risk of cervical cancer associated with extending the interval between cervical-cancer screenings. N Engl J Med 349 (16): 1501-9, 2003. [PUBMED Abstract]
Creighton P, Lew JB, Clements M, et al.: Cervical cancer screening in Australia: modelled evaluation of the impact of changing the recommended interval from two to three years. BMC Public Health 10: 734, 2010. [PUBMED Abstract]
HPV Testing
Noninvasive cervical squamous cell abnormalities are graded histologically as cervical intraepithelial neoplasia (CIN) mild dysplasia (CIN 1), moderate dysplasia (CIN 2), or severe dysplasia (CIN 3), according to the severity of the cell changes and the percent of the epithelium replaced by abnormal cell growth. CIN 3 is a reasonably reproducible diagnosis and, if untreated, has an approximate 30% risk of developing into invasive cancer over many years.[1] CIN 2 has poor interobserver reproducibility,[2] and the biological behavior is variable.[3] CIN 3 is therefore a more rigorous end point for clinical trials, while CIN 2 represents the threshold for treatment to provide an additional measure of safety.
Approximately 15 cancer-associated (high-risk or carcinogenic) HPV genotypes cause virtually all cases of cervical cancer and precursor lesions of CIN 2 and CIN 3. However, carcinogenic HPV infections are very common, particularly in young women, and most infections clear on their own within 1 to 2 years. Therefore, the challenge of incorporating HPV testing in cervical screening programs is to balance sensitivity for detection of CIN 2 or CIN 2+ and to minimize the over-referral of women with transient HPV infections and cervical changes that are destined to regress.
The U.S. Food and Drug Administration has approved several HPV tests. Most of these tests are based on the detection of DNA from one or more oncogenic types of HPV. One test detects HPV RNA. HPV testing is approved for use in two contexts: (1) as a second (i.e., triage) test after an equivocal cytology result of atypical squamous cells of undetermined significance (ASCUS); and (2) for primary screening in conjunction with cervical cytology for women aged 30 years and older.[4] Testing for low-risk HPV types does not identify women at risk of developing CIN 2 or 3.[5,6]
Triage
A large randomized clinical trial, the ASCUS/low-grade squamous intraepithelial lesion (LSIL) Triage Study (ALTS), demonstrated the cost-effectiveness of using HPV testing to clarify the risk of an ASCUS Pap result.[7] ALTS randomly assigned women with ASCUS to one of three management strategies: immediate colposcopy regardless of enrollment test results, referral to colposcopy if HPV test results were positive or if the enrollment cytology was high-grade squamous intraepithelial lesion (HSIL), and referral to colposcopy only if the cytology was HSIL. The HPV triage strategy was as sensitive as immediate colposcopy to detection CIN 2+, while referring only about half of the women for the procedure. Repeat cytology with referral to colposcopy at the threshold of HSIL was less sensitive for CIN 3+ (60%) compared with HPV triage (92%); however, using a cytologic threshold of ASCUS for referral increased sensitivity but resulted in 72% of women with ASCUS undergoing colposcopy.[8] HPV testing is not recommended for adolescent women with ASCUS because most of these women are HPV positive.[9,10]
HPV DNA testing is generally not appropriate or clinically useful after cytology results of LSIL, which is more severe than ASCUS, and most of these women (84%–96%) are carcinogenic HPV DNA positive.[11] One exception may be to clarify the risk for postmenopausal women with cytologic LSIL, which is an interpretation that can be falsely positive, presumably due to atrophic changes.[12]
Primary HPV Screening
Testing for HPV DNA as a primary screening test is an option for women aged 30 years and older. Women who are negative by cytology and HPV testing are at extremely low risk of CIN 3+ and therefore may be screened less frequently. A prospective cohort study of nearly 332,000 U.S. women aged 30 years and older undergoing HPV DNA and cervical cytology cotesting every 3 years found that the cumulative incidence of CIN 3+ in women with negative results for both tests at baseline was 0.047% at 3 years and 0.16% at 5 years.[13] A second study of more than 43,000 women aged 29 to 61 years, one-half of whom underwent three rounds of HPV DNA and cervical cytology cotesting every 5 years, found that the cumulative incidence of CIN 3+ in women with negative results for both tests at baseline was 0.01% (95% confidence interval [CI], 0.00%–0.05%) at 9 years and 0.07% (95% CI, 0.03%–0.17%) after 14 years of follow-up.[14] Screening more frequently than every 3 years would not improve sensitivity significantly but would increase costs and overtreatment.[15,16]
Numerous studies have demonstrated that, compared with cytology, HPV DNA testing is more sensitive for identifying women who have CIN 2+ (range of sensitivities, 84%–97%).[17–24] In one randomized trial using both Pap and HPV testing in random order among women aged 30 to 69 years, sensitivity of HPV was 95% compared with 55% for Pap cytology. The combination of HPV and cytology had 100% sensitivity and a referral rate of 7.9%.[18]
The lower specificity of HPV DNA testing compared with cytology is a consideration. Among women older than 30 years, cytology had a specificity of 97% compared with 94% for HPV testing.[18] The specificity of HPV DNA testing would likely be even lower among women younger than 30 years, who have more transient HPV infection that is of little consequence. Thus, detecting such women would potentially increase the number of follow-up diagnostic workups. Potential approaches to minimize over-referral with HPV DNA testing and improve specificity include: (1) triage HPV-positive results with cytology [23] or another more specific molecular assay;[25] and (2) trigger further workup only after two sequential positive HPV test results because it is the persistence of carcinogenic HPV that confers the greatest risk of CIN 2–3.[26,27]
An Italian population-based, randomized, controlled trial of HPV DNA testing versus cervical cytology performed at 3-year intervals in approximately 94,000 women aged 25 to 60 years found a statistically significant decrease in the number of invasive cervical cancer cases diagnosed in the HPV DNA arm at the second round of screening (0 cases vs. 9 cases; P = .004). However, about 48% of individuals in the HPV DNA arm also received conventional cytology testing at the first screening round, making it impossible to discern whether the observed difference resulted from the use of a combined testing strategy or HPV DNA testing alone. Of note, many more women in the HPV DNA arm than in the cytology-alone arm were referred to colposcopy for abnormal findings (4,436 women vs. 1,416 women), prompting the authors to conclude that if the HPV DNA test is used as a primary screening strategy, women with positive test results should be triaged by cytology before referral.[28] A Canadian study of 19,000 women aged 25 to 65 years that compared HPV DNA testing with cervical cytology found that HPV DNA testing identified most women with CIN 3 at initial screening. Women who initially tested HPV DNA negative were at low risk for cervical dysplasia 48 months later. Additionally, there were not an excessive number of women referred for additional diagnostic testing.[24]
A study using data from a population-based randomized trial of cervical cancer screening among women aged 32 to 38 years compared 11 different screening strategies using HPV DNA testing and cytology. The strategy of initial screening with an HPV DNA test and a triage of HPV-positive results with cytology, and subsequent repeat HPV DNA testing after 1 year for women who were HPV positive but cytology negative, increased the sensitivity for detection of CIN 3+ by 30% compared with cytology alone, and increased the total number of screening tests performed by only 12%.[29] In a review of data from a large integrated health system, the added benefit of cotesting versus HPV testing alone would improve detection of CIN 3 or early-stage cervical cancer in very few women. Only 5.1% of locally advanced invasive cancers and 3.6% of CIN 3 were cytology positive and HPV negative, representing a very small fraction of all screened women.[30]
Cytology can be used to triage after primary HPV screening. Triage with cytology can be improved with concomitant detection of p16 and Ki-67 in the same cell (p16/Ki-67 dual stain [DS]). DS can be assessed manually through immunostaining cervical cytology slides. Additionally, artificial intelligence–based deep learning algorithms are currently being investigated and applied to aid in automated identification of p16/Ki-67 dual-stained slides. This approach has been shown to improve specificity without sacrificing sensitivity over manual DS assessment,[31] but it has not yet been validated in population studies.
References
McCredie MR, Sharples KJ, Paul C, et al.: Natural history of cervical neoplasia and risk of invasive cancer in women with cervical intraepithelial neoplasia 3: a retrospective cohort study. Lancet Oncol 9 (5): 425-34, 2008. [PUBMED Abstract]
Stoler MH, Schiffman M; Atypical Squamous Cells of Undetermined Significance-Low-grade Squamous Intraepithelial Lesion Triage Study (ALTS) Group: Interobserver reproducibility of cervical cytologic and histologic interpretations: realistic estimates from the ASCUS-LSIL Triage Study. JAMA 285 (11): 1500-5, 2001. [PUBMED Abstract]
Castle PE, Schiffman M, Wheeler CM, et al.: Evidence for frequent regression of cervical intraepithelial neoplasia-grade 2. Obstet Gynecol 113 (1): 18-25, 2009. [PUBMED Abstract]
Halfon P, Trepo E, Antoniotti G, et al.: Prospective evaluation of the Hybrid Capture 2 and AMPLICOR human papillomavirus (HPV) tests for detection of 13 high-risk HPV genotypes in atypical squamous cells of uncertain significance. J Clin Microbiol 45 (2): 313-6, 2007. [PUBMED Abstract]
Thomsen LT, Frederiksen K, Munk C, et al.: High-risk and low-risk human papillomavirus and the absolute risk of cervical intraepithelial neoplasia or cancer. Obstet Gynecol 123 (1): 57-64, 2014. [PUBMED Abstract]
Castle PE, Hunt WC, Langsfeld E, et al.: Three-year risk of cervical precancer and cancer after the detection of low-risk human papillomavirus genotypes targeted by a commercial test. Obstet Gynecol 123 (1): 49-56, 2014. [PUBMED Abstract]
Kulasingam SL, Kim JJ, Lawrence WF, et al.: Cost-effectiveness analysis based on the atypical squamous cells of undetermined significance/low-grade squamous intraepithelial lesion Triage Study (ALTS). J Natl Cancer Inst 98 (2): 92-100, 2006. [PUBMED Abstract]
ASCUS-LSIL Traige Study (ALTS) Group: Results of a randomized trial on the management of cytology interpretations of atypical squamous cells of undetermined significance. Am J Obstet Gynecol 188 (6): 1383-92, 2003. [PUBMED Abstract]
Wright TC, Massad LS, Dunton CJ, et al.: 2006 consensus guidelines for the management of women with abnormal cervical cancer screening tests. Am J Obstet Gynecol 197 (4): 346-55, 2007. [PUBMED Abstract]
Sherman ME, Schiffman M, Cox JT, et al.: Effects of age and human papilloma viral load on colposcopy triage: data from the randomized Atypical Squamous Cells of Undetermined Significance/Low-Grade Squamous Intraepithelial Lesion Triage Study (ALTS). J Natl Cancer Inst 94 (2): 102-7, 2002. [PUBMED Abstract]
ASCUS-LSIL Traige Study (ALTS) Group: A randomized trial on the management of low-grade squamous intraepithelial lesion cytology interpretations. Am J Obstet Gynecol 188 (6): 1393-400, 2003. [PUBMED Abstract]
Zuna RE, Wang SS, Rosenthal DL, et al.: Determinants of human papillomavirus-negative, low-grade squamous intraepithelial lesions in the atypical squamous cells of undetermined significance/low-grade squamous intraepithelial lesions triage study (ALTS). Cancer 105 (5): 253-62, 2005. [PUBMED Abstract]
Katki HA, Kinney WK, Fetterman B, et al.: Cervical cancer risk for women undergoing concurrent testing for human papillomavirus and cervical cytology: a population-based study in routine clinical practice. Lancet Oncol 12 (7): 663-72, 2011. [PUBMED Abstract]
Dijkstra MG, van Zummeren M, Rozendaal L, et al.: Safety of extending screening intervals beyond five years in cervical screening programmes with testing for high risk human papillomavirus: 14 year follow-up of population based randomised cohort in the Netherlands. BMJ 355: i4924, 2016. [PUBMED Abstract]
Saslow D, Runowicz CD, Solomon D, et al.: American Cancer Society guideline for the early detection of cervical neoplasia and cancer. CA Cancer J Clin 52 (6): 342-62, 2002 Nov-Dec. [PUBMED Abstract]
Goldie SJ, Kim JJ, Wright TC: Cost-effectiveness of human papillomavirus DNA testing for cervical cancer screening in women aged 30 years or more. Obstet Gynecol 103 (4): 619-31, 2004. [PUBMED Abstract]
Arbyn M, Sasieni P, Meijer CJ, et al.: Chapter 9: Clinical applications of HPV testing: a summary of meta-analyses. Vaccine 24 (Suppl 3): S3/78-89, 2006. [PUBMED Abstract]
Mayrand MH, Duarte-Franco E, Rodrigues I, et al.: Human papillomavirus DNA versus Papanicolaou screening tests for cervical cancer. N Engl J Med 357 (16): 1579-88, 2007. [PUBMED Abstract]
Naucler P, Ryd W, Törnberg S, et al.: Human papillomavirus and Papanicolaou tests to screen for cervical cancer. N Engl J Med 357 (16): 1589-97, 2007. [PUBMED Abstract]
Bulkmans NW, Berkhof J, Rozendaal L, et al.: Human papillomavirus DNA testing for the detection of cervical intraepithelial neoplasia grade 3 and cancer: 5-year follow-up of a randomised controlled implementation trial. Lancet 370 (9601): 1764-72, 2007. [PUBMED Abstract]
Cuzick J, Szarewski A, Cubie H, et al.: Management of women who test positive for high-risk types of human papillomavirus: the HART study. Lancet 362 (9399): 1871-6, 2003. [PUBMED Abstract]
Hartmann KE, Hall SA, Nanda K, et al.: Screening for Cervical Cancer. Rockville, Md: Agency for Health Research and Quality, 2002. Available online. Last accessed December 18, 2024.
Cuzick J, Clavel C, Petry KU, et al.: Overview of the European and North American studies on HPV testing in primary cervical cancer screening. Int J Cancer 119 (5): 1095-101, 2006. [PUBMED Abstract]
Ogilvie GS, van Niekerk D, Krajden M, et al.: Effect of Screening With Primary Cervical HPV Testing vs Cytology Testing on High-grade Cervical Intraepithelial Neoplasia at 48 Months: The HPV FOCAL Randomized Clinical Trial. JAMA 320 (1): 43-52, 2018. [PUBMED Abstract]
Carozzi F, Confortini M, Dalla Palma P, et al.: Use of p16-INK4A overexpression to increase the specificity of human papillomavirus testing: a nested substudy of the NTCC randomised controlled trial. Lancet Oncol 9 (10): 937-45, 2008. [PUBMED Abstract]
Koshiol J, Lindsay L, Pimenta JM, et al.: Persistent human papillomavirus infection and cervical neoplasia: a systematic review and meta-analysis. Am J Epidemiol 168 (2): 123-37, 2008. [PUBMED Abstract]
Castle PE: Invited commentary: is monitoring of human papillomavirus infection for viral persistence ready for use in cervical cancer screening? Am J Epidemiol 168 (2): 138-44; discussion 145-8, 2008. [PUBMED Abstract]
Ronco G, Giorgi-Rossi P, Carozzi F, et al.: Efficacy of human papillomavirus testing for the detection of invasive cervical cancers and cervical intraepithelial neoplasia: a randomised controlled trial. Lancet Oncol 11 (3): 249-57, 2010. [PUBMED Abstract]
Naucler P, Ryd W, Törnberg S, et al.: Efficacy of HPV DNA testing with cytology triage and/or repeat HPV DNA testing in primary cervical cancer screening. J Natl Cancer Inst 101 (2): 88-99, 2009. [PUBMED Abstract]
Schiffman M, Kinney WK, Cheung LC, et al.: Relative Performance of HPV and Cytology Components of Cotesting in Cervical Screening. J Natl Cancer Inst 110 (5): 501-508, 2018. [PUBMED Abstract]
Wentzensen N, Lahrmann B, Clarke MA, et al.: Accuracy and Efficiency of Deep-Learning-Based Automation of Dual Stain Cytology in Cervical Cancer Screening. J Natl Cancer Inst 113 (1): 72-79, 2021. [PUBMED Abstract]
Screening Benefit According to Age
Cervical cancer mortality, usually in unscreened women, increases with age, with the maximum mortality for White women between the ages of 45 years and 70 years, and for Black women in their 70s.[1,2] (Also available online.)
Mortality among women with negative Pap screening is low at all ages.
Screening by Pap testing with associated diagnostic testing and treatment is effective in reducing the incidence of all histologies and stages of invasive cervical cancer.[3] The benefit increases with age. Whereas the odds ratio is 0.79 (95% confidence interval [CI], 0.57–1.1) among women screened at age 30 to 31 years for developing cancer at age 35 to 39 years, it improves to 0.26 (95% CI, 0.19–0.36) among women screened at age 52 to 54 years for developing cancer at age 55 to 59 years.
Women aged 20 years and younger are more likely to have Pap abnormalities leading to further testing and treatment, so forgoing Pap testing in these women may improve the benefit-risk balance for this intervention. For more information, see the Evidence of Harm section. Women in this age group have a very low risk of cervical cancer and a high likelihood that cervical cell abnormalities will go away on their own.[4]
High-grade squamous intraepithelial lesions are rare among women older than 65 years who have been previously screened. For women with a negative Pap test at age 60 years and older, the likelihood of having a new diagnosis of CIN 3+ on repeat screening is less than 1 in 1,000 (in some studies, as few as 2–6 in 10,000).[5]
References
Saslow D, Runowicz CD, Solomon D, et al.: American Cancer Society guideline for the early detection of cervical neoplasia and cancer. CA Cancer J Clin 52 (6): 342-62, 2002 Nov-Dec. [PUBMED Abstract]
National Institutes of Health Consensus Development Conference Statement: cervical cancer, April 1-3, 1996. National Institutes of Health Consensus Development Panel. J Natl Cancer Inst Monogr (21): vii-xix, 1996. [PUBMED Abstract]
Sasieni P, Castanon A, Cuzick J: Effectiveness of cervical screening with age: population based case-control study of prospectively recorded data. BMJ 339: b2968, 2009. [PUBMED Abstract]
Saslow D, Solomon D, Lawson HW, et al.: American Cancer Society, American Society for Colposcopy and Cervical Pathology, and American Society for Clinical Pathology screening guidelines for the prevention and early detection of cervical cancer. CA Cancer J Clin 62 (3): 147-72, 2012 May-Jun. [PUBMED Abstract]
Sawaya GF, Grady D, Kerlikowske K, et al.: The positive predictive value of cervical smears in previously screened postmenopausal women: the Heart and Estrogen/progestin Replacement Study (HERS). Ann Intern Med 133 (12): 942-50, 2000. [PUBMED Abstract]
Alternative Screening and Treatment Strategies Including Low-Resource Settings
Choice in methods of screening for cervical cancer in resource-limited countries or underserved populations has prompted the evaluation of alternative methods, including self-collected human papillomavirus (HPV) tests and one-time screen-and-treat approaches.
Visual Inspection of the Cervix With Acetic Acid (VIA)
A clustered, randomized, controlled trial in rural India evaluated the impact of one-time visual VIA and immediate colposcopy, directed biopsy, and cryotherapy (where indicated) on cervical cancer incidence and mortality in healthy women aged 30 to 59 years.[1] Fifty-seven clusters (n = 31,343 women) received the intervention, while 56 control clusters (n = 30,958 women) received counseling and education about cervical cancer screening. After 7 years of follow-up, with adjustments for age, education, marital status, parity, and cluster design, there was a 25% relative reduction in cervical cancer incidence in the intervention arm compared with the control group (hazard ratio [HR], 0.75; 95% confidence interval [CI], 0.55–0.95). Using the same adjustments, cervical cancer mortality rates had a 35% relative reduction in the intervention arm compared with the control group (HR, 0.65; 95% CI, 0.47–0.89); the age-standardized rate of death caused by cervical cancer was 39.6 per 100,000 person-years for the intervention group versus 56.7 per 100,000 person-years for the control group. However, using the same cohort, the same authors subsequently reported that HPV testing is superior at reducing cervical cancer mortality.[2] This population was essentially screen naive at study entry and demonstrated a much higher overall risk of cervical cancer death (11% in the control group) than that observed in the U.S. population; therefore, these findings are not applicable to U.S. and similar Western health care. Histological diagnosis of cervical lesions happened after treatment had already taken place, and approximately 27% of patients in this trial received cryotherapy for lesions later determined to be nonmalignant.[3]
A second cluster-randomized trial of VIA screening in low socioeconomic areas of urban Mumbai, India, similarly demonstrated its efficacy in reducing cervical cancer mortality. In this trial, primary community health workers (as opposed to medical personnel) were trained to provide biennial VIA screening to 75,360 women aged 35 to 64 years. Women with positive screening results were referred to a central hospital for free diagnostic confirmation (including Pap smear, colposcopy, and biopsy, if indicated) and treatment—where warranted—according to hospital protocol. A control group (n = 76,178) received general cancer education. After 12 years, the relative risk (RR) of dying from cervical cancer was reduced by 31% in the screening arm (rate ratio, 0.69; 95% CI, 0.54–0.88), corresponding to about 5 fewer deaths per 100,000 woman-years. Compliance with treatment was about 15% lower for those in the control arm, which may have inflated the observed mortality benefit somewhat.[4]
A demonstration project in Kolkata, India, enrolled 39,740 women aged 30 to 60 years who underwent screening with VIA and Hybrid Capture II HPV DNA testing with colposcopy referral for a positive test, followed by biopsy and treatment if indicated. Estimated test performance for detection of cervical intraepithelial neoplasia (CIN) severe dysplasia (CIN 3+), corrected for verification bias, demonstrated that VIA achieved a sensitivity of 59.9% (95% CI, 49.9%–69.1%) and a specificity of 93.2% (95% CI, 92.9%–93.4%) compared with HPV testing, which resulted in a sensitivity of 91.2% (95% CI, 85.4%–95.7%) and a specificity of 96.9% (95% CI, 96.7%–97.0%). HPV testing identified an additional 32 CIN 3+ cases and 7 invasive cancer cases missed by VIA.[5]
A randomized trial in South Africa evaluated the impact on diagnosis of CIN moderate dysplasia (CIN 2+) at 6 months with a screen-and-treat approach with VIA and HPV versus delayed evaluation.[6] Women underwent HPV DNA testing and VIA testing (N = 6,555) and then returned in 2 to 6 days and were randomly assigned to one of three groups to receive (1) cryotherapy if the HPV DNA test result was positive (n = 2,163; 473 HPV+ and 467 treated); (2) cryotherapy if the VIA test result was positive (n = 2,227; 492 VIA+ and 482 treated); or (3) delayed evaluation (n = 2,165). At 6 months, CIN 2+ was diagnosed in 0.80% of women in the HPV+/cryotherapy group, in 2.23% of the VIA+/cryotherapy group, and in 3.55% of the delayed evaluation group. Differences in the prevalence of CIN 2+ persisted among the subset of women evaluated at 12 months. For the secondary outcome of CIN 3+, the prevalence of CIN 3+ lesions was low among the three groups but followed the same pattern (two cases in the HPV DNA group, three cases in the VIA group, and eight cases in the delayed evaluation group).
While VIA is practical in resource-limited settings, the accuracy and reproducibility are low. Advances in machine deep learning may help improve these metrics. A supervised, deep learning–based approach to predicting cervical precancers and cancers was investigated in a retrospective data set of 9,406 women who underwent cervical cancer screening using photographic images of the cervix. The archived digitized cervical images, taken with a fixed-focus camera (cervicography), were used for training and validation of the deep learning–based algorithm. The automated algorithm achieved better accuracy in predicting precancer and cancer compared with the original physician readers who interpreted the cervicography; it also compared favorably to conventional Pap smear cytology. This automated visual evaluation method needs to be transferred from digitized cervigrams (now obsolete) to contemporary digital cameras.[7]
A study of the feasibility of single-visit management of high-grade cervical lesions was conducted among a predominantly Latina population in California.[8] Women were randomly assigned to a single-visit group (n = 1,716) in which the Pap test was evaluated immediately and treatment administered the same day for women with HSILs or atypical glandular cells of undetermined significance (AGUS); or to usual care (n = 1,805), with results of the Pap test provided within 2 to 4 weeks and referrals for treatment based on results. The program was feasible, with a high degree of acceptability: 14 of 16 women (88%) with abnormal test results completed treatment by 6 months, while 10 of 19 women (53%) in the usual-care arm completed treatment by 6 months. Follow-up at 12 months was also higher among women in the single-visit group with HSILs/AGUS than among those in the usual-care arm; among all women, only 36% in each group had a follow-up Pap test at 1 year.
Self-Collection of HPV Tests
Self-collected HPV testing may be an alternative method for primary cervical screening. Incorporating self-collection of samples for HPV testing may improve access to cervical cancer screening, especially in communities with limited access to health care providers. A pooled analysis of cervical screening studies conducted in China compared the sensitivity and specificity of self-collected cervical specimens for HPV DNA testing, physician-collected specimens for HPV testing, liquid-based cytology (LBC), and VIA. The study included 13,004 participants in the analysis. Women underwent screening with all three sampling methods; in one study included in the pooled analysis, all women had colposcopy and biopsy. The women were instructed in the self-collection methodology by physicians, which likely affected the quality of specimen collection and thus the accuracy of the test in these studies. HPV DNA testing on physician-collected specimens had the highest sensitivity, 97.0% for CIN 2+ (95% CI, 95.2%–98.3%) and 97.8% for CIN 3+ (95% CI, 95.3%–99.2%). The results of HPV DNA testing on self-collected specimens had moderate agreement with that of physician-collected specimens (kappa statistic, 0.67). Pooled sensitivity for self-collected HPV testing was 86.2% for CIN 2+ (95% CI, 82.9%–89.1%) and 86.1% for CIN 3+ (95% CI, 81.4%–90.0%). Pooled specificity for self-collected HPV DNA testing was 80.7% (95% CI, 75.6%–85.8%) for CIN 2+ and 79.5% (95% CI, 74.1%–84.8%) for CIN 3+. The specificity of HPV testing was lowest of all screening modalities. Whereas pooled sensitivity was highest for physician-collected HPV testing, it was lowest for the VIA screening methods—50.3% for CIN 2+ and 55.7% for CIN 3+. Pooled specificity was highest for LBC—94.0% for CIN 2+ and 92.8% for CIN 3+.[9]
A randomized noninferiority trial conducted in the Netherlands found that there was no difference in the CIN 2+ sensitivity or specificity of HPV testing between self-sampling based on written instructions and clinician-based sampling (relative sensitivity, 0.96 [95% CI, 0.90–1.03]; relative specificity, 1.00 [95% CI, 0.99–1.01]).[10] A population-based cluster-randomized trial in Argentina, comparing screening uptake using self-collection of samples for HPV DNA testing with that of clinic-based cervical sample collection with cytology and HPV triage, found that self-collection was associated with increased screening (RR, 4.02; 95% CI, 3.44–4.71), which translated into higher detection of CIN 2+ and treatment.[11] A Dutch study among women who participated in the national cervical cancer screening program found that vaginal self-sampling was highly concordant (96.8%; 95% CI, 96.0%–97.5%) with high-risk HPV prevalence in physician-collected samples and was both convenient and user friendly. Vaginal self-sampling will be offered in the Dutch national screening program for those who do not participate in their routine screening.[12]
A randomized trial within the U.S. Kaiser Permanente health care system evaluated the effectiveness of mailed HPV self-sampling kits versus usual-care reminders for in-clinic screening to increase the uptake of cervical cancer screening and the detection of CIN 2+. A total of 19,851 women who were overdue for screening were randomly assigned to either the self-sampling intervention or the usual-care control group. Screening uptake was higher in the intervention group (26.3%) than in the control group (17.4%) (RR, 1.51; 95% CI, 1.43–1.60). In the intervention group, 12 participants with CIN 2+ were detected compared with 8 participants in the control group (RR, 1.49; 95% CI, 0.61–3.64), and 12 patients were treated compared with 7 of those in the control group (RR, 1.70; 95% CI, 0.67–4.32).[13] As a follow-up to this study, the authors conducted the STEP study (self-testing options in the era for primary HPV screening for cervical cancer), a pragmatic, parallel, single-blinded, randomized clinical trial that compared cervical cancer screening completion across strata of individuals due for screening (screening adherent), overdue for screening, or with unknown screening histories.[14] Overall, 31,355 English-speaking individuals enrolled in Kaiser Permanente Washington were included and randomly assigned to receive usual care (patient reminders and clinical electronic health record alerts), education (usual care plus educational material about screening), direct mail (usual care plus educational materials plus a mailed HPV self-sampling kit), or to opt in (usual care plus educational materials plus the option to request a self-sampling kit). Direct mailing (of HPV self-sampling kits to individuals) increased cervical cancer screening by more than 14% in individuals who were due or overdue for cervical cancer screening, compared with education alone (attention control). Compared with the education group, time-to-screening completion was shorter for the direct-mail and opt-in groups. Time-to-screening completion for the education and the usual-care groups was similar across all screening-history strata. Furthermore, the education and usual-care groups had similar screening rates in this study population. Strengthened by the pragmatic design, this trial was highly inclusive of a diverse patient population with regards to age, race, ethnicity, health care utilization, and household income. Nevertheless, the generalizability of these study results may be limited given that participants were English-speaking and enrolled in a mixed-model managed care system, with both access to health care and insurance coverage. Notably, the HPV self-sampling test used in this study is now approved by the U.S. Food and Drug Administration for use in a health care setting.
A study including underscreened ethnic minority groups and immigrant populations in South Florida evaluated the effectiveness of HPV self-sampling by randomizing women to self-collection via a mailed self-sampling kit or through an in-person visit by a community health worker.[15] The participants self-identified as Hispanic, Haitian, or non-Hispanic Black women between the ages of 30 years and 65 years. After adjusting for study site, age, income, insurance education, Pap smear history, marital status, and citizenship status, women who received the self-sampling intervention via an in-person visit from a community health worker were more likely to complete the self-sampling (odds ratio, 1.81; 95% CI, 1.22–2.69). Completion of HPV self-sampling was high in both study arms, with 81.0% (n = 243) among the in-person visit group and 71.6% (n = 214) among those who received the self-sampling HPV kit via mail.
References
Sankaranarayanan R, Esmy PO, Rajkumar R, et al.: Effect of visual screening on cervical cancer incidence and mortality in Tamil Nadu, India: a cluster-randomised trial. Lancet 370 (9585): 398-406, 2007. [PUBMED Abstract]
Sankaranarayanan R, Nene BM, Shastri SS, et al.: HPV screening for cervical cancer in rural India. N Engl J Med 360 (14): 1385-94, 2009. [PUBMED Abstract]
Szarewski A: Cervical screening by visual inspection with acetic acid. Lancet 370 (9585): 365-6, 2007. [PUBMED Abstract]
Shastri SS, Mittra I, Mishra GA, et al.: Effect of VIA screening by primary health workers: randomized controlled study in Mumbai, India. J Natl Cancer Inst 106 (3): dju009, 2014. [PUBMED Abstract]
Basu P, Mittal S, Banerjee D, et al.: Diagnostic accuracy of VIA and HPV detection as primary and sequential screening tests in a cervical cancer screening demonstration project in India. Int J Cancer 137 (4): 859-67, 2015. [PUBMED Abstract]
Denny L, Kuhn L, De Souza M, et al.: Screen-and-treat approaches for cervical cancer prevention in low-resource settings: a randomized controlled trial. JAMA 294 (17): 2173-81, 2005. [PUBMED Abstract]
Hu L, Bell D, Antani S, et al.: An Observational Study of Deep Learning and Automated Evaluation of Cervical Images for Cancer Screening. J Natl Cancer Inst 111 (9): 923-932, 2019. [PUBMED Abstract]
Brewster WR, Hubbell FA, Largent J, et al.: Feasibility of management of high-grade cervical lesions in a single visit: a randomized controlled trial. JAMA 294 (17): 2182-7, 2005. [PUBMED Abstract]
Zhao FH, Lewkowitz AK, Chen F, et al.: Pooled analysis of a self-sampling HPV DNA Test as a cervical cancer primary screening method. J Natl Cancer Inst 104 (3): 178-88, 2012. [PUBMED Abstract]
Polman NJ, Ebisch RMF, Heideman DAM, et al.: Performance of human papillomavirus testing on self-collected versus clinician-collected samples for the detection of cervical intraepithelial neoplasia of grade 2 or worse: a randomised, paired screen-positive, non-inferiority trial. Lancet Oncol 20 (2): 229-238, 2019. [PUBMED Abstract]
Arrossi S, Thouyaret L, Herrero R, et al.: Effect of self-collection of HPV DNA offered by community health workers at home visits on uptake of screening for cervical cancer (the EMA study): a population-based cluster-randomised trial. Lancet Glob Health 3 (2): e85-94, 2015. [PUBMED Abstract]
Ketelaars PJW, Bosgraaf RP, Siebers AG, et al.: High-risk human papillomavirus detection in self-sampling compared to physician-taken smear in a responder population of the Dutch cervical screening: Results of the VERA study. Prev Med 101: 96-101, 2017. [PUBMED Abstract]
Winer RL, Lin J, Tiro JA, et al.: Effect of Mailed Human Papillomavirus Test Kits vs Usual Care Reminders on Cervical Cancer Screening Uptake, Precancer Detection, and Treatment: A Randomized Clinical Trial. JAMA Netw Open 2 (11): e1914729, 2019. [PUBMED Abstract]
Winer RL, Lin J, Anderson ML, et al.: Strategies to Increase Cervical Cancer Screening With Mailed Human Papillomavirus Self-Sampling Kits: A Randomized Clinical Trial. JAMA 330 (20): 1971-1981, 2023. [PUBMED Abstract]
Kobetz E, Seay J, Koru-Sengul T, et al.: A randomized trial of mailed HPV self-sampling for cervical cancer screening among ethnic minority women in South Florida. Cancer Causes Control 29 (9): 793-801, 2018. [PUBMED Abstract]
Evidence of Harm
Annually in the United States, an estimated 65 million women undergo cervical cancer screening;[1] about 3.9 million (6%) will be referred for further evaluation.[2] About 11,000 cases of invasive cervical cancer were diagnosed in 2008. Thus, Papanicolaou (Pap) test screening results in a large number of colposcopies for benign conditions.
The major potential harm of screening for cervical cancer lies in the screening detection of many cytologic abnormalities such as atypical squamous cells of undetermined significance (ASCUS) and low-grade squamous intraepithelial lesions (LSILs), the majority of which would never progress to cervical cancer. Women with human papillomavirus (HPV)-positive ASCUS or LSILs on Pap testing are usually referred for colposcopy. Histological CIN 2+ is treated with cryotherapy or loop electrosurgical excision procedure. These procedures permanently alter the cervix and have consequences on fertility and pregnancy.[3] Younger women are more likely to acquire HPV infections and be referred for diagnostic workup, and they are more likely to suffer harms from interventions for a condition that often resolves spontaneously.
On the basis of an analysis of screening records from nearly 350,000 women in Bristol, England, investigators projected that 1,000 women would need to be screened for cervical cancer for 35 years to prevent one death from the disease. For each death prevented, the authors estimated that more than 150 women have an abnormal result, more than 80 women are referred for investigation, and more than 50 women have treatment.[4]
References
Solomon D, Breen N, McNeel T: Cervical cancer screening rates in the United States and the potential impact of implementation of screening guidelines. CA Cancer J Clin 57 (2): 105-11, 2007 Mar-Apr. [PUBMED Abstract]
Davey DD, Woodhouse S, Styer P, et al.: Atypical epithelial cells and specimen adequacy: current laboratory practices of participants in the college of American pathologists interlaboratory comparison program in cervicovaginal cytology. Arch Pathol Lab Med 124 (2): 203-11, 2000. [PUBMED Abstract]
Sadler L, Saftlas A, Wang W, et al.: Treatment for cervical intraepithelial neoplasia and risk of preterm delivery. JAMA 291 (17): 2100-6, 2004. [PUBMED Abstract]
Raffle AE, Alden B, Quinn M, et al.: Outcomes of screening to prevent cancer: analysis of cumulative incidence of cervical abnormality and modelling of cases and deaths prevented. BMJ 326 (7395): 901, 2003. [PUBMED Abstract]
Latest Updates to This Summary (04/18/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 1).
This summary is written and maintained by the PDQ Screening and Prevention Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about cervical cancer screening. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Screening and Prevention Editorial Board. PDQ Cervical Cancer Screening. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/cervical/hp/cervical-screening-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389177]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
Gardasil 9 Vaccine Protects against Additional HPV Types
Summary
In a large randomized clinical trial, a new human papillomavirus (HPV) vaccine effectively prevented infection and disease caused by nine HPV types, including seven types that cause cervical and other cancers—five of which were not covered by the previously available HPV vaccines—and two types that cause genital warts.
Source
New England Journal of Medicine, February 18, 2015 (See the abstract.)
Background
HPV infections are the most common sexually transmitted infections in the United States. More than 40 HPV types can be spread through direct sexual contact. Of these, about a dozen, including HPV types 16, 18, 31, 33, 45, 52, and 58, are high-risk—that is, persistent infection with these HPV types can cause cellular changes that may progress to cancer, including cervical, anal, penile, vaginal, vulvar, and oropharyngeal cancers. HPV types 16 and 18 are responsible for approximately 70 percent of all cervical cancers and HPV types 31, 33, 45, 52, and 58 are responsible for another 20 percent of cervical cancers.
Until December 2014, the FDA had approved two vaccines to protect against HPV infection: Cervarix, a bivalent vaccine that protects against infection with HPV types 16 and 18, and Gardasil, a quadrivalent vaccine that protects against infection with HPV types 6, 11, 16, and 18. Types 6 and 11 are low-risk types that do not cause cancer but can cause warts on or around the genitals, anus, mouth, or throat.
Researchers have been working on developing next-generation HPV vaccines that protect against additional high-risk HPV types. In December 2014, based on the results of this clinical trial, the FDA approved the 9-valent vaccine Gardasil 9, which protects against infection with HPV types 6, 11, 16, 18, 31, 33, 45, 52 and 58.
The Study
In a phase II/III international double-blind clinical trial, researchers randomly assigned 14,215 women between the ages of 16 and 26 to receive Gardasil or Gardasil 9. As part of the clinical trial, a small dose-finding study was first done to determine the dose that would be used in the larger efficacy study. Participants were eligible if they had no history of an abnormal Pap test, no more than four lifetime sexual partners, and no previous abnormal finding on a cervical biopsy.
Women in each group received three intramuscular injections of the vaccine over 6 months.
The study’s primary endpoint was the incidence of high-grade cervical, vulvar, or vaginal disease, including high-grade cervical epithelial neoplasia, adenocarcinoma in situ, cervical cancer, high-grade vulvar intraepithelial neoplasia, high-grade vaginal intraepithelial neoplasia, vulvar cancer, and vaginal cancer. Secondary endpoints included safety and the ability of the vaccine to provoke an immune response (vaccine immunogenicity), as measured by antibody responses to HPV types 6, 11, 16, and 18.
Elmar A. Joura, M.D., of the Medical University of Vienna, Comprehensive Cancer Center in Austria, led the trial, which was sponsored by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., the maker of Gardasil and Gardasil 9.
Results
Among study participants who did not have a documented HPV infection at the time of their first vaccine injection and who received all three doses, the number of cases of cervical, vulvar, or vaginal disease related to HPV types 31, 33, 45, 52, and 58 was much lower among patients who received the 9-valent vaccine compared with those who received the quadrivalent vaccine (1 versus 30 cases). The efficacy rate of the 9-valent vaccine was 96.7 percent.
Gardasil 9 was as effective as Gardasil at generating an antibody response against the four HPV types targeted by both vaccines. All cases of high-grade disease detected in the Gardasil 9 group occurred in participants who were HPV-infected when they received their first vaccine dose, which underscores the importance of vaccinating women and girls before they are exposed to HPV, the authors noted.
The safety of Gardasil 9 was evaluated in more than 7,000 women. Adverse events related to the injection site, including mild or moderate pain, swelling, redness, and itching, were more common in the Gardasil 9 group than in the Gardasil group. The rate of adverse systemic events—headache, fever, nausea, dizziness, and fatigue—were similar in the two groups.
Limitations
The researchers compared Gardasil 9 to Gardasil and did not include a placebo control group. Consequently, the researchers were unable to directly determine the efficacy of Gardasil 9 in preventing diseases associated with the HPV types the vaccines have in common (HPV types 6, 11, 16, and 18). However, because Gardasil 9 generated an antibody response similar to that generated by Gardasil and because the incidence of disease related to these four HPV types were similar, the authors concluded that Gardasil 9 would have similar efficacy to Gardasil in preventing diseases associated with these four HPV types.
The study authors noted that longer-term follow-up of participants vaccinated with Gardasil 9 is needed to provide information on the durability of protection.
Comment
This study represents “a milestone in expanding the coverage of cancers associated with the human papillomavirus,” wrote Anne Schuchat, M.D., of the Centers for Disease Control and Prevention, in an accompanying editorial in NEJM.
But even with a vaccine that provides protection against more high-risk HPV types, Dr. Schuchat noted that “vaccination of a much higher proportion of preteens is needed. Otherwise, decades from now oncologists will still be talking about HPV-associated cancers with thousands of new patients every year.”
Aimée Kreimer, Ph.D., of NCI’s Division of Cancer Epidemiology and Genetics, said she is hopeful that Gardasil 9 will protect against HPV infections for the same duration as the earlier HPV vaccines have demonstrated thus far.
Dr. Kreimer also noted that “it will be important to know if currently approved HPV vaccines, including Gardasil 9, may also be given in a reduced dosage schedule without diminishing their efficacy.” This would help “to maximize their benefit in regions of the world where HPV-associated cancers are the leading cause of cancer death in women and among segments of the U.S. population where coverage has been low.”
Cervical Cancer Treatment (PDQ®)–Health Professional Version
General Information About Cervical Cancer
Cervical cancer is relatively rare but is diagnosed most frequently in women aged 35 to 44 years.[1] It is the fourth most common cancer in women worldwide for both incidence and mortality, with 661,021 new cases and 348,189 deaths in 2022.[2] Most cases of cervical cancer are preventable with routine screening and treatment of precancerous lesions. As a result, most 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 2025:[3]
New cases: 13,360.
Deaths: 4,320.
Anatomy
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 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 precancerous and cancerous changes arise in this zone.
Cervical carcinoma begins at the squamocolumnar junction. It can involve the outer squamous cells, 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 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.[4–7]
Other risk factors for cervical cancer include the following:
Exposure to diethylstilbestrol (DES) in utero.[15]
Human papillomavirus (HPV) infection
HPV infection is a necessary step in the development of virtually all precancerous and cancerous lesions. Epidemiological studies convincingly demonstrate that the major risk factor for 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,[16] while cervical cancer is rare. The persistence of an HPV infection leads to increased risk of developing precancerous and cancerous lesions.[17,18]
The strain of HPV infection is also important in conferring risk. Multiple subtypes of HPV infect humans; 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.[19–21] Further studies have shown that infection with either HPV 16 or 18 is more predictive than cytological screening of high-grade CIN or greater disease, and that the predictive ability is seen for up to 18 years after the initial test.[22–24]
There are two commercially available vaccines that target anogenital-related strains of HPV. The vaccines are directed toward HPV-naïve adolescents and young adults. Although penetration of the vaccine has been moderate, significant decreases in HPV-related diseases have been documented.[25] For more information, see Cervical Cancer Prevention.
Clinical Features
Early cervical cancer may not cause noticeable signs or symptoms.
Possible signs and symptoms of cervical cancer include:
Vaginal bleeding.
Unusual vaginal discharge.
Pelvic pain.
Dyspareunia.
Postcoital bleeding.
Diagnosis
The following procedures may be used to diagnose cervical cancer:
History and physical examination.
Pelvic examination.
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:
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 cytological specimens were assayed by a sensitive (hybrid capture) technique.[26] The authors of the study and an accompanying editorial concluded that using HPV DNA testing in this setting does not add sufficient information to justify its cost.[26]
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.[26–28]
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 microinvasive histopathology on biopsy.[19,29–31] 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 of high-grade dysplasia and invasive cancer.[32]
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.[33] Other studies have shown the effectiveness of a primary HPV DNA–screening strategy with cytology triage over the previously used cytology-based screening algorithms.[34,35]
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 by using the Pap test and HPV testing.[36] 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 pathological findings in surgically treated patients.
Evidence (clinical stage and other findings):
In a large, surgicopathological staging study of patients with clinical stage IB disease reported by the Gynecologic Oncology Group (GOG) GOG-49, the factors that most prominently predicted 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.[37,38]
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 histological stage.[39]
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:[40]
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.[41] Several population-based and retrospective studies showed a worse outcome for patients with adenocarcinoma, with an increase in distant metastasis compared with those with squamous histology.[42–45] Reports conflict about the effect of adenosquamous cell type on outcome.[46,47] 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.[48]
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.[49] A multivariate analysis of factors influencing the incidence of distant metastases showed stage, endometrial extension of tumor, and pelvic tumor control to be significant indicators of distant dissemination.[49]
GOG studies have indicated that prognostic factors vary depending on whether clinical or surgical staging is used and with different treatments. Delay in radiation delivery completion is associated with poorer progression-free survival when clinical staging is used. Stage, tumor grade, race, and age are uncertain prognostic factors in studies using chemoradiation.[50]
Other prognostic factors
Other prognostic factors that may affect outcome include:
HIV status: Women with HIV have more aggressive and advanced disease and a poorer prognosis.[51]
MYC overexpression: A study of patients with known invasive squamous carcinoma of the cervix found that overexpression of the MYC oncogene was associated with a poorer prognosis.[52]
Number of cells in S phase: The number of cells in S phase may also have prognostic significance in early cervical carcinoma.[53]
HPV-18 DNA: HPV-18 DNA is 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.[54,55]
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.[56]
Follow-Up After Treatment
High-quality studies are lacking, and the optimal follow-up for patients after treatment for cervical cancer is unknown. Retrospective studies have shown that cancer recurrence is most likely within the first 2 years.[57] 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,[58,59] 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:
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.
References
National Cancer Institute: SEER Cancer Stat Facts: Cervical Cancer. Bethesda, Md: National Cancer Institute. Available online. Last accessed February 28, 2025.
Bray F, Laversanne M, Sung H, et al.: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 74 (3): 229-263, 2024. [PUBMED Abstract]
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Human papillomaviruses. IARC Monogr Eval Carcinog Risks Hum 100 (Pt B), 255-296, 2012. Available online. Last accessed January 31, 2025.
Schiffman M, Castle PE, Jeronimo J, et al.: Human papillomavirus and cervical cancer. Lancet 370 (9590): 890-907, 2007. [PUBMED Abstract]
Trottier H, Franco EL: The epidemiology of genital human papillomavirus infection. Vaccine 24 (Suppl 1): S1-15, 2006. [PUBMED Abstract]
Ault KA: Epidemiology and natural history of human papillomavirus infections in the female genital tract. Infect Dis Obstet Gynecol 2006 (Suppl): 40470, 2006. [PUBMED Abstract]
Muñoz N, Franceschi S, Bosetti C, et al.: Role of parity and human papillomavirus in cervical cancer: the IARC multicentric case-control study. Lancet 359 (9312): 1093-101, 2002. [PUBMED Abstract]
Plummer M, Herrero R, Franceschi S, et al.: Smoking and cervical cancer: pooled analysis of the IARC multi-centric case–control study. Cancer Causes Control 14 (9): 805-14, 2003. [PUBMED Abstract]
Moreno V, Bosch FX, Muñoz N, et al.: Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet 359 (9312): 1085-92, 2002. [PUBMED Abstract]
Appleby P, Beral V, Berrington de González A, et al.: Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet 370 (9599): 1609-21, 2007. [PUBMED Abstract]
Abraham AG, D’Souza G, Jing Y, et al.: Invasive cervical cancer risk among HIV-infected women: a North American multicohort collaboration prospective study. J Acquir Immune Defic Syndr 62 (4): 405-13, 2013. [PUBMED Abstract]
Grulich AE, van Leeuwen MT, Falster MO, et al.: Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet 370 (9581): 59-67, 2007. [PUBMED Abstract]
International Collaboration of Epidemiological Studies of Cervical Cancer: Cervical carcinoma and reproductive factors: collaborative reanalysis of individual data on 16,563 women with cervical carcinoma and 33,542 women without cervical carcinoma from 25 epidemiological studies. Int J Cancer 119 (5): 1108-24, 2006. [PUBMED Abstract]
Hoover RN, Hyer M, Pfeiffer RM, et al.: Adverse health outcomes in women exposed in utero to diethylstilbestrol. N Engl J Med 365 (14): 1304-14, 2011. [PUBMED Abstract]
Dunne EF, Unger ER, Sternberg M, et al.: Prevalence of HPV infection among females in the United States. JAMA 297 (8): 813-9, 2007. [PUBMED Abstract]
Rodríguez AC, Schiffman M, Herrero R, et al.: Rapid clearance of human papillomavirus and implications for clinical focus on persistent infections. J Natl Cancer Inst 100 (7): 513-7, 2008. [PUBMED Abstract]
Jaisamrarn U, Castellsagué X, Garland SM, et al.: Natural history of progression of HPV infection to cervical lesion or clearance: analysis of the control arm of the large, randomised PATRICIA study. PLoS One 8 (11): e79260, 2013. [PUBMED Abstract]
Brisson J, Morin C, Fortier M, et al.: Risk factors for cervical intraepithelial neoplasia: differences between low- and high-grade lesions. Am J Epidemiol 140 (8): 700-10, 1994. [PUBMED Abstract]
Koutsky LA, Holmes KK, Critchlow CW, et al.: A cohort study of the risk of cervical intraepithelial neoplasia grade 2 or 3 in relation to papillomavirus infection. N Engl J Med 327 (18): 1272-8, 1992. [PUBMED Abstract]
Schiffman MH, Bauer HM, Hoover RN, et al.: Epidemiologic evidence showing that human papillomavirus infection causes most cervical intraepithelial neoplasia. J Natl Cancer Inst 85 (12): 958-64, 1993. [PUBMED Abstract]
Castle PE, Glass AG, Rush BB, et al.: Clinical human papillomavirus detection forecasts cervical cancer risk in women over 18 years of follow-up. J Clin Oncol 30 (25): 3044-50, 2012. [PUBMED Abstract]
Khan MJ, Castle PE, Lorincz AT, et al.: The elevated 10-year risk of cervical precancer and cancer in women with human papillomavirus (HPV) type 16 or 18 and the possible utility of type-specific HPV testing in clinical practice. J Natl Cancer Inst 97 (14): 1072-9, 2005. [PUBMED Abstract]
Schlecht NF, Kulaga S, Robitaille J, et al.: Persistent human papillomavirus infection as a predictor of cervical intraepithelial neoplasia. JAMA 286 (24): 3106-14, 2001. [PUBMED Abstract]
Muñoz N, Kjaer SK, Sigurdsson K, et al.: Impact of human papillomavirus (HPV)-6/11/16/18 vaccine on all HPV-associated genital diseases in young women. J Natl Cancer Inst 102 (5): 325-39, 2010. [PUBMED Abstract]
Human papillomavirus testing for triage of women with cytologic evidence of low-grade squamous intraepithelial lesions: baseline data from a randomized trial. The Atypical Squamous Cells of Undetermined Significance/Low-Grade Squamous Intraepithelial Lesions Triage Study (ALTS) Group. J Natl Cancer Inst 92 (5): 397-402, 2000. [PUBMED Abstract]
Wright TC, Massad LS, Dunton CJ, et al.: 2006 consensus guidelines for the management of women with abnormal cervical cancer screening tests. Am J Obstet Gynecol 197 (4): 346-55, 2007. [PUBMED Abstract]
Wright TC, Massad LS, Dunton CJ, et al.: 2006 consensus guidelines for the management of women with cervical intraepithelial neoplasia or adenocarcinoma in situ. Am J Obstet Gynecol 197 (4): 340-5, 2007. [PUBMED Abstract]
Tabbara S, Saleh AD, Andersen WA, et al.: The Bethesda classification for squamous intraepithelial lesions: histologic, cytologic, and viral correlates. Obstet Gynecol 79 (3): 338-46, 1992. [PUBMED Abstract]
Cuzick J, Terry G, Ho L, et al.: Human papillomavirus type 16 in cervical smears as predictor of high-grade cervical intraepithelial neoplasia [corrected] Lancet 339 (8799): 959-60, 1992. [PUBMED Abstract]
Richart RM, Wright TC: Controversies in the management of low-grade cervical intraepithelial neoplasia. Cancer 71 (4 Suppl): 1413-21, 1993. [PUBMED Abstract]
Klaes R, Woerner SM, Ridder R, et al.: Detection of high-risk cervical intraepithelial neoplasia and cervical cancer by amplification of transcripts derived from integrated papillomavirus oncogenes. Cancer Res 59 (24): 6132-6, 1999. [PUBMED Abstract]
Katki HA, Kinney WK, Fetterman B, et al.: Cervical cancer risk for women undergoing concurrent testing for human papillomavirus and cervical cytology: a population-based study in routine clinical practice. Lancet Oncol 12 (7): 663-72, 2011. [PUBMED Abstract]
Naucler P, Ryd W, Törnberg S, et al.: Efficacy of HPV DNA testing with cytology triage and/or repeat HPV DNA testing in primary cervical cancer screening. J Natl Cancer Inst 101 (2): 88-99, 2009. [PUBMED Abstract]
Castle PE, Stoler MH, Wright TC, et al.: Performance of carcinogenic human papillomavirus (HPV) testing and HPV16 or HPV18 genotyping for cervical cancer screening of women aged 25 years and older: a subanalysis of the ATHENA study. Lancet Oncol 12 (9): 880-90, 2011. [PUBMED Abstract]
The 1988 Bethesda System for reporting cervical/vaginal cytological diagnoses. National Cancer Institute Workshop. JAMA 262 (7): 931-4, 1989. [PUBMED Abstract]
Delgado G, Bundy B, Zaino R, et al.: Prospective surgical-pathological study of disease-free interval in patients with stage IB squamous cell carcinoma of the cervix: a Gynecologic Oncology Group study. Gynecol Oncol 38 (3): 352-7, 1990. [PUBMED Abstract]
Zaino RJ, Ward S, Delgado G, et al.: Histopathologic predictors of the behavior of surgically treated stage IB squamous cell carcinoma of the cervix. A Gynecologic Oncology Group study. Cancer 69 (7): 1750-8, 1992. [PUBMED Abstract]
Burghardt E, Baltzer J, Tulusan AH, et al.: Results of surgical treatment of 1028 cervical cancers studied with volumetry. Cancer 70 (3): 648-55, 1992. [PUBMED Abstract]
Stehman FB, Bundy BN, DiSaia PJ, et al.: Carcinoma of the cervix treated with radiation therapy. I. A multi-variate analysis of prognostic variables in the Gynecologic Oncology Group. Cancer 67 (11): 2776-85, 1991. [PUBMED Abstract]
Steren A, Nguyen HN, Averette HE, et al.: Radical hysterectomy for stage IB adenocarcinoma of the cervix: the University of Miami experience. Gynecol Oncol 48 (3): 355-9, 1993. [PUBMED Abstract]
Park JY, Kim DY, Kim JH, et al.: Outcomes after radical hysterectomy in patients with early-stage adenocarcinoma of uterine cervix. Br J Cancer 102 (12): 1692-8, 2010. [PUBMED Abstract]
Eifel PJ, Burke TW, Morris M, et al.: Adenocarcinoma as an independent risk factor for disease recurrence in patients with stage IB cervical carcinoma. Gynecol Oncol 59 (1): 38-44, 1995. [PUBMED Abstract]
Lee YY, Choi CH, Kim TJ, et al.: A comparison of pure adenocarcinoma and squamous cell carcinoma of the cervix after radical hysterectomy in stage IB-IIA. Gynecol Oncol 120 (3): 439-43, 2011. [PUBMED Abstract]
Galic V, Herzog TJ, Lewin SN, et al.: Prognostic significance of adenocarcinoma histology in women with cervical cancer. Gynecol Oncol 125 (2): 287-91, 2012. [PUBMED Abstract]
Gallup DG, Harper RH, Stock RJ: Poor prognosis in patients with adenosquamous cell carcinoma of the cervix. Obstet Gynecol 65 (3): 416-22, 1985. [PUBMED Abstract]
Yazigi R, Sandstad J, Munoz AK, et al.: Adenosquamous carcinoma of the cervix: prognosis in stage IB. Obstet Gynecol 75 (6): 1012-5, 1990. [PUBMED Abstract]
Bethwaite P, Yeong ML, Holloway L, et al.: The prognosis of adenosquamous carcinomas of the uterine cervix. Br J Obstet Gynaecol 99 (9): 745-50, 1992. [PUBMED Abstract]
Fagundes H, Perez CA, Grigsby PW, et al.: Distant metastases after irradiation alone in carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys 24 (2): 197-204, 1992. [PUBMED Abstract]
Monk BJ, Tian C, Rose PG, et al.: Which clinical/pathologic factors matter in the era of chemoradiation as treatment for locally advanced cervical carcinoma? Analysis of two Gynecologic Oncology Group (GOG) trials. Gynecol Oncol 105 (2): 427-33, 2007. [PUBMED Abstract]
Maiman M, Fruchter RG, Guy L, et al.: Human immunodeficiency virus infection and invasive cervical carcinoma. Cancer 71 (2): 402-6, 1993. [PUBMED Abstract]
Bourhis J, Le MG, Barrois M, et al.: Prognostic value of c-myc proto-oncogene overexpression in early invasive carcinoma of the cervix. J Clin Oncol 8 (11): 1789-96, 1990. [PUBMED Abstract]
Strang P, Eklund G, Stendahl U, et al.: S-phase rate as a predictor of early recurrences in carcinoma of the uterine cervix. Anticancer Res 7 (4B): 807-10, 1987 Jul-Aug. [PUBMED Abstract]
Burger RA, Monk BJ, Kurosaki T, et al.: Human papillomavirus type 18: association with poor prognosis in early stage cervical cancer. J Natl Cancer Inst 88 (19): 1361-8, 1996. [PUBMED Abstract]
Lai CH, Chang CJ, Huang HJ, et al.: Role of human papillomavirus genotype in prognosis of early-stage cervical cancer undergoing primary surgery. J Clin Oncol 25 (24): 3628-34, 2007. [PUBMED Abstract]
Silva IH, Nogueira-Silva C, Figueiredo T, et al.: The impact of GGH -401C>T polymorphism on cisplatin-based chemoradiotherapy response and survival in cervical cancer. Gene 512 (2): 247-50, 2013. [PUBMED Abstract]
Ansink A, de Barros Lopes A, Naik R, et al.: Recurrent stage IB cervical carcinoma: evaluation of the effectiveness of routine follow up surveillance. Br J Obstet Gynaecol 103 (11): 1156-8, 1996. [PUBMED Abstract]
Duyn A, Van Eijkeren M, Kenter G, et al.: Recurrent cervical cancer: detection and prognosis. Acta Obstet Gynecol Scand 81 (4): 351-5, 2002. [PUBMED Abstract]
Morice P, Deyrolle C, Rey A, et al.: Value of routine follow-up procedures for patients with stage I/II cervical cancer treated with combined surgery-radiation therapy. Ann Oncol 15 (2): 218-23, 2004. [PUBMED Abstract]
Cellular Classification of Cervical Cancer
Squamous cell (epidermoid) carcinoma makes up approximately 90% of cervical cancers, and adenocarcinoma makes up 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 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.[3,4]
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.[3]
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
–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.
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.
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
aAdapted from FIGO Committee for Gynecologic Oncology.[3]
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.
Gold MA, Tian C, Whitney CW, et al.: Surgical versus radiographic determination of para-aortic lymph node metastases before chemoradiation for locally advanced cervical carcinoma: a Gynecologic Oncology Group Study. Cancer 112 (9): 1954-63, 2008. [PUBMED Abstract]
Epstein E, Testa A, Gaurilcikas A, et al.: Early-stage cervical cancer: tumor delineation by magnetic resonance imaging and ultrasound – a European multicenter trial. Gynecol Oncol 128 (3): 449-53, 2013. [PUBMED Abstract]
Bhatla N, Aoki D, Sharma DN, et al.: Cancer of the cervix uteri: 2021 update. Int J Gynaecol Obstet 155 (Suppl 1): 28-44, 2021. [PUBMED Abstract]
Olawaiye AB, Mutch DG, Bhosale P, et al.: Cervix uteri. In: Goodman KA, Gollub M, Eng C, et al.: AJCC Cancer Staging System. Version 9. American Joint Committee on Cancer; American College of Surgeons, 2020.
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).
Phase I and phase II clinical trials of new anticancer drugs
Chemoradiation Therapy
Five randomized phase III trials have shown an overall survival advantage for cisplatin-based therapy given concurrently with radiation therapy,[2–6] while one trial examining this regimen demonstrated no benefit.[7] 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 (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 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.
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.[2–6]
The DPYD gene encodes an enzyme that catabolizes pyrimidines and fluoropyrimidines, like capecitabine and fluorouracil. An estimated 1% to 2% of the population has germline pathogenic variants in DPYD, which lead to reduced DPD protein function and an accumulation of pyrimidines and fluoropyrimidines in the body.[11,12] Patients with the DPYD*2A variant who receive fluoropyrimidines may experience severe, life-threatening toxicities that are sometimes fatal. Many other DPYD variants have been identified, with a range of clinical effects.[11–13] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[14–16] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[17] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[18]
Surgery and Radiation Therapy
Surgery and radiation therapy are equally effective for early stage, small-volume disease.[19] 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.[20]
References
Lanciano RM, Won M, Hanks GE: A reappraisal of the International Federation of Gynecology and Obstetrics staging system for cervical cancer. A study of patterns of care. Cancer 69 (2): 482-7, 1992. [PUBMED Abstract]
Whitney CW, Sause W, Bundy BN, et al.: Randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an adjunct to radiation therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic lymph nodes: a Gynecologic Oncology Group and Southwest Oncology Group study. J Clin Oncol 17 (5): 1339-48, 1999. [PUBMED Abstract]
Morris M, Eifel PJ, Lu J, et al.: Pelvic radiation with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk cervical cancer. N Engl J Med 340 (15): 1137-43, 1999. [PUBMED Abstract]
Rose PG, Bundy BN, Watkins EB, et al.: Concurrent cisplatin-based radiotherapy and chemotherapy for locally advanced cervical cancer. N Engl J Med 340 (15): 1144-53, 1999. [PUBMED Abstract]
Keys HM, Bundy BN, Stehman FB, et al.: Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hysterectomy for bulky stage IB cervical carcinoma. N Engl J Med 340 (15): 1154-61, 1999. [PUBMED Abstract]
Peters WA, Liu PY, Barrett RJ, et al.: Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol 18 (8): 1606-13, 2000. [PUBMED Abstract]
Pearcey R, Brundage M, Drouin P, et al.: Phase III trial comparing radical radiotherapy with and without cisplatin chemotherapy in patients with advanced squamous cell cancer of the cervix. J Clin Oncol 20 (4): 966-72, 2002. [PUBMED Abstract]
Thomas GM: Improved treatment for cervical cancer–concurrent chemotherapy and radiotherapy. N Engl J Med 340 (15): 1198-200, 1999. [PUBMED Abstract]
Rose PG, Bundy BN: Chemoradiation for locally advanced cervical cancer: does it help? J Clin Oncol 20 (4): 891-3, 2002. [PUBMED Abstract]
Chemoradiotherapy for Cervical Cancer Meta-Analysis Collaboration: Reducing uncertainties about the effects of chemoradiotherapy for cervical cancer: a systematic review and meta-analysis of individual patient data from 18 randomized trials. J Clin Oncol 26 (35): 5802-12, 2008. [PUBMED Abstract]
Sharma BB, Rai K, Blunt H, et al.: Pathogenic DPYD Variants and Treatment-Related Mortality in Patients Receiving Fluoropyrimidine Chemotherapy: A Systematic Review and Meta-Analysis. Oncologist 26 (12): 1008-1016, 2021. [PUBMED Abstract]
Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
Shakeel F, Fang F, Kwon JW, et al.: Patients carrying DPYD variant alleles have increased risk of severe toxicity and related treatment modifications during fluoropyrimidine chemotherapy. Pharmacogenomics 22 (3): 145-155, 2021. [PUBMED Abstract]
Amstutz U, Henricks LM, Offer SM, et al.: Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther 103 (2): 210-216, 2018. [PUBMED Abstract]
Henricks LM, Lunenburg CATC, de Man FM, et al.: DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol 19 (11): 1459-1467, 2018. [PUBMED Abstract]
Lau-Min KS, Varughese LA, Nelson MN, et al.: Preemptive pharmacogenetic testing to guide chemotherapy dosing in patients with gastrointestinal malignancies: a qualitative study of barriers to implementation. BMC Cancer 22 (1): 47, 2022. [PUBMED Abstract]
Brooks GA, Tapp S, Daly AT, et al.: Cost-effectiveness of DPYD Genotyping Prior to Fluoropyrimidine-based Adjuvant Chemotherapy for Colon Cancer. Clin Colorectal Cancer 21 (3): e189-e195, 2022. [PUBMED Abstract]
Baker SD, Bates SE, Brooks GA, et al.: DPYD Testing: Time to Put Patient Safety First. J Clin Oncol 41 (15): 2701-2705, 2023. [PUBMED Abstract]
Eifel PJ, Burke TW, Delclos L, et al.: Early stage I adenocarcinoma of the uterine cervix: treatment results in patients with tumors less than or equal to 4 cm in diameter. Gynecol Oncol 41 (3): 199-205, 1991. [PUBMED Abstract]
Kovalic JJ, Grigsby PW, Perez CA, et al.: Cervical stump carcinoma. Int J Radiat Oncol Biol Phys 20 (5): 933-8, 1991. [PUBMED Abstract]
Treatment of In Situ Cervical Cancer
Consensus guidelines have been issued for managing women with cervical intraepithelial neoplasia or adenocarcinoma in situ.[1] 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.[2] 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.
Treatment Options for In Situ Cervical Cancer
Treatment options for in situ cervical cancer include:
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.[6]
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.[7–9] 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.[10]
Evidence (conization using LEEP):
A trial comparing LEEP with cold-knife cone biopsy showed no difference in the likelihood of complete excision of dysplasia.[6]
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.[11]
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 of 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.[12]
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.[13]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Wright TC, Massad LS, Dunton CJ, et al.: 2006 consensus guidelines for the management of women with cervical intraepithelial neoplasia or adenocarcinoma in situ. Am J Obstet Gynecol 197 (4): 340-5, 2007. [PUBMED Abstract]
Shumsky AG, Stuart GC, Nation J: Carcinoma of the cervix following conservative management of cervical intraepithelial neoplasia. Gynecol Oncol 53 (1): 50-4, 1994. [PUBMED Abstract]
Wright VC, Chapman W: Intraepithelial neoplasia of the lower female genital tract: etiology, investigation, and management. Semin Surg Oncol 8 (4): 180-90, 1992 Jul-Aug. [PUBMED Abstract]
Bloss JD: The use of electrosurgical techniques in the management of premalignant diseases of the vulva, vagina, and cervix: an excisional rather than an ablative approach. Am J Obstet Gynecol 169 (5): 1081-5, 1993. [PUBMED Abstract]
Tsukamoto N: Treatment of cervical intraepithelial neoplasia with the carbon dioxide laser. Gynecol Oncol 21 (3): 331-6, 1985. [PUBMED Abstract]
Girardi F, Heydarfadai M, Koroschetz F, et al.: Cold-knife conization versus loop excision: histopathologic and clinical results of a randomized trial. Gynecol Oncol 55 (3 Pt 1): 368-70, 1994. [PUBMED Abstract]
Wright TC, Gagnon S, Richart RM, et al.: Treatment of cervical intraepithelial neoplasia using the loop electrosurgical excision procedure. Obstet Gynecol 79 (2): 173-8, 1992. [PUBMED Abstract]
Naumann RW, Bell MC, Alvarez RD, et al.: LLETZ is an acceptable alternative to diagnostic cold-knife conization. Gynecol Oncol 55 (2): 224-8, 1994. [PUBMED Abstract]
Duesing N, Schwarz J, Choschzick M, et al.: Assessment of cervical intraepithelial neoplasia (CIN) with colposcopic biopsy and efficacy of loop electrosurgical excision procedure (LEEP). Arch Gynecol Obstet 286 (6): 1549-54, 2012. [PUBMED Abstract]
Widrich T, Kennedy AW, Myers TM, et al.: Adenocarcinoma in situ of the uterine cervix: management and outcome. Gynecol Oncol 61 (3): 304-8, 1996. [PUBMED Abstract]
Eddy GL, Spiegel GW, Creasman WT: Adverse effect of electrosurgical loop excision on assignment of FIGO stage in cervical cancer: report of two cases. Gynecol Oncol 55 (2): 313-7, 1994. [PUBMED Abstract]
Massad LS: New guidelines on cervical cancer screening: more than just the end of annual Pap testing. J Low Genit Tract Dis 16 (3): 172-4, 2012. [PUBMED Abstract]
Grigsby PW, Perez CA: Radiotherapy alone for medically inoperable carcinoma of the cervix: stage IA and carcinoma in situ. Int J Radiat Oncol Biol Phys 21 (2): 375-8, 1991. [PUBMED Abstract]
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.[1]
Total hysterectomy
If the depth of invasion is less than 3 mm, which is proven by cone biopsy with clear margins,[2] 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.
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%.[2] 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.[3] Patients with stages IA1 (with lymphovascular space invasion), IA2, and IB1 disease and histological subtypes of squamous cell, adenocarcinoma, or adenosquamous carcinoma were eligible for inclusion. The primary end point was noninferiority of MIS compared with open surgery. The metric used was the percent of disease-free patients at 4.5 years postsurgery. The secondary end points 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) rate at 4.5 years was 86% for the MIS group and 96.5% for the open-surgery group (95% confidence interval [CI], -16.4 to -4.7). At 3 years, the MIS group had a DFS rate 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 3-year overall survival (OS) rate (93.8% vs. 99.0% for the open-surgery group; HR for death from any cause, 6.0; 95% CI, 1.77–20.30).[3][Level of evidence A1]
The study concluded that MIS was inferior to an open abdominal approach and should not replace open surgery as the standard for patients with cervical cancer.
An epidemiological study used two large U.S. databases, the National Cancer Database (NCDB) and the 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.[4][Level of evidence C1]
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.
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 use 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.[5–9]
Intracavitary radiation therapy
Intracavitary radiation therapy is an option when palliative treatment is being considered for women who are not surgical candidates or who have other medical contraindications.
If the depth of invasion is less than 3 mm, 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.[10]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Sevin BU, Nadji M, Averette HE, et al.: Microinvasive carcinoma of the cervix. Cancer 70 (8): 2121-8, 1992. [PUBMED Abstract]
Jones WB, Mercer GO, Lewis JL, et al.: Early invasive carcinoma of the cervix. Gynecol Oncol 51 (1): 26-32, 1993. [PUBMED Abstract]
Ramirez PT, Frumovitz M, Pareja R, et al.: Minimally Invasive versus Abdominal Radical Hysterectomy for Cervical Cancer. N Engl J Med 379 (20): 1895-1904, 2018. [PUBMED Abstract]
Melamed A, Margul DJ, Chen L, et al.: Survival after Minimally Invasive Radical Hysterectomy for Early-Stage Cervical Cancer. N Engl J Med 379 (20): 1905-1914, 2018. [PUBMED Abstract]
Covens A, Shaw P, Murphy J, et al.: Is radical trachelectomy a safe alternative to radical hysterectomy for patients with stage IA-B carcinoma of the cervix? Cancer 86 (11): 2273-9, 1999. [PUBMED Abstract]
Dargent D, Martin X, Sacchetoni A, et al.: Laparoscopic vaginal radical trachelectomy: a treatment to preserve the fertility of cervical carcinoma patients. Cancer 88 (8): 1877-82, 2000. [PUBMED Abstract]
Plante M, Renaud MC, Hoskins IA, et al.: Vaginal radical trachelectomy: a valuable fertility-preserving option in the management of early-stage cervical cancer. A series of 50 pregnancies and review of the literature. Gynecol Oncol 98 (1): 3-10, 2005. [PUBMED Abstract]
Shepherd JH, Spencer C, Herod J, et al.: Radical vaginal trachelectomy as a fertility-sparing procedure in women with early-stage cervical cancer-cumulative pregnancy rate in a series of 123 women. BJOG 113 (6): 719-24, 2006. [PUBMED Abstract]
Wethington SL, Cibula D, Duska LR, et al.: An international series on abdominal radical trachelectomy: 101 patients and 28 pregnancies. Int J Gynecol Cancer 22 (7): 1251-7, 2012. [PUBMED Abstract]
Grigsby PW, Perez CA: Radiotherapy alone for medically inoperable carcinoma of the cervix: stage IA and carcinoma in situ. Int J Radiat Oncol Biol Phys 21 (2): 375-8, 1991. [PUBMED Abstract]
Treatment of Stages IB and IIA Cervical Cancer
Treatment Options for Stages IB and IIA Cervical Cancer
Tumor size is an important prognostic factor to carefully evaluate when 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.[2]
In patients with stage IB2 disease, for tumors that expand the cervix more than 4 cm, the primary treatment should be concomitant chemotherapy and radiation therapy.[3]
Radiation therapy with concomitant chemotherapy
Concurrent cisplatin-based chemotherapy with radiation therapy is the standard of care for women who require radiation therapy for treatment of cervical cancer.[4–10] 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,[4–7] while one trial that examined this regimen demonstrated no benefit.[8] 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.
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 has the advantages 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.[11,12]
Evidence (brachytherapy):
In three randomized trials, HDR brachytherapy was comparable with LDR brachytherapy in terms of local-regional control and complication rates.[13–15][Level of evidence B1]
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.[16]
Pelvic node disease
The resection of macroscopically involved pelvic nodes may improve rates of local control with postoperative radiation therapy.[17] Patients who underwent extraperitoneal lymph–node sampling had fewer bowel complications than those who had transperitoneal lymph–node sampling.[18–20] Patients with close vaginal margins (<0.5 cm) may also benefit from pelvic radiation therapy.[21]
Radical hysterectomy and bilateral pelvic lymphadenectomy with or without total pelvic radiation therapy plus chemotherapy
Women with stages IB to IIA disease may consider radical hysterectomy and bilateral pelvic lymphadenectomy.
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 pathological 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.[2][Level of evidence A1]
The primary outcome was 5-year OS, with secondary measures of rate of recurrence and complications. With a median follow-up of 87 months, the OS rate was the same in both groups at 83% (hazard ratio [HR], 1.2; 95% 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.[22] Patients with stages IA1 (with lymphovascular space invasion), IA2, and IB1 disease and histological subtypes of squamous cell, adenocarcinoma or adenosquamous carcinoma were eligible for inclusion. The primary end point was noninferiority of MIS compared with open surgery; the metric used was the percent of disease-free patients at 4.5 years postsurgery. The secondary end points 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 rate at 4.5 years was 86% for the MIS group and 96.5% for the open-surgery group (95% CI, -16.4 to -4.7). At 3 years, the MIS group had a DFS rate of 91.2% versus 97.1% for the open-surgery group (HR for disease recurrence or death, 3.74; 95% CI, 1.63–8.58).
The MIS group also had a lower 3-year OS rate (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 A1]
The study concluded that MIS was inferior to an open abdominal approach and should not replace open surgery as the standard for cervical cancer patients.
An epidemiological study using two large U.S. databases (National Cancer Database [NCDB] and Surveillance, Epidemiology, and End Results [SEER] Program database) 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.[23][Level of evidence C1]
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 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.[24] 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 (1,000 mg/m2/d every 3 weeks for four cycles); the first two cycles were concurrent with the radiation therapy.[6][Level of evidence A1]
There were 268 patients evaluated with a primary end point of OS. The study results were reported early because of the positive results in other trials of concomitant cisplatin and radiation therapy.
The estimated 4-year survival rate 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 pathological 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.[25] 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.[18] 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 histological evidence of disease.[26] 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.[26] The use of intensity-modulated radiation therapy (IMRT) may minimize the effects to the small bowel usually associated with this treatment.[27]
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:
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.[28–32]
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.
Immunotherapy
Evidence (immunotherapy):
KEYNOTE-A18 (NCT04221945) was a multicenter, phase III, randomized trial that included 1,060 women with newly diagnosed squamous cell carcinoma, adenocarcinoma, or adenosquamous carcinoma of the cervix. Patients had stage IB2 or IIB node-positive disease or stage III to IVA disease and had received no prior treatment. Patients were randomly assigned to receive either chemoradiation therapy (with cisplatin) plus pembrolizumab (every 3 weeks for five cycles) or chemoradiation therapy plus placebo. All patients received maintenance therapy with pembrolizumab or placebo every 6 weeks for 15 cycles. All patients received brachytherapy, and 94% of patients had programmed death-ligand 1 (PD-L1)–positive disease. The dual primary end points were progression-free survival (PFS) and OS. The median follow-up time was 17.9 months.[33]
The 24-month OS rate was 87% in the pembrolizumab group and 81% in the placebo group (HR, 0.73; 95% CI, 0.49–1.07).[33][Level of evidence B1]
There was a statistically significant improvement in PFS (HR, 0.70; 95% CI, 0.55–0.89). The 24-month PFS rate was 68% in the pembrolizumab group and 57% in the placebo group.
Patients with stage III or IV disease had a greater improvement in PFS (HR, 0.58; 95% CI, 0.42–0.80).
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.[34–38] Multiple regimens have been used; however, almost all use 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.[39] As a result, the control arm received radiation therapy alone. Although there was an improvement in OS for the experimental arm, the results do not reflect 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 end point, this trial may delineate whether there is a role for neoadjuvant chemotherapy for this patient population.
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.[40–42] A Radiation Therapy Oncology Group (RTOG) phase II trial (RTOG-0418 [NCT00331760]) evaluated 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. General information about clinical trials is also available.
References
Perez CA, Grigsby PW, Nene SM, et al.: Effect of tumor size on the prognosis of carcinoma of the uterine cervix treated with irradiation alone. Cancer 69 (11): 2796-806, 1992. [PUBMED Abstract]
Landoni F, Maneo A, Colombo A, et al.: Randomised study of radical surgery versus radiotherapy for stage Ib-IIa cervical cancer. Lancet 350 (9077): 535-40, 1997. [PUBMED Abstract]
Eifel PJ, Burke TW, Delclos L, et al.: Early stage I adenocarcinoma of the uterine cervix: treatment results in patients with tumors less than or equal to 4 cm in diameter. Gynecol Oncol 41 (3): 199-205, 1991. [PUBMED Abstract]
Morris M, Eifel PJ, Lu J, et al.: Pelvic radiation with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk cervical cancer. N Engl J Med 340 (15): 1137-43, 1999. [PUBMED Abstract]
Keys HM, Bundy BN, Stehman FB, et al.: Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hysterectomy for bulky stage IB cervical carcinoma. N Engl J Med 340 (15): 1154-61, 1999. [PUBMED Abstract]
Peters WA, Liu PY, Barrett RJ, et al.: Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol 18 (8): 1606-13, 2000. [PUBMED Abstract]
Thomas GM: Improved treatment for cervical cancer–concurrent chemotherapy and radiotherapy. N Engl J Med 340 (15): 1198-200, 1999. [PUBMED Abstract]
Pearcey R, Brundage M, Drouin P, et al.: Phase III trial comparing radical radiotherapy with and without cisplatin chemotherapy in patients with advanced squamous cell cancer of the cervix. J Clin Oncol 20 (4): 966-72, 2002. [PUBMED Abstract]
Rose PG, Bundy BN: Chemoradiation for locally advanced cervical cancer: does it help? J Clin Oncol 20 (4): 891-3, 2002. [PUBMED Abstract]
Chemoradiotherapy for Cervical Cancer Meta-Analysis Collaboration: Reducing uncertainties about the effects of chemoradiotherapy for cervical cancer: a systematic review and meta-analysis of individual patient data from 18 randomized trials. J Clin Oncol 26 (35): 5802-12, 2008. [PUBMED Abstract]
Nag S, Chao C, Erickson B, et al.: The American Brachytherapy Society recommendations for low-dose-rate brachytherapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 52 (1): 33-48, 2002. [PUBMED Abstract]
Nag S, Erickson B, Thomadsen B, et al.: The American Brachytherapy Society recommendations for high-dose-rate brachytherapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 48 (1): 201-11, 2000. [PUBMED Abstract]
Patel FD, Sharma SC, Negi PS, et al.: Low dose rate vs. high dose rate brachytherapy in the treatment of carcinoma of the uterine cervix: a clinical trial. Int J Radiat Oncol Biol Phys 28 (2): 335-41, 1994. [PUBMED Abstract]
Hareyama M, Sakata K, Oouchi A, et al.: High-dose-rate versus low-dose-rate intracavitary therapy for carcinoma of the uterine cervix: a randomized trial. Cancer 94 (1): 117-24, 2002. [PUBMED Abstract]
Lertsanguansinchai P, Lertbutsayanukul C, Shotelersuk K, et al.: Phase III randomized trial comparing LDR and HDR brachytherapy in treatment of cervical carcinoma. Int J Radiat Oncol Biol Phys 59 (5): 1424-31, 2004. [PUBMED Abstract]
Thoms WW, Eifel PJ, Smith TL, et al.: Bulky endocervical carcinoma: a 23-year experience. Int J Radiat Oncol Biol Phys 23 (3): 491-9, 1992. [PUBMED Abstract]
Downey GO, Potish RA, Adcock LL, et al.: Pretreatment surgical staging in cervical carcinoma: therapeutic efficacy of pelvic lymph node resection. Am J Obstet Gynecol 160 (5 Pt 1): 1055-61, 1989. [PUBMED Abstract]
Vigliotti AP, Wen BC, Hussey DH, et al.: Extended field irradiation for carcinoma of the uterine cervix with positive periaortic nodes. Int J Radiat Oncol Biol Phys 23 (3): 501-9, 1992. [PUBMED Abstract]
Weiser EB, Bundy BN, Hoskins WJ, et al.: Extraperitoneal versus transperitoneal selective paraaortic lymphadenectomy in the pretreatment surgical staging of advanced cervical carcinoma (a Gynecologic Oncology Group study). Gynecol Oncol 33 (3): 283-9, 1989. [PUBMED Abstract]
Fine BA, Hempling RE, Piver MS, et al.: Severe radiation morbidity in carcinoma of the cervix: impact of pretherapy surgical staging and previous surgery. Int J Radiat Oncol Biol Phys 31 (4): 717-23, 1995. [PUBMED Abstract]
Estape RE, Angioli R, Madrigal M, et al.: Close vaginal margins as a prognostic factor after radical hysterectomy. Gynecol Oncol 68 (3): 229-32, 1998. [PUBMED Abstract]
Ramirez PT, Frumovitz M, Pareja R, et al.: Minimally Invasive versus Abdominal Radical Hysterectomy for Cervical Cancer. N Engl J Med 379 (20): 1895-1904, 2018. [PUBMED Abstract]
Melamed A, Margul DJ, Chen L, et al.: Survival after Minimally Invasive Radical Hysterectomy for Early-Stage Cervical Cancer. N Engl J Med 379 (20): 1905-1914, 2018. [PUBMED Abstract]
Sedlis A, Bundy BN, Rotman MZ, et al.: A randomized trial of pelvic radiation therapy versus no further therapy in selected patients with stage IB carcinoma of the cervix after radical hysterectomy and pelvic lymphadenectomy: A Gynecologic Oncology Group Study. Gynecol Oncol 73 (2): 177-83, 1999. [PUBMED Abstract]
Cunningham MJ, Dunton CJ, Corn B, et al.: Extended-field radiation therapy in early-stage cervical carcinoma: survival and complications. Gynecol Oncol 43 (1): 51-4, 1991. [PUBMED Abstract]
Rotman M, Pajak TF, Choi K, et al.: Prophylactic extended-field irradiation of para-aortic lymph nodes in stages IIB and bulky IB and IIA cervical carcinomas. Ten-year treatment results of RTOG 79-20. JAMA 274 (5): 387-93, 1995. [PUBMED Abstract]
Poorvu PD, Sadow CA, Townamchai K, et al.: Duodenal and other gastrointestinal toxicity in cervical and endometrial cancer treated with extended-field intensity modulated radiation therapy to paraaortic lymph nodes. Int J Radiat Oncol Biol Phys 85 (5): 1262-8, 2013. [PUBMED Abstract]
Covens A, Shaw P, Murphy J, et al.: Is radical trachelectomy a safe alternative to radical hysterectomy for patients with stage IA-B carcinoma of the cervix? Cancer 86 (11): 2273-9, 1999. [PUBMED Abstract]
Dargent D, Martin X, Sacchetoni A, et al.: Laparoscopic vaginal radical trachelectomy: a treatment to preserve the fertility of cervical carcinoma patients. Cancer 88 (8): 1877-82, 2000. [PUBMED Abstract]
Plante M, Renaud MC, Hoskins IA, et al.: Vaginal radical trachelectomy: a valuable fertility-preserving option in the management of early-stage cervical cancer. A series of 50 pregnancies and review of the literature. Gynecol Oncol 98 (1): 3-10, 2005. [PUBMED Abstract]
Shepherd JH, Spencer C, Herod J, et al.: Radical vaginal trachelectomy as a fertility-sparing procedure in women with early-stage cervical cancer-cumulative pregnancy rate in a series of 123 women. BJOG 113 (6): 719-24, 2006. [PUBMED Abstract]
Wethington SL, Cibula D, Duska LR, et al.: An international series on abdominal radical trachelectomy: 101 patients and 28 pregnancies. Int J Gynecol Cancer 22 (7): 1251-7, 2012. [PUBMED Abstract]
Lorusso D, Xiang Y, Hasegawa K, et al.: Pembrolizumab or placebo with chemoradiotherapy followed by pembrolizumab or placebo for newly diagnosed, high-risk, locally advanced cervical cancer (ENGOT-cx11/GOG-3047/KEYNOTE-A18): a randomised, double-blind, phase 3 clinical trial. Lancet 403 (10434): 1341-1350, 2024. [PUBMED Abstract]
Ferrandina G, Margariti PA, Smaniotto D, et al.: Long-term analysis of clinical outcome and complications in locally advanced cervical cancer patients administered concomitant chemoradiation followed by radical surgery. Gynecol Oncol 119 (3): 404-10, 2010. [PUBMED Abstract]
Ferrandina G, Distefano MG, De Vincenzo R, et al.: Paclitaxel, epirubicin, and cisplatin (TEP) regimen as neoadjuvant treatment in locally advanced cervical cancer: long-term results. Gynecol Oncol 128 (3): 518-23, 2013. [PUBMED Abstract]
Zanaboni F, Grijuela B, Giudici S, et al.: Weekly topotecan and cisplatin (TOPOCIS) as neo-adjuvant chemotherapy for locally-advanced squamous cervical carcinoma: Results of a phase II multicentric study. Eur J Cancer 49 (5): 1065-72, 2013. [PUBMED Abstract]
Manci N, Marchetti C, Di Tucci C, et al.: A prospective phase II study of topotecan (Hycamtin®) and cisplatin as neoadjuvant chemotherapy in locally advanced cervical cancer. Gynecol Oncol 122 (2): 285-90, 2011. [PUBMED Abstract]
Gong L, Lou JY, Wang P, et al.: Clinical evaluation of neoadjuvant chemotherapy followed by radical surgery in the management of stage IB2-IIB cervical cancer. Int J Gynaecol Obstet 117 (1): 23-6, 2012. [PUBMED Abstract]
Benedetti-Panici P, Greggi S, Colombo A, et al.: Neoadjuvant chemotherapy and radical surgery versus exclusive radiotherapy in locally advanced squamous cell cervical cancer: results from the Italian multicenter randomized study. J Clin Oncol 20 (1): 179-88, 2002. [PUBMED Abstract]
Chen MF, Tseng CJ, Tseng CC, et al.: Adjuvant concurrent chemoradiotherapy with intensity-modulated pelvic radiotherapy after surgery for high-risk, early stage cervical cancer patients. Cancer J 14 (3): 200-6, 2008 May-Jun. [PUBMED Abstract]
Hasselle MD, Rose BS, Kochanski JD, et al.: Clinical outcomes of intensity-modulated pelvic radiation therapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 80 (5): 1436-45, 2011. [PUBMED Abstract]
Folkert MR, Shih KK, Abu-Rustum NR, et al.: Postoperative pelvic intensity-modulated radiotherapy and concurrent chemotherapy in intermediate- and high-risk cervical cancer. Gynecol Oncol 128 (2): 288-93, 2013. [PUBMED Abstract]
Treatment of Stages IIB, III, and IVA Cervical Cancer
Treatment Options for Stages IIB, III, and IVA Cervical Cancer
Primary tumor size is an important prognostic factor to carefully evaluate when choosing optimal therapy.[1] Survival and local control are better with unilateral rather than bilateral parametrial involvement.[2] Patterns-of-care studies in patients with stages IIIA and IIIB disease 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.[2] 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.[3]
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).[5–12]
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,[5–10] but one trial that examined this regimen demonstrated no benefit.[13] 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.[14,15]
In three randomized trials, HDR brachytherapy was comparable with LDR brachytherapy in terms of local-regional control and complication rates.[16–18][Level of evidence B1]
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.[19][Level of evidence A1] 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 end point 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.[20] 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.
Interstitial brachytherapy
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.[21]
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.[22–26] Multiple regimens have been used; however, almost all use 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.[27] 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 end point, this trial may delineate whether there is a role for neoadjuvant chemotherapy for this patient population.
Immunotherapy
Evidence (immunotherapy):
KEYNOTE-A18 (NCT04221945) was a multicenter, phase III, randomized trial that included 1,060 women with newly diagnosed squamous cell carcinoma, adenocarcinoma, or adenosquamous carcinoma of the cervix. Patients had stage IB2 or IIB node-positive disease or stage III to IVA disease and had received no prior treatment. Patients were randomly assigned to receive either chemoradiation therapy (with cisplatin) plus pembrolizumab (every 3 weeks for five cycles) or chemoradiation therapy plus placebo. All patients received maintenance therapy with pembrolizumab or placebo every 6 weeks for 15 cycles. All patients received brachytherapy, and 94% of patients had programmed death-ligand 1 (PD-L1)–positive disease. The dual primary end points were PFS and OS. The median follow-up time was 17.9 months.[28]
The 24-month OS rate was 87% in the pembrolizumab group and 81% in the placebo group (HR, 0.73; 95% CI, 0.49–1.07).[28][Level of evidence B1]
There was a statistically significant improvement in PFS (HR, 0.70; 95% CI, 0.55–0.89). The 24-month PFS rate was 68% in the pembrolizumab group and 57% in the placebo group.
Patients with stage III or IV disease had a greater improvement in PFS (HR, 0.58; 95% CI, 0.42–0.80).
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.[29] 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.[30] A single study (RTOG-7920) showed a survival advantage in patients who received radiation therapy to para-aortic nodes without histological evidence of disease.[31] Toxic effects are greater with para-aortic radiation than with pelvic radiation alone but were mostly confined to patients with previous abdominopelvic surgery.[31]
If postoperative EBRT is planned following surgery, extraperitoneal lymph–node sampling is associated with fewer radiation-induced complications than a transperitoneal approach.[32] Patients who underwent extraperitoneal lymph–node sampling had fewer bowel complications than those who had transperitoneal lymph–node sampling.[30,32,33]
The resection of macroscopically involved pelvic nodes may improve rates of local control with postoperative radiation therapy.[34] In addition, prospective data indicated improved outcomes for patients who underwent resection of positive para-aortic lymph nodes before curative intent chemoradiation therapy; however, only patients with minimal nodal involvement (<5 mm) benefited.[35]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Perez CA, Grigsby PW, Nene SM, et al.: Effect of tumor size on the prognosis of carcinoma of the uterine cervix treated with irradiation alone. Cancer 69 (11): 2796-806, 1992. [PUBMED Abstract]
Lanciano RM, Won M, Hanks GE: A reappraisal of the International Federation of Gynecology and Obstetrics staging system for cervical cancer. A study of patterns of care. Cancer 69 (2): 482-7, 1992. [PUBMED Abstract]
Lanciano RM, Martz K, Coia LR, et al.: Tumor and treatment factors improving outcome in stage III-B cervix cancer. Int J Radiat Oncol Biol Phys 20 (1): 95-100, 1991. [PUBMED Abstract]
Monk BJ, Tewari KS, Koh WJ: Multimodality therapy for locally advanced cervical carcinoma: state of the art and future directions. J Clin Oncol 25 (20): 2952-65, 2007. [PUBMED Abstract]
Whitney CW, Sause W, Bundy BN, et al.: Randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an adjunct to radiation therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic lymph nodes: a Gynecologic Oncology Group and Southwest Oncology Group study. J Clin Oncol 17 (5): 1339-48, 1999. [PUBMED Abstract]
Morris M, Eifel PJ, Lu J, et al.: Pelvic radiation with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk cervical cancer. N Engl J Med 340 (15): 1137-43, 1999. [PUBMED Abstract]
Rose PG, Bundy BN, Watkins EB, et al.: Concurrent cisplatin-based radiotherapy and chemotherapy for locally advanced cervical cancer. N Engl J Med 340 (15): 1144-53, 1999. [PUBMED Abstract]
Keys HM, Bundy BN, Stehman FB, et al.: Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hysterectomy for bulky stage IB cervical carcinoma. N Engl J Med 340 (15): 1154-61, 1999. [PUBMED Abstract]
Peters WA, Liu PY, Barrett RJ, et al.: Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol 18 (8): 1606-13, 2000. [PUBMED Abstract]
Thomas GM: Improved treatment for cervical cancer–concurrent chemotherapy and radiotherapy. N Engl J Med 340 (15): 1198-200, 1999. [PUBMED Abstract]
Rose PG, Bundy BN: Chemoradiation for locally advanced cervical cancer: does it help? J Clin Oncol 20 (4): 891-3, 2002. [PUBMED Abstract]
Chemoradiotherapy for Cervical Cancer Meta-Analysis Collaboration: Reducing uncertainties about the effects of chemoradiotherapy for cervical cancer: a systematic review and meta-analysis of individual patient data from 18 randomized trials. J Clin Oncol 26 (35): 5802-12, 2008. [PUBMED Abstract]
Pearcey R, Brundage M, Drouin P, et al.: Phase III trial comparing radical radiotherapy with and without cisplatin chemotherapy in patients with advanced squamous cell cancer of the cervix. J Clin Oncol 20 (4): 966-72, 2002. [PUBMED Abstract]
Nag S, Chao C, Erickson B, et al.: The American Brachytherapy Society recommendations for low-dose-rate brachytherapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 52 (1): 33-48, 2002. [PUBMED Abstract]
Nag S, Erickson B, Thomadsen B, et al.: The American Brachytherapy Society recommendations for high-dose-rate brachytherapy for carcinoma of the cervix. Int J Radiat Oncol Biol Phys 48 (1): 201-11, 2000. [PUBMED Abstract]
Patel FD, Sharma SC, Negi PS, et al.: Low dose rate vs. high dose rate brachytherapy in the treatment of carcinoma of the uterine cervix: a clinical trial. Int J Radiat Oncol Biol Phys 28 (2): 335-41, 1994. [PUBMED Abstract]
Hareyama M, Sakata K, Oouchi A, et al.: High-dose-rate versus low-dose-rate intracavitary therapy for carcinoma of the uterine cervix: a randomized trial. Cancer 94 (1): 117-24, 2002. [PUBMED Abstract]
Lertsanguansinchai P, Lertbutsayanukul C, Shotelersuk K, et al.: Phase III randomized trial comparing LDR and HDR brachytherapy in treatment of cervical carcinoma. Int J Radiat Oncol Biol Phys 59 (5): 1424-31, 2004. [PUBMED Abstract]
Dueñas-González A, Zarbá JJ, Patel F, et al.: Phase III, open-label, randomized study comparing concurrent gemcitabine plus cisplatin and radiation followed by adjuvant gemcitabine and cisplatin versus concurrent cisplatin and radiation in patients with stage IIB to IVA carcinoma of the cervix. J Clin Oncol 29 (13): 1678-85, 2011. [PUBMED Abstract]
Dueňas-González A, Orlando M, Zhou Y, et al.: Efficacy in high burden locally advanced cervical cancer with concurrent gemcitabine and cisplatin chemoradiotherapy plus adjuvant gemcitabine and cisplatin: prognostic and predictive factors and the impact of disease stage on outcomes from a prospective randomized phase III trial. Gynecol Oncol 126 (3): 334-40, 2012. [PUBMED Abstract]
Pinn-Bingham M, Puthawala AA, Syed AM, et al.: Outcomes of high-dose-rate interstitial brachytherapy in the treatment of locally advanced cervical cancer: long-term results. Int J Radiat Oncol Biol Phys 85 (3): 714-20, 2013. [PUBMED Abstract]
Ferrandina G, Margariti PA, Smaniotto D, et al.: Long-term analysis of clinical outcome and complications in locally advanced cervical cancer patients administered concomitant chemoradiation followed by radical surgery. Gynecol Oncol 119 (3): 404-10, 2010. [PUBMED Abstract]
Ferrandina G, Distefano MG, De Vincenzo R, et al.: Paclitaxel, epirubicin, and cisplatin (TEP) regimen as neoadjuvant treatment in locally advanced cervical cancer: long-term results. Gynecol Oncol 128 (3): 518-23, 2013. [PUBMED Abstract]
Zanaboni F, Grijuela B, Giudici S, et al.: Weekly topotecan and cisplatin (TOPOCIS) as neo-adjuvant chemotherapy for locally-advanced squamous cervical carcinoma: Results of a phase II multicentric study. Eur J Cancer 49 (5): 1065-72, 2013. [PUBMED Abstract]
Manci N, Marchetti C, Di Tucci C, et al.: A prospective phase II study of topotecan (Hycamtin®) and cisplatin as neoadjuvant chemotherapy in locally advanced cervical cancer. Gynecol Oncol 122 (2): 285-90, 2011. [PUBMED Abstract]
Gong L, Lou JY, Wang P, et al.: Clinical evaluation of neoadjuvant chemotherapy followed by radical surgery in the management of stage IB2-IIB cervical cancer. Int J Gynaecol Obstet 117 (1): 23-6, 2012. [PUBMED Abstract]
Benedetti-Panici P, Greggi S, Colombo A, et al.: Neoadjuvant chemotherapy and radical surgery versus exclusive radiotherapy in locally advanced squamous cell cervical cancer: results from the Italian multicenter randomized study. J Clin Oncol 20 (1): 179-88, 2002. [PUBMED Abstract]
Lorusso D, Xiang Y, Hasegawa K, et al.: Pembrolizumab or placebo with chemoradiotherapy followed by pembrolizumab or placebo for newly diagnosed, high-risk, locally advanced cervical cancer (ENGOT-cx11/GOG-3047/KEYNOTE-A18): a randomised, double-blind, phase 3 clinical trial. Lancet 403 (10434): 1341-1350, 2024. [PUBMED Abstract]
Cunningham MJ, Dunton CJ, Corn B, et al.: Extended-field radiation therapy in early-stage cervical carcinoma: survival and complications. Gynecol Oncol 43 (1): 51-4, 1991. [PUBMED Abstract]
Vigliotti AP, Wen BC, Hussey DH, et al.: Extended field irradiation for carcinoma of the uterine cervix with positive periaortic nodes. Int J Radiat Oncol Biol Phys 23 (3): 501-9, 1992. [PUBMED Abstract]
Rotman M, Pajak TF, Choi K, et al.: Prophylactic extended-field irradiation of para-aortic lymph nodes in stages IIB and bulky IB and IIA cervical carcinomas. Ten-year treatment results of RTOG 79-20. JAMA 274 (5): 387-93, 1995. [PUBMED Abstract]
Weiser EB, Bundy BN, Hoskins WJ, et al.: Extraperitoneal versus transperitoneal selective paraaortic lymphadenectomy in the pretreatment surgical staging of advanced cervical carcinoma (a Gynecologic Oncology Group study). Gynecol Oncol 33 (3): 283-9, 1989. [PUBMED Abstract]
Fine BA, Hempling RE, Piver MS, et al.: Severe radiation morbidity in carcinoma of the cervix: impact of pretherapy surgical staging and previous surgery. Int J Radiat Oncol Biol Phys 31 (4): 717-23, 1995. [PUBMED Abstract]
Downey GO, Potish RA, Adcock LL, et al.: Pretreatment surgical staging in cervical carcinoma: therapeutic efficacy of pelvic lymph node resection. Am J Obstet Gynecol 160 (5 Pt 1): 1055-61, 1989. [PUBMED Abstract]
Gouy S, Morice P, Narducci F, et al.: Prospective multicenter study evaluating the survival of patients with locally advanced cervical cancer undergoing laparoscopic para-aortic lymphadenectomy before chemoradiotherapy in the era of positron emission tomography imaging. J Clin Oncol 31 (24): 3026-33, 2013. [PUBMED Abstract]
Treatment of Stage IVB and Recurrent Cervical Cancer
Treatment Options for Stage IVB and 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 stage IVB and recurrent cervical cancer include:
Phase I and phase II clinical trials of new anticancer drugs.
Immunotherapy
Based on the results of the phase II KEYNOTE-158 trial (NCT02628067), the U.S. Food and Drug Administration (FDA) approved the anti–programmed cell death-1 (PD-1) immune checkpoint inhibitor, pembrolizumab, for women with recurrent or metastatic cervical cancer whose tumors express programmed death-ligand 1 (PD-L1) (combined positive score [CPS], ≥1). Additional data on the benefits of pembrolizumab have been gathered from several trials.
Evidence (immunotherapy):
The international multicenter BEATcc trial (NCT03556839) included 410 women with stage IVB, persistent, or recurrent squamous cell or adenocarcinoma of the cervix not amenable to curative surgery or radiation therapy. Patients had not received previous systemic therapy. Patients were randomly assigned to receive either bevacizumab plus chemotherapy (carboplatin or cisplatin with paclitaxel) (the control arm) or the experimental arm, which added atezolizumab to the control regimen. The dual primary end points were progression-free survival (PFS) and overall survival (OS), and the trial was biomarker unselective for PD-L1 expression.[1]
After a median follow-up of 32.9 months, the median PFS was 10.4 months for patients in the control arm and 13.7 months for patients in the experimental arm (hazard ratio [HR], 0.62; 95% confidence interval [CI], 0.48–0.78; P < .0001). The median OS was 22.8 months for patients in the control arm and 32.1 months for patients in the experimental arm (HR, 0.68; 95% CI, 0.52–0.88; P = .0046).[1][Level of evidence A1]
KEYNOTE-826 (NCT03635567) was a multicenter, phase III, randomized trial that included 617 women with persistent, recurrent, or metastatic adenosquamous or squamous cell carcinoma of the cervix not amenable to curative treatment. Patients had measurable disease. All patients were treated with cisplatin or carboplatin combined with paclitaxel, with the option to add bevacizumab at the treating physician’s discretion. Patients received six cycles and were randomly assigned 1:1 to receive pembrolizumab or placebo every 3 weeks for up to 35 cycles. The median follow-up was 39.1 months, with dual primary end points of PFS and OS. Approximately 89% of women had a PD-L1 CPS score of 1 or higher.[2]
The median OS was 26.4 months for patients who received pembrolizumab and 16.8 months for patients who received placebo (HR, 0.63; 95% CI, 0.52–0.77). This benefit was not maintained in women with a CPS score less than 1. The benefit was maintained in the group of women who did not receive bevacizumab.[2][Level of evidence A1]
The median PFS was 10.4 months for patients who received pembrolizumab and 8.2 months for patients who received placebo (HR, 0.61; 95% CI, 0.50–0.74).
KEYNOTE-A18 (NCT04221945) was a multicenter, phase III, randomized trial that included 1,060 women with newly diagnosed squamous cell carcinoma, adenocarcinoma, or adenosquamous carcinoma of the cervix. Patients had stage IB2 or IIB node-positive disease or stage III to IVA disease and had received no prior treatment. Patients were randomly assigned to receive either chemoradiation therapy (with cisplatin) plus pembrolizumab (every 3 weeks for five cycles) or chemoradiation therapy plus placebo. All patients received maintenance therapy with pembrolizumab or placebo every 6 weeks for 15 cycles. All patients received brachytherapy, and 94% of patients had PD-L1–positive disease. The dual primary end points were PFS and OS. The median follow-up time was 17.9 months.[3]
The 24-month OS rate was 87% in the pembrolizumab group and 81% in the placebo group (HR, 0.73; 95% CI, 0.49–1.07).[3][Level of evidence B1]
There was a statistically significant improvement in PFS (HR, 0.70; 95% CI, 0.55–0.89). The 24-month PFS rate was 68% in the pembrolizumab group and 57% in the placebo group.
Patients with stage III or IV disease had a greater improvement in PFS (HR, 0.58; 95% CI, 0.42–0.80).
KEYNOTE-158 (NCT02628067) was a multicenter nonrandomized trial that included 98 patients with recurrent or metastatic cervical cancer. Patients received 200 mg of pembrolizumab every 3 weeks intravenously until unacceptable toxicity or disease progression.[4] 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.
The median PFS was 2.1 months, and OS 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%).[4][Level of evidence B4]
GOG-0240 (NCT00803062), which enrolled patients with metastatic, persistent, or recurrent cervical carcinoma, was designed to answer the following two questions:[5]
Can a nonplatinum combination show improvement over the standard of cisplatin-paclitaxel in this population, which was previously treated with cisplatin during radiation therapy?
Can the addition of bevacizumab improve combination chemotherapy in patients with stage IVB, persistent, or recurrent cervical cancer?
Patients were randomly assigned to one of 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) on days 1–3 + paclitaxel (175 mg/m2) on day 1 (PT).
PT + bevacizumab (15 mg/kg) on day 1.
Additional study methods and results included the following:
The primary end point was OS, and 452 patients were evaluable.
The combination PT was not superior to PC and had a HRdeath 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: (1) improved OS: 17 months for chemotherapy plus bevacizumab versus 13.3 months for chemotherapy alone (HR, 0.71; 98% CI, 0.54–0.95) and (2) 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 who received 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 who received chemotherapy alone.
As a result, the addition of bevacizumab may be considered for this patient population.
In a phase III international trial (NCT04697628) 502 women were randomly assigned to receive either tisotumab vedotin (2 mg/kg every 3 weeks) or the investigator’s choice of chemotherapy. Patients had metastatic or recurrent cervical cancer (squamous cell, adenocarcinoma, or adenosquamous histology) with measurable disease after prior systemic therapy (paclitaxel plus a platinum or topotecan, with or without bevacizumab and with or without an anti–PD-1 or anti–PD-L1 agent). One or two prior systemic treatments were required (not including adjuvant, maintenance, or neoadjuvant chemotherapy or chemoradiation therapy). Imaging was done every 6 weeks for 30 weeks and then every 12 weeks thereafter. The primary end point was OS.[6]
After a median follow-up of 10.8 months, the median OS was 11.5 months for patients who received tisotumab vedotin and 9.5 months for patients who received chemotherapy.[6][Level of evidence A1]
Common adverse events in the tisotumab vedotin group included ocular events (52.8%), peripheral neuropathy (38.4%), and bleeding (42%).
Radiation therapy and chemotherapy
Radiation therapy and chemotherapy (fluorouracil with or without mitomycin) may cure 40% to 50% of patients with recurrence in the pelvis after initial radical surgery.[7]
Palliative chemotherapy and other systemic therapy
Chemotherapy can be used for palliation. Drugs used for palliative chemotherapy are shown in Table 6.
Table 6. Drugs Used to Treat Stage IVB and Recurrent Cervical Cancer
Since the drug was initially introduced in the 1970s, the regimen used most often to treat recurrent cervical cancer has been single-agent cisplatin given intravenously at 50 mg/m² every 3 weeks.[8] The Gynecologic Oncology Group (GOG) reported on sequential randomized trials of combination chemotherapy for stage IVB, recurrent, or persistent cervical cancer.[15,18,20–23]
Evidence (cisplatin in combination with other drugs):
GOG-110: The ifosfamide + cisplatin combination was superior to cisplatin alone in the secondary end point 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 FDA. However, cisplatin alone underperformed in this trial because as many as 40% of the patients had already received cisplatin up front as a radiosensitizer.[18]
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, 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 HRdeath relative to PC:[23]
1.15 (95% CI, 0.79–1.67) for vinorelbine + cisplatin (VC).
1.32 (95% CI, 0.91–1.92) for gemcitabine plus cisplatin.
1.27 (95% CI, 0.90–1.78) for CT. Trends in response rate, PFS, and OS favored CT.
Patients in the four study arms experienced different grades of neutropenia, infection, and alopecia.[23] There were no differences in health-related quality of life during treatment.[24] However, patients who received PC had more neurological side effects.
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 5-year survival rates of 32% to 62% in selected patients.[25,26] These patients are appropriate candidates for clinical trials testing drug combinations or new anticancer agents.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Oaknin A, Gladieff L, Martínez-García J, et al.: Atezolizumab plus bevacizumab and chemotherapy for metastatic, persistent, or recurrent cervical cancer (BEATcc): a randomised, open-label, phase 3 trial. Lancet 403 (10421): 31-43, 2024. [PUBMED Abstract]
Monk BJ, Colombo N, Tewari KS, et al.: First-Line Pembrolizumab + Chemotherapy Versus Placebo + Chemotherapy for Persistent, Recurrent, or Metastatic Cervical Cancer: Final Overall Survival Results of KEYNOTE-826. J Clin Oncol 41 (36): 5505-5511, 2023. [PUBMED Abstract]
Lorusso D, Xiang Y, Hasegawa K, et al.: Pembrolizumab or placebo with chemoradiotherapy followed by pembrolizumab or placebo for newly diagnosed, high-risk, locally advanced cervical cancer (ENGOT-cx11/GOG-3047/KEYNOTE-A18): a randomised, double-blind, phase 3 clinical trial. Lancet 403 (10434): 1341-1350, 2024. [PUBMED Abstract]
Chung HC, Schellens JHM, Delord JP, et al.: Pembrolizumab treatment of advanced cervical cancer: updated results from the phase 2 KEYNOTE-158 study. [Abstract] J Clin Oncol 36 (Suppl 18): A-5522, 2018.
Tewari KS, Sill MW, Long HJ, et al.: Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med 370 (8): 734-43, 2014. [PUBMED Abstract]
Vergote I, González-Martín A, Fujiwara K, et al.: Tisotumab Vedotin as Second- or Third-Line Therapy for Recurrent Cervical Cancer. N Engl J Med 391 (1): 44-55, 2024. [PUBMED Abstract]
Thomas GM, Dembo AJ, Black B, et al.: Concurrent radiation and chemotherapy for carcinoma of the cervix recurrent after radical surgery. Gynecol Oncol 27 (3): 254-63, 1987. [PUBMED Abstract]
Thigpen JT, Blessing JA, DiSaia PJ, et al.: A randomized comparison of a rapid versus prolonged (24 hr) infusion of cisplatin in therapy of squamous cell carcinoma of the uterine cervix: a Gynecologic Oncology Group study. Gynecol Oncol 32 (2): 198-202, 1989. [PUBMED Abstract]
Coleman RE, Harper PG, Gallagher C, et al.: A phase II study of ifosfamide in advanced and relapsed carcinoma of the cervix. Cancer Chemother Pharmacol 18 (3): 280-3, 1986. [PUBMED Abstract]
Sutton GP, Blessing JA, McGuire WP, et al.: Phase II trial of ifosfamide and mesna in patients with advanced or recurrent squamous carcinoma of the cervix who had never received chemotherapy: a Gynecologic Oncology Group study. Am J Obstet Gynecol 168 (3 Pt 1): 805-7, 1993. [PUBMED Abstract]
McGuire WP, Blessing JA, Moore D, et al.: Paclitaxel has moderate activity in squamous cervix cancer. A Gynecologic Oncology Group study. J Clin Oncol 14 (3): 792-5, 1996. [PUBMED Abstract]
Verschraegen CF, Levy T, Kudelka AP, et al.: Phase II study of irinotecan in prior chemotherapy-treated squamous cell carcinoma of the cervix. J Clin Oncol 15 (2): 625-31, 1997. [PUBMED Abstract]
Monk BJ, Sill MW, Burger RA, et al.: Phase II trial of bevacizumab in the treatment of persistent or recurrent squamous cell carcinoma of the cervix: a gynecologic oncology group study. J Clin Oncol 27 (7): 1069-74, 2009. [PUBMED Abstract]
Buxton EJ, Meanwell CA, Hilton C, et al.: Combination bleomycin, ifosfamide, and cisplatin chemotherapy in cervical cancer. J Natl Cancer Inst 81 (5): 359-61, 1989. [PUBMED Abstract]
Omura GA, Blessing JA, Vaccarello L, et al.: Randomized trial of cisplatin versus cisplatin plus mitolactol versus cisplatin plus ifosfamide in advanced squamous carcinoma of the cervix: a Gynecologic Oncology Group study. J Clin Oncol 15 (1): 165-71, 1997. [PUBMED Abstract]
Rose PG, Blessing JA, Gershenson DM, et al.: Paclitaxel and cisplatin as first-line therapy in recurrent or advanced squamous cell carcinoma of the cervix: a gynecologic oncology group study. J Clin Oncol 17 (9): 2676-80, 1999. [PUBMED Abstract]
Burnett AF, Roman LD, Garcia AA, et al.: A phase II study of gemcitabine and cisplatin in patients with advanced, persistent, or recurrent squamous cell carcinoma of the cervix. Gynecol Oncol 76 (1): 63-6, 2000. [PUBMED Abstract]
Long HJ, Bundy BN, Grendys EC, et al.: Randomized phase III trial of cisplatin with or without topotecan in carcinoma of the uterine cervix: a Gynecologic Oncology Group Study. J Clin Oncol 23 (21): 4626-33, 2005. [PUBMED Abstract]
Morris M, Blessing JA, Monk BJ, et al.: Phase II study of cisplatin and vinorelbine in squamous cell carcinoma of the cervix: a Gynecologic Oncology Group study. J Clin Oncol 22 (16): 3340-4, 2004. [PUBMED Abstract]
Tewari KS, Monk BJ: Gynecologic oncology group trials of chemotherapy for metastatic and recurrent cervical cancer. Curr Oncol Rep 7 (6): 419-34, 2005. [PUBMED Abstract]
Moore DH, Blessing JA, McQuellon RP, et al.: Phase III study of cisplatin with or without paclitaxel in stage IVB, recurrent, or persistent squamous cell carcinoma of the cervix: a Gynecologic Oncology Group study. J Clin Oncol 22 (15): 3113-9, 2004. [PUBMED Abstract]
Tewari KS, Monk BJ: Recent achievements and future developments in advanced and recurrent cervical cancer: trials of the Gynecologic Oncology Group. Semin Oncol 36 (2): 170-80, 2009. [PUBMED Abstract]
Monk BJ, Sill MW, McMeekin DS, et al.: Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: a Gynecologic Oncology Group study. J Clin Oncol 27 (28): 4649-55, 2009. [PUBMED Abstract]
Cella D, Huang HQ, Monk BJ, et al.: Health-related quality of life outcomes associated with four cisplatin-based doublet chemotherapy regimens for stage IVB recurrent or persistent cervical cancer: a Gynecologic Oncology Group study. Gynecol Oncol 119 (3): 531-7, 2010. [PUBMED Abstract]
Alberts DS, Kronmal R, Baker LH, et al.: Phase II randomized trial of cisplatin chemotherapy regimens in the treatment of recurrent or metastatic squamous cell cancer of the cervix: a Southwest Oncology Group Study. J Clin Oncol 5 (11): 1791-5, 1987. [PUBMED Abstract]
Tumors of the cervix. In: Morrow CP, Curtin JP: Synopsis of Gynecologic Oncology. 5th ed. Churchill Livingstone, 1998, pp 107-151.
Latest Updates to This Summary (05/13/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Editorial changes were made to this summary.
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of cervical cancer. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewer for Cervical Cancer Treatment is:
Olga T. Filippova, MD (Lenox Hill Hospital)
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Cervical Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/cervical/hp/cervical-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389493]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
Carcinogenic types of human papillomavirus (HPV) are the primary, etiologic, infectious agents that cause virtually all cases of cervical cancer. HPV type 16 (HPV-16) and HPV type 18 (HPV-18) are most often associated with invasive disease.[1,2] Because HPV can be transmitted during sexual activity, there is an association between beginning sexual activity at a younger age, as well as having a greater number of lifetime sexual partners and an increased risk of developing cervical cancer.[3] Immunosuppression is another risk factor for cervical cancer; for example, coinfection with HIV may lead to long-term persistence of viral infection (i.e., failure to clear).[4,5] Once HPV infection occurs, several additional risk factors are associated with a higher risk of the eventual development of cervical cancer. These risk factors include high parity, long-term use of oral contraceptives, and active and passive cigarette smoking.[6–8] The risk increases with longer duration and intensity of smoking. Diethylstilbestrol (DES) exposure in utero is also associated with an increased risk of developing cervical dysplasia.[9]
Factors With Adequate Evidence of an Increased Risk of Cervical Cancer
Human papillomavirus (HPV)
Based on solid evidence from observational studies, HPV infection is associated with the development of cervical cancer.
Magnitude of Effect: HPV has been implicated as the primary etiologic infectious agent causing virtually all cases of cervical cancer.
Study Design: Evidence obtained from cohort and case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Immunosuppression
Based on solid evidence, being immunosuppressed is associated with an increased risk of cervical cancer.
Study Design: Evidence obtained from cohort and case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Sexual activity at an early age and with a greater number of partners
Based on solid evidence, sexual activity at a younger age and an increasing number of sexual partners are both associated with an increased risk of HPV infection and subsequent development of cervical cancer.
Magnitude of Effect: Women who experience first sexual intercourse at age 17 years or younger or women who have had six or more lifetime sexual partners have approximately two to three times the risk of squamous cell carcinoma or adenocarcinoma of the cervix, compared with women aged 21 years or older or who have a single sexual partner.[3]
Study Design: Evidence obtained from cohort and case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
High parity
Based on solid evidence, high parity is associated with increased risk of cervical cancer in HPV-infected women.
Magnitude of Effect: Among HPV-infected women, those who have had seven or more full-term pregnancies have approximately four times the risk of squamous cell cancer compared with nulliparous women, and HPV-infected women also have two to three times the risk of women who have had one or two full-term pregnancies.[6]
Study Design: Evidence obtained from cohort or case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Long-term use of oral contraceptives
Based on solid evidence, long-term use of oral contraceptives is associated with increased risk of cervical cancer in HPV-infected women.
Magnitude of Effect: Among HPV-infected women, those who used oral contraceptives for 5 to 9 years have approximately three times the incidence of invasive cancer, and those who used them for 10 years or longer have approximately four times the risk.[7]
Study Design: Evidence obtained from cohort or case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Cigarette smoke exposure
Based on solid evidence, cigarette smoking, both active and passive, is associated with an increased risk of cervical cancer in HPV-infected women.
Magnitude of Effect: Among HPV-infected women, current and former smokers have approximately two to three times the incidence of high-grade cervical intraepithelial neoplasia or invasive cancer. Passive smoking is also associated with increased risk but to a lesser extent.
Study Design: Evidence obtained from cohort or case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Diethylstilbestrol (DES) exposure
Based on solid evidence, DES exposure is associated with an increased risk of developing clear cell adenocarcinoma of the cervix.
Magnitude of Effect: About one in 1,000 women exposed to DES in utero will develop a clear cell adenocarcinoma of the cervix.
Study Design: Evidence obtained from cohort studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Factors With Adequate Evidence of a Decreased Risk of Cervical Cancer
Sexual abstinence
Based on solid evidence, abstinence from sexual activity is associated with a near-total reduction in the risk of developing cervical cancer.
Magnitude of Effect: Sexual abstinence essentially precludes HPV transmission.
Study Design: Evidence obtained from cohort or case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Interventions With Adequate Evidence of a Decreased Risk of Cervical Cancer
Note: Based on solid evidence, screening with the Papanicolaou (Pap) test and screening with the HPV DNA test reduces cervical cancer incidence. For more information on these screening tests, see Cervical Cancer Screening.
HPV vaccination: Benefits
Based on solid evidence, vaccination against HPV-16/HPV-18 is effective in preventing HPV infection in HPV-naive individuals and is associated with a reduced incidence of cervical intraepithelial neoplasia 2 and 3. By extrapolation, these vaccines should also be associated with a reduced incidence of cervical cancer.
Magnitude of Effect: Vaccination against HPV-16 and HPV-18 reduces incident and persistent infections with efficacy of 91.6% (95% confidence interval [CI], 64.5%–98.0%) and 100% (95% CI, 45%–100%), respectively.
Study Design: Evidence obtained from randomized controlled trials (for intraepithelial precursor lesions) and cohort study analyses (for invasive cervical cancers).
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
HPV vaccination: Harms
Based on solid evidence, harms of HPV vaccines include injection-site reactions, dizziness and syncope, headache, and fever. Vaccination during pregnancy has not been associated with adverse pregnancy outcomes.[10] Allergic reactions occur rarely.
Study Design: Evidence obtained from randomized controlled trials.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Use of barrier protection during sexual intercourse: Benefits
Based on solid evidence, the use of barrier methods (e.g., condoms) during sexual intercourse is associated with a decreased risk of cervical cancer.
Magnitude of Effect: Total use of barrier protection decreases cervical cancer incidence (relative risk, 0.4; 95% CI, 0.2–0.9).
Study Design: Evidence obtained from cohort and case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
Use of barrier protection during sexual intercourse: Harms
Based on fair evidence, the use of barrier methods during sexual intercourse is associated with few serious harms. Barrier methods can break, potentially resulting in unintended pregnancy. Allergic reactions to the barrier material (e.g., natural latex) can occur.
Study Design: Evidence obtained from cohort and case-control studies.
Internal Validity: Good.
Consistency: Good.
External Validity: Good.
References
Schiffman M, Castle PE, Jeronimo J, et al.: Human papillomavirus and cervical cancer. Lancet 370 (9590): 890-907, 2007. [PUBMED Abstract]
Trottier H, Franco EL: The epidemiology of genital human papillomavirus infection. Vaccine 24 (Suppl 1): S1-15, 2006. [PUBMED Abstract]
Berrington de González A, Green J; International Collaboration of Epidemiological Studies of Cervical Cancer: Comparison of risk factors for invasive squamous cell carcinoma and adenocarcinoma of the cervix: collaborative reanalysis of individual data on 8,097 women with squamous cell carcinoma and 1,374 women with adenocarcinoma from 12 epidemiological studies. Int J Cancer 120 (4): 885-91, 2007. [PUBMED Abstract]
Abraham AG, D’Souza G, Jing Y, et al.: Invasive cervical cancer risk among HIV-infected women: a North American multicohort collaboration prospective study. J Acquir Immune Defic Syndr 62 (4): 405-13, 2013. [PUBMED Abstract]
Grulich AE, van Leeuwen MT, Falster MO, et al.: Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet 370 (9581): 59-67, 2007. [PUBMED Abstract]
Muñoz N, Franceschi S, Bosetti C, et al.: Role of parity and human papillomavirus in cervical cancer: the IARC multicentric case-control study. Lancet 359 (9312): 1093-101, 2002. [PUBMED Abstract]
Moreno V, Bosch FX, Muñoz N, et al.: Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet 359 (9312): 1085-92, 2002. [PUBMED Abstract]
Appleby P, Beral V, Berrington de González A, et al.: Carcinoma of the cervix and tobacco smoking: collaborative reanalysis of individual data on 13,541 women with carcinoma of the cervix and 23,017 women without carcinoma of the cervix from 23 epidemiological studies. Int J Cancer 118 (6): 1481-95, 2006. [PUBMED Abstract]
Hoover RN, Hyer M, Pfeiffer RM, et al.: Adverse health outcomes in women exposed in utero to diethylstilbestrol. N Engl J Med 365 (14): 1304-14, 2011. [PUBMED Abstract]
Scheller NM, Pasternak B, Mølgaard-Nielsen D, et al.: Quadrivalent HPV Vaccination and the Risk of Adverse Pregnancy Outcomes. N Engl J Med 376 (13): 1223-1233, 2017. [PUBMED Abstract]
Incidence and Mortality
An estimated 13,360 new cervical cancers and 4,320 cervical cancer deaths will occur in the United States in 2025.[1] When corrected for the prevalence of hysterectomy, the mortality rate for Black women is nearly twice the mortality rate for White women.[2] Also, approximately 1,250,000 women will be diagnosed with precancers annually by cytology using the Papanicolaou (Pap) smear. A continuum of pathological changes may be diagnosed, ranging from atypical squamous cells of undetermined significance to low-grade squamous intraepithelial lesions (LSIL) to high-grade squamous intraepithelial lesions (HSIL) to invasive cancer. The precancerous conditions LSIL and HSIL are also referred to as cervical intraepithelial neoplasia (CIN) 1, 2, and 3. Lesions can regress, persist, or progress to an invasive malignancy, with LSIL (CIN 1) more likely to regress spontaneously and HSIL (CIN 2/CIN 3) more likely to persist or progress. The average time for progression of CIN 3 to invasive cancer has been estimated to be 10 to 15 years.[3]
References
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
Beavis AL, Gravitt PE, Rositch AF: Hysterectomy-corrected cervical cancer mortality rates reveal a larger racial disparity in the United States. Cancer 123 (6): 1044-1050, 2017. [PUBMED Abstract]
Holowaty P, Miller AB, Rohan T, et al.: Natural history of dysplasia of the uterine cervix. J Natl Cancer Inst 91 (3): 252-8, 1999. [PUBMED Abstract]
Factors With Adequate Evidence of an Increased Risk of Cervical Cancer
HPV
Epidemiological studies to evaluate risk factors for the development of squamous intraepithelial lesions and cervical malignancy demonstrate conclusively a sexual mode of transmission of a carcinogen.[1] It is now widely accepted that human papillomavirus (HPV) is the primary etiologic infectious agent that causes virtually all cases of cervical cancer.[2,3] Other sexually transmitted factors, including herpes simplex virus 2 and Chlamydia trachomatis, may play a co-causative role.[4] More than 80 distinct types of HPV have been identified, approximately 30 of which infect the human genital tract. HPV type 16 (HPV-16) and HPV type 18 (HPV-18) are most often associated with invasive disease. Characterization of carcinogenic risk associated with HPV types is an important step in the process of developing a combination HPV vaccine for the prevention of cervical neoplasia. In a population-based study of HPV infection and cervical neoplasia in Costa Rica, 80% of high-grade squamous intraepithelial lesions (HSIL) and invasive lesions were associated with HPV infection by one or more of 13 cancer-associated types.[5] In this study, the risk of about 50% of HSIL and invasive cervical cancer was attributable to HPV-16. HPV-18 was associated with 15% of invasive disease but only 5% of HSIL, suggesting that HPV-18 may have a role in more aggressive cases of cervical malignancy. There may be differences in the prevalence and carcinogenic risk of individual high-risk HPV genotypes by race and geographical regions, and additional studies are ongoing.[6]
Immunosuppression
Most cases of HPV infection are resolved by the host immune system. Immunosuppression leads to persistence of viral infection with a subsequent increased risk of cervical neoplasia. Women with immunosuppression resulting from HIV infection have been studied over the past three decades of the AIDS epidemic. In one North American study, a group of 13,690 HIV-infected women were studied for a median of 5 years. The rate of invasive cervical cancer in the HIV-infected women was 26 cases per 100,000 women, and this was approximately four times greater than an HIV-uninfected control group.[7] HIV-infected women with the lowest CD4 lymphocyte counts were at the highest risk of invasive cancer. Women who are immunosuppressed resulting from organ transplantation are also at risk of invasive cervical cancer, and one meta-analysis found a twofold increased risk.[8]
Sexual Activity at an Early Age and With a Greater Number of Partners
HPV infection has been established as a necessary cause of almost all cases of cervical cancer, and the primary mode of transmission is sexual contact. This provides context for the findings that younger age at first intercourse and an increasing number of lifetime sexual partners are both associated with an increased risk of developing cervical cancer. Pooled, individual, patient-level data from 12 cohort and case-control studies demonstrated statistically significantly increased risks of developing cervical cancer in women who were aged 17 years or younger at first intercourse, compared with women who were aged 21 years or older at first intercourse (relative risk [RR] for squamous cell cancer, 2.24; 95% confidence interval [CI], 2.11–2.38 and RR for adenocarcinoma, 2.06; 95% CI, 1.83–2.33). Similar findings were observed in women who had six or more lifetime sexual partners, compared with women who had one lifetime sexual partner (RR for squamous cell cancer, 2.98; 95% CI, 2.62–3.40 and RR for adenocarcinoma, 2.64; 95% CI, 2.07–3.36).[9]
High Parity
High parity has long been recognized as a risk factor for cervical cancer, but the relation of parity to HPV infection was uncertain. A meta-analysis of 25 epidemiological studies, including 16,563 women with cervical cancer and 33,542 women without cervical cancer, showed that the number of full-term pregnancies was associated with increased risk, regardless of age at first pregnancy. This finding was also true if analyses were limited to patients with high-risk HPV infections (RR, 4.99; 95% CI, 3.49–7.13 for seven or more pregnancies vs. no pregnancies; linear trend test X2 = 30.69; P < .001).[10]
Long-Term Use of Oral Contraceptives
Long-term use of oral contraceptives has also been known to be associated with cervical cancer, but its relation to HPV infection was also uncertain. A pooled analysis of HPV-positive women from the studies described above was undertaken. Compared with women who have never used oral contraceptives, those who have used them for fewer than 5 years did not have an increased risk of cervical cancer (odds ratio [OR], 0.73; 95% CI, 0.52–1.03). The OR for women who used oral contraceptives for 5 to 9 years was 2.82 (95% CI, 1.46–5.42), and for 10 or more years, the OR was 4.03 (95% CI, 2.09–8.02).[11] A meta-analysis of 24 epidemiological studies confirmed the increased risk associated with oral contraceptives, which is proportionate to the duration of use. Risk decreases after cessation and returns to normal risk levels in 10 years.[12]
Cigarette Smoke Exposure
Cigarette smoking by women is associated with an increased risk of squamous cell carcinoma.[1,13,14] This risk increases with longer duration and intensity of smoking. The risk among smokers may be present with exposure to environmental tobacco smoke and may be as high as four times that of women who are nonsmokers who are not exposed to environmental smoking.[1] Case-control studies of women infected with HPV have examined the effect of various types and levels of tobacco exposure and found similar results.[14–16]
DES Exposure
Diethylstilbestrol (DES) is a synthetic form of estrogen that was prescribed to pregnant women in the United States between 1940 and 1971 to prevent miscarriage and premature labor. DES is associated with a substantially increased risk of developing clear cell adenocarcinoma of the vagina and cervix among the daughters of women who used the drug during pregnancy (standardized incidence ratio, 24.23; 95% CI, 8.89–52.74); the risk persists as these women age into their 40s.[17] Despite the greatly elevated risk relative to the general population, this type of cancer is still rare; about one in 1,000 daughters exposed to DES will develop a clear cell adenocarcinoma.
DES exposure in utero is also associated with an increased risk of developing cervical dysplasia. An evaluation of three cohorts, including the Diethylstilbestrol Adenosis study, the Dieckmann study, and the Women’s Health Study, with long-term follow-up of more than 4,500 women exposed in utero to DES, found that 6.9% of exposed women developed grade II or higher cervical intraepithelial neoplasia, compared with 3.4% of nonexposed women (hazard ratio, 2.28; 95% CI, 1.59–3.27).[18]
Schiffman M, Castle PE, Jeronimo J, et al.: Human papillomavirus and cervical cancer. Lancet 370 (9590): 890-907, 2007. [PUBMED Abstract]
Trottier H, Franco EL: The epidemiology of genital human papillomavirus infection. Vaccine 24 (Suppl 1): S1-15, 2006. [PUBMED Abstract]
Ault KA: Epidemiology and natural history of human papillomavirus infections in the female genital tract. Infect Dis Obstet Gynecol 2006 (Suppl): 40470, 2006. [PUBMED Abstract]
Herrero R, Hildesheim A, Bratti C, et al.: Population-based study of human papillomavirus infection and cervical neoplasia in rural Costa Rica. J Natl Cancer Inst 92 (6): 464-74, 2000. [PUBMED Abstract]
Risley C, Clarke MA, Geisinger KR, et al.: Racial differences in HPV type 16 prevalence in women with ASCUS of the uterine cervix. Cancer Cytopathol 128 (8): 528-534, 2020. [PUBMED Abstract]
Abraham AG, D’Souza G, Jing Y, et al.: Invasive cervical cancer risk among HIV-infected women: a North American multicohort collaboration prospective study. J Acquir Immune Defic Syndr 62 (4): 405-13, 2013. [PUBMED Abstract]
Grulich AE, van Leeuwen MT, Falster MO, et al.: Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet 370 (9581): 59-67, 2007. [PUBMED Abstract]
Berrington de González A, Green J; International Collaboration of Epidemiological Studies of Cervical Cancer: Comparison of risk factors for invasive squamous cell carcinoma and adenocarcinoma of the cervix: collaborative reanalysis of individual data on 8,097 women with squamous cell carcinoma and 1,374 women with adenocarcinoma from 12 epidemiological studies. Int J Cancer 120 (4): 885-91, 2007. [PUBMED Abstract]
International Collaboration of Epidemiological Studies of Cervical Cancer: Cervical carcinoma and reproductive factors: collaborative reanalysis of individual data on 16,563 women with cervical carcinoma and 33,542 women without cervical carcinoma from 25 epidemiological studies. Int J Cancer 119 (5): 1108-24, 2006. [PUBMED Abstract]
Moreno V, Bosch FX, Muñoz N, et al.: Effect of oral contraceptives on risk of cervical cancer in women with human papillomavirus infection: the IARC multicentric case-control study. Lancet 359 (9312): 1085-92, 2002. [PUBMED Abstract]
Appleby P, Beral V, Berrington de González A, et al.: Cervical cancer and hormonal contraceptives: collaborative reanalysis of individual data for 16,573 women with cervical cancer and 35,509 women without cervical cancer from 24 epidemiological studies. Lancet 370 (9599): 1609-21, 2007. [PUBMED Abstract]
Hellberg D, Nilsson S, Haley NJ, et al.: Smoking and cervical intraepithelial neoplasia: nicotine and cotinine in serum and cervical mucus in smokers and nonsmokers. Am J Obstet Gynecol 158 (4): 910-3, 1988. [PUBMED Abstract]
Brock KE, MacLennan R, Brinton LA, et al.: Smoking and infectious agents and risk of in situ cervical cancer in Sydney, Australia. Cancer Res 49 (17): 4925-8, 1989. [PUBMED Abstract]
Ho GY, Kadish AS, Burk RD, et al.: HPV 16 and cigarette smoking as risk factors for high-grade cervical intra-epithelial neoplasia. Int J Cancer 78 (3): 281-5, 1998. [PUBMED Abstract]
Plummer M, Herrero R, Franceschi S, et al.: Smoking and cervical cancer: pooled analysis of the IARC multi-centric case–control study. Cancer Causes Control 14 (9): 805-14, 2003. [PUBMED Abstract]
Verloop J, van Leeuwen FE, Helmerhorst TJ, et al.: Cancer risk in DES daughters. Cancer Causes Control 21 (7): 999-1007, 2010. [PUBMED Abstract]
Hoover RN, Hyer M, Pfeiffer RM, et al.: Adverse health outcomes in women exposed in utero to diethylstilbestrol. N Engl J Med 365 (14): 1304-14, 2011. [PUBMED Abstract]
Factors With Adequate Evidence of a Decreased Risk of Cervical Cancer
Sexual Abstinence
Nearly all cases of cervical cancer are associated with human papillomavirus (HPV) infection, which is transmitted during sexual activity. Therefore, cervical cancer is seen more frequently in women with sexual activity at an early age and with multiple partners.[1] Lifetime abstinence from sexual activity is associated with a near-total reduction in the risk of developing cervical cancer. For more information, see the HPV section.
References
IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Human papillomaviruses. IARC Monogr Eval Carcinog Risks Hum 100 (Pt B), 255-296, 2012. Available online. Last accessed January 31, 2025.
Interventions With Adequate Evidence of a Decreased Risk of Cervical Cancer
HPV Vaccination
Given the etiological role of human papillomavirus (HPV) in the pathogenesis of cervical neoplasia, vaccines to immunize individuals against HPV infection offer a primary prevention strategy for cervical cancer. A quadrivalent (HPV type 6 [HPV-6], type 11 [HPV-11], type 16 [HPV-16], and type 18 [HPV-18]) vaccine using a late protein L1 construct to induce antibody-mediated immunity was approved for use by the U.S. Food and Drug Administration in 2006; a bivalent (HPV-16, -18) vaccine was approved in 2009; and a vaccine targeting nine HPV types was approved in 2014. Vaccination during pregnancy has not been associated with adverse pregnancy outcomes.[1]
Persistent infection with oncogenic types of HPV, such as HPV-16 and HPV-18, is associated with the development of cervical cancer.[2] A vaccine to prevent HPV infection with oncogenic-type viruses has the potential to reduce the incidence of cervical cancer. A vaccine against HPV-16 using empty-viral capsids called virus-like particles (VLP) was developed and tested for efficacy in preventing persistent infection with HPV-16.
A multicenter, double-blind, placebo-controlled trial enrolled 2,391 women aged 16 to 23 years and randomly assigned them to receive either 40 µg of HPV-16 L1 VLP vaccine or placebo on day 1, at 2 months, and at 6 months. Papanicolaou (Pap) tests and genital samples for HPV-16 DNA were obtained on day 1, at 7 months, and every 6 months for 48 months. Colposcopy and cervical biopsies were obtained when clinically indicated at study exit. Serum HPV-16 antibody titers were obtained at study entry, at 7 months, and then every 6 months. A total of 1,505 women (755 receiving vaccine and 750 receiving placebo) completed all three vaccinations and had follow-up after month 7. After immunization, HPV titers peaked at month 7, declined through month 18, and then stabilized in months 30 through 48. There were no cases of cervical intraepithelial neoplasia (CIN) in the vaccine-treated women, but there were 12 cases in the placebo group (six CIN 2 and six CIN 3). HPV-16 infection that persisted for at least 4 months was seen in seven vaccine-treated women, compared with 111 placebo-treated women.[3]
An international, double-blind, placebo-controlled trial of a bivalent HPV-16/HPV-18 VLP vaccine was performed in 1,113 women aged 15 to 25 years with normal cervical cytology who were seronegative for HPV-16, HPV-18, and 12 other oncogenic HPV types at enrollment. Women received either vaccine or placebo at 0, 1, and 6 months and were assessed by cervical cytology and self-obtained cervicovaginal samples for at least 18 months. A masked treatment-allocation follow-up study was performed for an additional 3 years, for a combined analysis of up to 6.4 years of follow-up. The 12-month persistent infection rate of HPV-16 or HPV-18 in an according-to-protocol cohort (i.e., women who received all three doses of vaccine or placebo on the correct schedule) was 0 of 401 women in the vaccine arm, compared with 20 of 372 women in the placebo arm, with a vaccine efficacy rate of 100% (95% confidence interval [CI], 81.8%–100%). Diagnoses of CIN 2 or higher in a total vaccinated cohort (i.e., women who received at least one dose of vaccine or placebo) were 0 of 481 women in the vaccine arm compared with 9 of 470 women in the placebo arm, with a vaccine efficacy of 100% (95% CI, 51.3%–100%). Adverse events were similar in vaccinated and placebo-treated women. Neither analysis was intention-to-treat (ITT), making it difficult to know what the true vaccine efficacy for either virological or cytohistological end points would be in the routine clinical setting. Furthermore, cytohistological outcomes were reported only as composite end points (CIN 2+), making it impossible to distinguish the vaccine’s efficacy against invasive cervical cancer alone and potentially inflating the observed efficacy by including lesions with a relatively high probability (approximately 50% for CIN 2 [4]) of spontaneous regression.[5] A register-based observational study in England reported the impact of a national bivalent vaccination program on cervical cancer and CIN 3.[6] Routine vaccinations were offered to girls aged 12 to 13 years with a catch-up program for those aged 14 to 18 years. Data from 13.7 million years of follow-up in women aged 20 years to younger than 30 years showed a substantial reduction in cervical cancer and CIN 3 incidence after a national HPV program was introduced. This was especially true in individuals who were offered the vaccine between ages 12 and 13 years (see Table 1).
Table 1. Estimated Relative Reduction in Cervical Cancer or CIN 3 by Age When Vaccine Was Offered Compared With the Reference Unvaccinated Cohort
Estimated Relative Reduction (95% CI)
Age Vaccine Was Offered
Cervical Cancer
CIN 3
CI = confidence interval; CIN = cervical intraepithelial neoplasia.
16–18 y
34% (25–41)
39% (36–41)
14–16 y
62% (52–71)
75% (72–77)
12–13 y
87% (72–94)
97% (96–78)
A quadrivalent vaccine (HPV types-6, -11, -16, and -18) was evaluated in a multinational, double-blind, randomized controlled trial of 17,622 women aged 15 to 26 years (FUTURE I and II).[7] Women received either the HPV vaccine or placebo at 0, 2, and 6 months; participants were assessed by clinical exam, Pap test, and HPV DNA testing for 4 or more years. Two analyses were reported. One group was considered to be HPV naive: negative to 14 HPV types. The second group was an ITT analysis, which approximates a sexually active population. The composite end point for cervical disease included the incidence of HPV-16/HPV-18–related, CIN 2, CIN 3, adenocarcinoma in situ, or invasive carcinoma. Outcomes were reported as follows:
Table 2. Vaccine Efficacy of the Quadrivalent HPV Vaccine
Population
Point Estimate and 95% CI
CI = confidence interval; CIN = cervical intraepithelial neoplasia; HPV = human papillomavirus; ITT = intention-to-treat.
HPV-naive population for HPV-CIN 3
100% (90.5%–100%) for lesions associated with HPV-6, -11, -16, or -18
ITT CIN 3
45.3% (29.8%–57.6%) for lesions associated with HPV-6, -11, -16, or -18
This study also demonstrated decreased rates of abnormal Pap tests and subsequent diagnostic procedures. No cases of invasive cervical cancer were identified during the trial.
A 9-valent HPV (9vHPV) vaccine was studied in another international randomized trial, which included 14,215 women. This new vaccine, 9vHPV, includes the four HPV types in the quadrivalent vaccine, qHPV (6, 11, 16, 18) and also 5 more oncogenic types (31, 33, 45, 52, 58). Sexually active women aged 16 to 26 years with fewer than five lifetime sexual partners received three intramuscular injections (day 1, month 2, and month 6) of either the qHPV vaccine or the 9vHPV vaccine. Women were evaluated every 6 months for up to 5 years. The rate of high-grade cervical, vulvar, or vaginal disease was the same in both groups (14.0 per 1,000 person-years) because of preexisting HPV infection, but the rate of disease related to HPV-31, -35, -45, -52, and -58 was lower in the 9vHPV vaccine group (0.1 vs. 1.6 per 1,000 person-years). Injection-site reactions were more common in the 9vHPV group.[8] Although not addressed in this study, the benefit of HPV vaccination is optimal in younger females before the onset of sexual activity.
All forms of the HPV vaccine are currently recommended by the Centers for Disease Control and Prevention (CDC) in the United States as a two-dose schedule at least 6 months apart for adolescents younger than 15 years. The current CDC recommendation for older individuals is to receive the original three-dose series. Recently, given issues of cost and adherence, there has been published data investigating whether similar vaccine efficacy could be obtainable using a reduced-dose schedule. A post hoc combined analysis of two phase III randomized controlled trials of the bivalent HPV vaccine (the Costa Rica Vaccine Trial and the PApilloma TRIal against Cancer In young Adults [PATRICIA] Trial) found that among women who were not HPV positive at enrollment for the specific virus type being studied, vaccine efficacy against either one-time incident detection of HPV 16/18 or incident infection that persisted at least 6 months was not statistically significantly different for those who received all three, two, or only one of the scheduled HPV vaccine doses (resulting from nonadherence or other factors) for up to 4 years of follow-up. Vaccine efficacy rates for persistent HPV 16/18 infection ranged from 89.1% (95% CI, 86.8%–91.0%) for three doses, to 89.7% (95% CI, 73.3%–99.8%) for two doses, to 96.6% (95% CI, 81.7%–99.8%) for one dose. To date, there are no randomized controlled trials that directly assess this clinical question.[9] A recent international study compared a two-dose schedule with a three-dose schedule in adolescents younger than 15 years who received the 9vHPV vaccine. The antibody response was noninferior in the two-dose schedule, leading to the current recommendation that two doses are sufficient for this age group.[10] Long-term follow-up data from the Costa Rica Vaccine Trial suggested that all HPV-vaccinated women aged 18 to 25 years at the time of initial vaccination remained HPV-16/HPV-18 seropositive more than a decade after initial vaccination, regardless of the number of doses received. The antibody levels were lower in the women who received only one dose than in the women who received two or three doses of the bivalent vaccine, but the levels remained higher than estimated levels achieved via natural immunity. The long-term vaccine efficacy rates against prevalent HPV-16 or HPV-18 infection were 80.2% (95% CI, 70.7%–87.0%) among women who received three doses, 83.8% (95% CI, 19.5%–99.2%) among those who received two doses, and 82.1% (95% CI, 40.2%–97.0%) among those who received one dose.[11] Additionally, there was prolonged efficacy against CIN 2 and CIN 3 after 7 to 11 years of follow-up.[12] The women in this long-term follow-up study were not randomly assigned to one, two, or three doses, and the number of women who received only one dose is low. However, the promising findings of the long-term stability of HPV-antibody levels and vaccine efficacy in women who were older than the recommended age at the time of initial vaccination has influenced the design of a currently ongoing trial, which will answer the question of the efficacy of a single dose more definitively. The ESCUDDO study (NCT03180034) is a trial enrolling adolescent girls who will be randomly assigned to either one dose or two doses of the bivalent or nonavalent vaccines. A concurrent population survey of comparable, unvaccinated, age-matched females in the same region will be used for comparison. Results are anticipated in 2025.
On the basis of their mechanism of action, L1/2 HPV vaccines do not appear to impact preexisting infections. The FUTURE II trial demonstrated a markedly lower vaccine efficacy rate in the total randomized study population, which included individuals who were positive for HPV at baseline, compared with the per-protocol population (44% for lesions associated with HPV-16 or HPV-18, and 17% for lesions associated with any HPV type vs. 98%; see Table 2 above).[7] Additionally, an intermediate analysis of a randomized controlled trial primarily evaluating the efficacy of the HPV-16/18 vaccine in preventing infection found no effect on viral clearance rates in women aged 18 to 25 years who were positive at the time of study enrollment.[13]
The type-specific vaccines, if successful in preventing invasive cancer, will offer protection for only a subset of cases, the proportion of which will vary worldwide.[14] Using data from a multicenter case-control study conducted in 25 countries, it was estimated that a vaccine containing the seven most common HPV types could prevent 87% of cervical cancers worldwide. A vaccine with the two most common strains, HPV-16 and HPV-18, would prevent 71% of cervical cancers worldwide.[14]
There is growing evidence of population-level impacts and herd immunity with HPV vaccination. There are data that explore the impact of national HPV vaccination programs and report on vaccine effectiveness. These data come from studies conducted in different countries throughout the world including England, Denmark, Australia, Costa Rica, and the United States. In England, 15,459 residual genital specimens from women aged 16 to 24 years, collected for Chlamydia trachomatis screening between 2010 and 2016, were utilized for national HPV surveillance.[15] In this study, vaccine effectiveness for HPV-16/HPV-18 was 82% (95% CI, 60.6%–91.8%) for women who were vaccinated before age 15 years. Within the younger age groups, the prevalence of HPV-16/HPV-18 significantly decreased within the postvaccination period between 2010 and 2011 to 2016 from 8.2% to 1.6% in 16 to 18 year olds and from 14.0% to 1.6% in 19 to 21 year olds (compared with 17.6% and 16.9% in the prevaccination era).[15] Data from a Danish nationwide cohort study reported the dose-related effectiveness of the quadrivalent HPV vaccine.[16] In this cohort of 590,083 women aged 17 to 25 years, 215,309 (36%) women were vaccinated at age 16 years or younger, and 40,742 (19%) women received less than three doses. Cervical cancer screening rates were similar in the vaccinated and unvaccinated cohorts. In the overall cohort, there were 5,561 cases of CIN 3+ during the follow-up period. Only 5% of cases were in vaccinated women. The incidence rate was 355 cases per 100,000 person-years in unvaccinated women compared with 41 cases per 100,000 person-years in vaccinated women. The incidence rate was independent of the number of vaccine doses administered (incidence rates 40, 41, 40 cases per 100,000 person-years for 1, 2 and 3 doses, respectively).[16]
A study of cervical HPV DNA among 202 Australian women aged 18 to 24 years who were sampled between 2005 and 2007, before implementation of a national quadrivalent prophylactic HPV vaccine program, compared the results with a matched group of 1,058 women who were sampled in the postvaccination era (2010–2012). This study found an adjusted prevalence ratio (PR) among fully vaccinated women of 0.07 (95% CI, 0.04–0.14; P < .0001) for vaccine-related HPV types and a smaller but statistically significant magnitude of protection of 0.65 (95% CI, 0.43–0.96; P < .03) among unvaccinated women, suggesting herd immunity (protection of unvaccinated individuals).[17] These data strengthen previous results that suggest herd immunity in this population, manifested as a reduction in genital warts among heterosexual men, a group that includes sexual partners of vaccinated women.[18] Data also suggest cross protection against carcinogenic types that are not directly targeted by the quadrivalent vaccine but are included in the new nonvalent HPV vaccine.[17] Pooled data from the Costa Rica Vaccine Trial and PATRICIA Study showed that the AS04-adjuvanted HPV-16/HPV-18 vaccine provides additional cross protection beyond established protected types (e.g., 34/35/39/40/42/43/44/51/52/53/54/56/58/59/66/68/73/70/74; efficacy 9/9%; 95% CI, 1.7%–1.4%). This may partially explain the high efficacy of the AS04-adjuvanted HPV-16/HPV-18 vaccine against CIN 3+ (87.8%; 95% CI, 71.1%–95.7%).[19] A meta-analysis that included data from 14 high-income countries with cumulated data from more than 60 million individuals over 8 years reported an 83% decrease in prevalence of HPV-16 and HPV-18 (RR, 0.17; 95% CI, 0.11–0.25) among girls aged 13 to 19 years. There was also evidence of benefit to a more proximal cancer end point. After 5 to 9 years of HPV vaccination, decreased risk of CIN 2+ was also identified among screened girls aged 15 to 19 years (RR, 0.49; 95% CI, 0.42–0.58), and among women aged 20 to 24 years (RR, 0.69; 95% CI, 0.57–0.84) in comparison to an increase seen among screened and mostly unvaccinated women aged 25 to 29 years (RR, 1.19; 95% CI, 1.06–1.32) and aged 30 to 39 years (RR, 1.23; 95% CI, 1.13–1.34).[20]
Data from the National Health and Nutrition Examination Survey (NHANES) from 2003 to 2018 demonstrated an increasing impact of the HPV vaccination program and herd protection in the United States.[21] Overall, the data demonstrated an increase in HPV vaccination coverage among sexually experienced females and males. Importantly, vaccination before age 15 years also increased from 2011 to 2014 and from 2015 to 2018 in both females (27.2%, 48.6%, respectively) and males (18.6%, 48.7%, respectively). From 2015 to 2018, the 4-valent HPV (4vHPV)-type prevalence among sexually experience females aged 14 to 24 years was 85% overall, 90% in vaccinated females, and 74% in unvaccinated females. Estimates of the vaccine’s effectiveness and its impact among vaccinated females were similar from 2007 to 2010 (64% and 61%, respectively) and from 2011 to 2014 (84% and 89%, respectively). However, in 2015 to 2018, these statistics diverged (60% and 90%, respectively). This indicates that as herd protection increases and prevalence among unvaccinated individuals decreases, vaccine effectiveness can be difficult to estimate (1-prevalence ratio between vaccinated and unvaccinated individuals x 100). In 2013 to 2016, the prevalence of 4vHPV types was 1.8% in sexually experienced males who were vaccinated and 3.5% in sexually experienced males who were unvaccinated (PR, 0.49; 95% CI, 0.11–2.20), resulting in an estimated vaccine effectiveness of 51%. Significant declines were not observed in non–4vHPV-type prevalence for females or males. Although notable limitations of this survey study included self-report of HPV vaccine and dose, small sample sizes, and estimates of the impact and effectiveness based on history of at least one vaccine dose, this nationally representative data reflects an increasing impact of the U.S. vaccination program and herd protection.
Association of HPV vaccination with reduced incidence of invasive cervical cancer
In a nationwide population-based cohort study of the impact of the national vaccination program in Sweden using quadrivalent vaccine, more than 1.67 million women aged 10 to 30 years with no previous history of HPV vaccination were followed through the national registry using individual person linkage.[22] The cumulative risk of cervical cancer by age 30 years was 47 cases per 100,000 in vaccinated women, compared with 94 cases per 100,000 in unvaccinated women (incidence rate ratio [IRR], 0.51; 95% CI, 0.32–0.82, adjusting only for age at follow-up). After adjusting for all collected potential confounding factors, the IRR for women vaccinated before age 17 years was 0.12 (95% CI, 0.00–0.34).
Use of Barrier Protection During Sexual Intercourse
Barrier methods of contraception are associated with a reduced incidence of squamous intraepithelial lesions (SIL) presumptively secondary to protection from sexually transmitted disease.[23,24] The effectiveness of condom use for the prevention of HPV infections has been evaluated in a prospective study of women aged 18 to 22 years who were virgins.[25] The number of vulvovaginal HPV infections was reduced with consistent condom use, and the HPV infection rate was 37.8 infections per 100 patient-years among women whose partners used condoms 100% of the time in the 8 months before testing, compared with 89.3 infections per 100 patient-years among women whose partners used condoms less than 5% of the time (P trend = .005). No cervical SIL were detected among women reporting 100% condom use by their partner.[25]
References
Scheller NM, Pasternak B, Mølgaard-Nielsen D, et al.: Quadrivalent HPV Vaccination and the Risk of Adverse Pregnancy Outcomes. N Engl J Med 376 (13): 1223-1233, 2017. [PUBMED Abstract]
Wallin KL, Wiklund F, Angström T, et al.: Type-specific persistence of human papillomavirus DNA before the development of invasive cervical cancer. N Engl J Med 341 (22): 1633-8, 1999. [PUBMED Abstract]
Mao C, Koutsky LA, Ault KA, et al.: Efficacy of human papillomavirus-16 vaccine to prevent cervical intraepithelial neoplasia: a randomized controlled trial. Obstet Gynecol 107 (1): 18-27, 2006. [PUBMED Abstract]
Castle PE, Schiffman M, Wheeler CM, et al.: Evidence for frequent regression of cervical intraepithelial neoplasia-grade 2. Obstet Gynecol 113 (1): 18-25, 2009. [PUBMED Abstract]
Romanowski B, de Borba PC, Naud PS, et al.: Sustained efficacy and immunogenicity of the human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine: analysis of a randomised placebo-controlled trial up to 6.4 years. Lancet 374 (9706): 1975-85, 2009. [PUBMED Abstract]
Falcaro M, Castañon A, Ndlela B, et al.: The effects of the national HPV vaccination programme in England, UK, on cervical cancer and grade 3 cervical intraepithelial neoplasia incidence: a register-based observational study. Lancet 398 (10316): 2084-2092, 2021. [PUBMED Abstract]
FUTURE II Study Group: Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med 356 (19): 1915-27, 2007. [PUBMED Abstract]
Joura EA, Giuliano AR, Iversen OE, et al.: A 9-valent HPV vaccine against infection and intraepithelial neoplasia in women. N Engl J Med 372 (8): 711-23, 2015. [PUBMED Abstract]
Kreimer AR, Struyf F, Del Rosario-Raymundo MR, et al.: Efficacy of fewer than three doses of an HPV-16/18 AS04-adjuvanted vaccine: combined analysis of data from the Costa Rica Vaccine and PATRICIA trials. Lancet Oncol 16 (7): 775-86, 2015. [PUBMED Abstract]
Iversen OE, Miranda MJ, Ulied A, et al.: Immunogenicity of the 9-Valent HPV Vaccine Using 2-Dose Regimens in Girls and Boys vs a 3-Dose Regimen in Women. JAMA 316 (22): 2411-2421, 2016. [PUBMED Abstract]
Kreimer AR, Sampson JN, Porras C, et al.: Evaluation of Durability of a Single Dose of the Bivalent HPV Vaccine: The CVT Trial. J Natl Cancer Inst 112 (10): 1038-1046, 2020. [PUBMED Abstract]
Porras C, Tsang SH, Herrero R, et al.: Efficacy of the bivalent HPV vaccine against HPV 16/18-associated precancer: long-term follow-up results from the Costa Rica Vaccine Trial. Lancet Oncol 21 (12): 1643-1652, 2020. [PUBMED Abstract]
Hildesheim A, Herrero R, Wacholder S, et al.: Effect of human papillomavirus 16/18 L1 viruslike particle vaccine among young women with preexisting infection: a randomized trial. JAMA 298 (7): 743-53, 2007. [PUBMED Abstract]
Muñoz N, Bosch FX, Castellsagué X, et al.: Against which human papillomavirus types shall we vaccinate and screen? The international perspective. Int J Cancer 111 (2): 278-85, 2004. [PUBMED Abstract]
Mesher D, Panwar K, Thomas SL, et al.: The Impact of the National HPV Vaccination Program in England Using the Bivalent HPV Vaccine: Surveillance of Type-Specific HPV in Young Females, 2010-2016. J Infect Dis 218 (6): 911-921, 2018. [PUBMED Abstract]
Verdoodt F, Dehlendorff C, Kjaer SK: Dose-related Effectiveness of Quadrivalent Human Papillomavirus Vaccine Against Cervical Intraepithelial Neoplasia: A Danish Nationwide Cohort Study. Clin Infect Dis 70 (4): 608-614, 2020. [PUBMED Abstract]
Tabrizi SN, Brotherton JM, Kaldor JM, et al.: Assessment of herd immunity and cross-protection after a human papillomavirus vaccination programme in Australia: a repeat cross-sectional study. Lancet Infect Dis 14 (10): 958-66, 2014. [PUBMED Abstract]
Donovan B, Franklin N, Guy R, et al.: Quadrivalent human papillomavirus vaccination and trends in genital warts in Australia: analysis of national sentinel surveillance data. Lancet Infect Dis 11 (1): 39-44, 2011. [PUBMED Abstract]
Tota JE, Struyf F, Hildesheim A, et al.: Efficacy of AS04-Adjuvanted Vaccine Against Human Papillomavirus (HPV) Types 16 and 18 in Clearing Incident HPV Infections: Pooled Analysis of Data From the Costa Rica Vaccine Trial and the PATRICIA Study. J Infect Dis 223 (9): 1576-1581, 2021. [PUBMED Abstract]
Drolet M, Bénard É, Pérez N, et al.: Population-level impact and herd effects following the introduction of human papillomavirus vaccination programmes: updated systematic review and meta-analysis. Lancet 394 (10197): 497-509, 2019. [PUBMED Abstract]
Rosenblum HG, Lewis RM, Gargano JW, et al.: Human Papillomavirus Vaccine Impact and Effectiveness Through 12 Years After Vaccine Introduction in the United States, 2003 to 2018. Ann Intern Med 175 (7): 918-926, 2022. [PUBMED Abstract]
Lei J, Ploner A, Elfström KM, et al.: HPV Vaccination and the Risk of Invasive Cervical Cancer. N Engl J Med 383 (14): 1340-1348, 2020. [PUBMED Abstract]
Parazzini F, Negri E, La Vecchia C, et al.: Barrier methods of contraception and the risk of cervical neoplasia. Contraception 40 (5): 519-30, 1989. [PUBMED Abstract]
Hildesheim A, Brinton LA, Mallin K, et al.: Barrier and spermicidal contraceptive methods and risk of invasive cervical cancer. Epidemiology 1 (4): 266-72, 1990. [PUBMED Abstract]
Winer RL, Hughes JP, Feng Q, et al.: Condom use and the risk of genital human papillomavirus infection in young women. N Engl J Med 354 (25): 2645-54, 2006. [PUBMED Abstract]
Latest Updates to This Summary (04/18/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 1).
This summary is written and maintained by the PDQ Screening and Prevention Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about cervical cancer prevention. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Screening and Prevention Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Screening and Prevention Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Screening and Prevention Editorial Board. PDQ Cervical Cancer Prevention. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/cervical/hp/cervical-prevention-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389433]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
Carcinomas of the vagina are uncommon tumors comprising about 2% of the cancers that arise in the female genital system.[1] Squamous cell carcinoma (SCC) accounts for approximately 80% to 90% of vaginal cancer cases and adenocarcinoma accounts for 5% to 10% of vaginal cancer cases.[1]
Rarely, melanomas (often nonpigmented), sarcomas, small-cell carcinomas, lymphomas, or carcinoid tumors have been described as primary vaginal cancers. The natural history, prognosis, and treatment of other primary vaginal cancers are different and are not covered in this summary.
Distant hematogenous metastases occur most commonly in the lungs, and, less frequently, in the liver, bone, or other sites.[1]
The American Joint Committee on Cancer staging system classifies tumors in the vagina that involve the cervix of women with an intact uterus as cervical cancers.[2] Therefore, tumors that originated in the apical vagina but extend to the cervix are classified as cervical cancers. For more information, see Cervical Cancer Treatment.
Incidence and Mortality
Estimated new cases and deaths from vaginal and other female genital cancer in the United States in 2025:[3]
Increasing age is the most important risk factor for most cancers. Other risk factors for vaginal cancer include:
Human papillomavirus (HPV) infection. SCC of the vagina is associated with a high rate of infection with oncogenic strains of HPV. SCC of the vagina and SCC of the cervix have many common risk factors.[4–6] HPV infection has also been described in a case of vaginal adenocarcinoma.[6] For more information, see Cervical Cancer Treatment.
Diethylstilbestrol (DES) exposure in utero. A rare form of adenocarcinoma, known as clear cell carcinoma, occurs in association with in utero exposure to DES, with a peak incidence before age 30 years. This association was first reported in 1971.[7] The incidence of this disease, which is highest for those exposed during the first trimester, peaked in the mid-1970s, reflecting the use of DES in the 1950s. It is extremely rare now.[1] However, women with a known history of in utero DES exposure should be carefully monitored for possible presence of this tumor. (This association was mainly applicable to vaginal cancers diagnosed in younger women since adenocarcinomas that are not associated with DES exposure occur primarily during postmenopausal years.)
Vaginal adenosis is most commonly found in young women who had in utero exposure to DES and may coexist with a clear cell adenocarcinoma, although it rarely progresses to adenocarcinoma. Adenosis is replaced by squamous metaplasia, which occurs naturally, and requires follow-up but not removal.
History of hysterectomy. Women who have had a hysterectomy for benign, premalignant, or malignant disease are at risk of vaginal carcinomas.[8] In a retrospective series of 100 women studied over 30 years, 50% had undergone hysterectomy before the diagnosis of vaginal cancer.[8] In the posthysterectomy group, 31 of 50 women (62%) developed cancers limited to the upper third of the vagina. In women who had not previously undergone hysterectomy, upper vaginal lesions were found in 17 of 50 women (34%).
Clinical Features
Although early vaginal cancer may not cause noticeable signs or symptoms, possible signs and symptoms of vaginal cancer include:
Metrorrhagia.
Dyspareunia.
Pelvic pain.
Vaginal mass.
Dysuria.
Constipation.
Diagnostic Evaluation
The following procedures may be used to diagnose vaginal cancer:
History and physical examination.
Pelvic examination.
Cervical cytology (Pap smear).
HPV testing.
Colposcopy.
Biopsy. If the cervix is intact, biopsies are mandatory to rule out a primary carcinoma of the cervix. Carcinoma of the vulva should also be ruled out.
Prognostic Factors
Prognosis depends primarily on the stage of disease, but survival is reduced among women with the following features:
Age older than 60 years.
Symptomatic at the time of diagnosis.
Lesions of the middle and lower third of the vagina.
Poorly differentiated tumors.
In addition, the length of vaginal wall involvement has been found to be associated with survival and stage of disease in patients with vaginal SCC.
Follow-Up After Treatment
Similar to other gynecologic malignancies, the evidence to support surveillance after initial management of vaginal cancer is weak because of a lack of randomized or prospective clinical studies.[9] There is no reliable evidence that routine cytological or imaging procedures in patients improves health outcomes beyond what is achieved by careful physical examination and assessment of new symptoms. Therefore, outside the investigational setting, imaging procedures may be reserved for patients in whom physical examination or symptoms raise clinical suspicion of a recurrence or progression.
References
Eifel PJ, Klopp AH, Berek JS, et al.: Cancer of the cervix, vagina, and vulva. In: DeVita VT Jr, Lawrence TS, Rosenberg SA, et al., eds.: DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 11th ed. Wolters Kluwer, 2019, pp 1171-1210.
American Cancer Society: Cancer Facts and Figures 2025. American Cancer Society, 2025. Available online. Last accessed January 16, 2025.
Daling JR, Madeleine MM, Schwartz SM, et al.: A population-based study of squamous cell vaginal cancer: HPV and cofactors. Gynecol Oncol 84 (2): 263-70, 2002. [PUBMED Abstract]
Parkin DM: The global health burden of infection-associated cancers in the year 2002. Int J Cancer 118 (12): 3030-44, 2006. [PUBMED Abstract]
Ikenberg H, Runge M, Göppinger A, et al.: Human papillomavirus DNA in invasive carcinoma of the vagina. Obstet Gynecol 76 (3 Pt 1): 432-8, 1990. [PUBMED Abstract]
Herbst AL, Ulfelder H, Poskanzer DC: Adenocarcinoma of the vagina. Association of maternal stilbestrol therapy with tumor appearance in young women. N Engl J Med 284 (15): 878-81, 1971. [PUBMED Abstract]
Stock RG, Chen AS, Seski J: A 30-year experience in the management of primary carcinoma of the vagina: analysis of prognostic factors and treatment modalities. Gynecol Oncol 56 (1): 45-52, 1995. [PUBMED Abstract]
Salani R, Backes FJ, Fung MF, et al.: Posttreatment surveillance and diagnosis of recurrence in women with gynecologic malignancies: Society of Gynecologic Oncologists recommendations. Am J Obstet Gynecol 204 (6): 466-78, 2011. [PUBMED Abstract]
Stage Information for Vaginal Cancer
FIGO Staging System
The Fédération Internationale de Gynécologie et d’Obstétrique (FIGO) and the American Joint Committee on Cancer (AJCC) have designated staging to define vaginal cancer. The FIGO system is the most commonly used staging system for vaginal cancer.[1–3]
Table 1. Carcinoma of the Vaginaa
FIGO Nomenclature
FIGO = Fédération Internationale de Gynécologie et d’Obstétrique.
aAdapted from FIGO Committee on Gynecologic Oncology.[1,2]
Stage I
The carcinoma is limited to the vaginal wall.
Stage II
The carcinoma has involved the subvaginal tissue but has not extended to the pelvic wall.
Stage III
The carcinoma has extended to the pelvic wall.
Stage IV
The carcinoma has extended beyond the true pelvis or has involved the mucosa of the bladder or rectum; bullous edemas as such does not permit a case to be allotted to stage IV.
IVa – Tumor invades bladder and/or rectal mucosa and/or direct extension beyond the true pelvis.
IVb – Spread to distant organs.
In addition, the FIGO staging system incorporates a modified World Health Organization prognostic scoring system. The scores from the eight risk factors are summed and incorporated into the FIGO stage, separated by a colon (e.g., stage II:4, stage IV:9, etc.). Unfortunately, a variety of risk-scoring systems have been published, making comparisons of results difficult.
References
FIGO Committee on Gynecologic Oncology: Current FIGO staging for cancer of the vagina, fallopian tube, ovary, and gestational trophoblastic neoplasia. Int J Gynaecol Obstet 105 (1): 3-4, 2009. [PUBMED Abstract]
Adams TS, Rogers LJ, Cuello MA: Cancer of the vagina: 2021 update. Int J Gynaecol Obstet 155 (Suppl 1): 19-27, 2021. [PUBMED Abstract]
Because vaginal cancer is rare, studies are limited to retrospective case series, usually from single-referral institutions.[Level of evidence C2] During the long span of time covered by these case series, available staging tests and radiation techniques often changed, including the shift to high-energy accelerators and conformal and intensity-modulated radiation therapy.[1,2] Comparing treatment approaches is further complicated by the frequent failure of investigators to provide precise staging criteria (particularly for stage I vs. stage II disease) or criteria for the choice of treatment modality. This has led to a broad range of reported disease control and survival rates for any given stage and treatment modality.[3]
The following factors should be considered when planning treatment for vaginal cancer:
Stage and size of the lesion.
Ability to retain a functional vagina.
Presence or absence of the uterus.
Whether the patient has received previous pelvic radiation therapy.
Whether the lymphatics drain to pelvic or inguinal nodes or both, depending on tumor location.
Proximity of the vagina to the bladder or rectum. This limits surgical treatment options and increases short-term and long-term surgical complications and functional deficits.
Proximity to radiosensitive organs or organs that preclude radical resection without unacceptable functional deficits (e.g., bladder, rectum, urethra).
Radiation-induced damage to nearby organs may include:[1,2]
Rectovaginal fistulas.
Vesicovaginal fistulas.
Rectal or vaginal strictures.
Cystitis.
Proctitis.
Premature menopause from ovarian damage.
Soft tissue or bone necrosis.
Management of the extremely rare vaginal clear cell carcinoma is similar to the management of squamous cell carcinoma. However, techniques that preserve vaginal and ovarian function should be strongly considered during treatment planning, given the young age of the patients at diagnosis.[4]
For patients with early-stage vaginal carcinoma, radiation therapy, surgery, or a combination of these treatments are standard. Data from randomized trials are lacking, and the choice of therapy is generally determined by institutional experience and the factors listed above.[3]
For patients with stages III and IVa disease, radiation therapy is standard and includes external-beam radiation therapy (EBRT), alone or with brachytherapy. Regional lymph nodes are included in the radiation portal. When used alone, EBRT involves a tumor dose of 65 Gy to 70 Gy, using shrinking fields, delivered within 6 to 7 weeks. Intracavitary brachytherapy provides insufficient dose penetration for locally advanced tumors, so interstitial brachytherapy is used if brachytherapy is given.[3,5]
For patients with stage IVb or recurrent disease that cannot be managed with local treatments, current therapy is inadequate. No established anticancer drugs have demonstrated proven clinical benefit, although patients are often treated with regimens used to treat cervical cancer. Concurrent chemotherapy, using fluorouracil or cisplatin-based therapy, and radiation are sometimes advocated, based solely on extrapolation from cervical cancer management strategies.[6–8] Evidence is limited to small case series and the incremental impact on survival and local control is not well defined.[Level of evidence C3]
Local control is a problem with bulky tumors. Some investigators have also used concurrent chemotherapy with agents such as cisplatin, bleomycin, mitomycin, floxuridine, and vincristine without improved outcomes.[1] It is an extrapolation from treatment approaches used in cervical cancer, based on shared etiologic and risk factors.
Because vaginal cancer is rare, these patients are candidates for clinical trials of anticancer drugs and/or radiosensitizers to attempt to improve survival or local control. Discussion of clinical trials should be considered with eligible patients. Information about ongoing clinical trials is available from the NCI website.
Fluorouracil Dosing
The DPYD gene encodes an enzyme that catabolizes pyrimidines and fluoropyrimidines, like capecitabine and fluorouracil. An estimated 1% to 2% of the population has germline pathogenic variants in DPYD, which lead to reduced DPD protein function and an accumulation of pyrimidines and fluoropyrimidines in the body.[9,10] Patients with the DPYD*2A variant who receive fluoropyrimidines may experience severe, life-threatening toxicities that are sometimes fatal. Many other DPYD variants have been identified, with a range of clinical effects.[9–11] Fluoropyrimidine avoidance or a dose reduction of 50% may be recommended based on the patient’s DPYD genotype and number of functioning DPYD alleles.[12–14] DPYD genetic testing costs less than $200, but insurance coverage varies due to a lack of national guidelines.[15] In addition, testing may delay therapy by 2 weeks, which would not be advisable in urgent situations. This controversial issue requires further evaluation.[16]
References
Frank SJ, Jhingran A, Levenback C, et al.: Definitive radiation therapy for squamous cell carcinoma of the vagina. Int J Radiat Oncol Biol Phys 62 (1): 138-47, 2005. [PUBMED Abstract]
Tran PT, Su Z, Lee P, et al.: Prognostic factors for outcomes and complications for primary squamous cell carcinoma of the vagina treated with radiation. Gynecol Oncol 105 (3): 641-9, 2007. [PUBMED Abstract]
Eifel PJ, Klopp AH, Berek JS, et al.: Cancer of the cervix, vagina, and vulva. In: DeVita VT Jr, Lawrence TS, Rosenberg SA, et al., eds.: DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 11th ed. Wolters Kluwer, 2019, pp 1171-1210.
Senekjian EK, Frey KW, Anderson D, et al.: Local therapy in stage I clear cell adenocarcinoma of the vagina. Cancer 60 (6): 1319-24, 1987. [PUBMED Abstract]
Chyle V, Zagars GK, Wheeler JA, et al.: Definitive radiotherapy for carcinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol Phys 35 (5): 891-905, 1996. [PUBMED Abstract]
Dalrymple JL, Russell AH, Lee SW, et al.: Chemoradiation for primary invasive squamous carcinoma of the vagina. Int J Gynecol Cancer 14 (1): 110-7, 2004 Jan-Feb. [PUBMED Abstract]
Samant R, Lau B, E C, et al.: Primary vaginal cancer treated with concurrent chemoradiation using Cis-platinum. Int J Radiat Oncol Biol Phys 69 (3): 746-50, 2007. [PUBMED Abstract]
Sharma BB, Rai K, Blunt H, et al.: Pathogenic DPYD Variants and Treatment-Related Mortality in Patients Receiving Fluoropyrimidine Chemotherapy: A Systematic Review and Meta-Analysis. Oncologist 26 (12): 1008-1016, 2021. [PUBMED Abstract]
Lam SW, Guchelaar HJ, Boven E: The role of pharmacogenetics in capecitabine efficacy and toxicity. Cancer Treat Rev 50: 9-22, 2016. [PUBMED Abstract]
Shakeel F, Fang F, Kwon JW, et al.: Patients carrying DPYD variant alleles have increased risk of severe toxicity and related treatment modifications during fluoropyrimidine chemotherapy. Pharmacogenomics 22 (3): 145-155, 2021. [PUBMED Abstract]
Amstutz U, Henricks LM, Offer SM, et al.: Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther 103 (2): 210-216, 2018. [PUBMED Abstract]
Henricks LM, Lunenburg CATC, de Man FM, et al.: DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol 19 (11): 1459-1467, 2018. [PUBMED Abstract]
Lau-Min KS, Varughese LA, Nelson MN, et al.: Preemptive pharmacogenetic testing to guide chemotherapy dosing in patients with gastrointestinal malignancies: a qualitative study of barriers to implementation. BMC Cancer 22 (1): 47, 2022. [PUBMED Abstract]
Brooks GA, Tapp S, Daly AT, et al.: Cost-effectiveness of DPYD Genotyping Prior to Fluoropyrimidine-based Adjuvant Chemotherapy for Colon Cancer. Clin Colorectal Cancer 21 (3): e189-e195, 2022. [PUBMED Abstract]
Baker SD, Bates SE, Brooks GA, et al.: DPYD Testing: Time to Put Patient Safety First. J Clin Oncol 41 (15): 2701-2705, 2023. [PUBMED Abstract]
Treatment of Vaginal Intraepithelial Neoplasia
Vaginal intraepithelial neoplasia (VaIN), the presence of noninvasive squamous cell atypia, is classified by the degree of involvement of the epithelium, as follows:
VaIN 1 is defined as involvement of the upper one-third of the epithelial thickness.
VaIN 2 is defined as involvement of the upper two-thirds of the epithelial thickness.
VaIN 3 is defined as involvement of more than two-thirds of the epithelial thickness. VaIN 3 lesions that involve the full thickness of the epithelium are called carcinoma in situ.
VaIN is associated with a high rate of human papillomavirus (HPV) infection and is thought to have an etiology that is similar to that of cervical intraepithelial neoplasia (CIN).[1–3]
The cervix and vulva are carefully evaluated because vaginal carcinoma in situ is associated with other genital neoplasia, and in some cases, may be an extension of CIN. Vaginal carcinoma in situ is often multifocal and commonly occurs in the vaginal vault. For more information, see Cervical Cancer Treatment.
The extent and type of surgical treatment needed is dependent upon anatomical location, evidence of multifocality, general patient comorbidities, and other specific factors (e.g., anatomical distortion of vaginal vault from prior hysterectomy).[4]
Treatment Options for VaIN
The following treatments have not been directly compared in randomized trials, so their relative efficacy is uncertain.[Level of evidence C3]
Laser therapy [5] after biopsy to rule out invasive components that could be missed with this treatment approach.
Wide local excision with or without skin grafting.[6]
Partial or total vaginectomy, with skin grafting for multifocal or extensive disease.[7]
Intravaginal chemotherapy with 5% fluorouracil (5-FU) cream. This option may be useful in the setting of multifocal lesions.[5,8]
Intracavitary radiation therapy.[9,10] Because of its attendant toxicity and inherent carcinogenicity, this treatment is primarily used in the setting of multifocal or recurrent disease, or when the risk of surgery is high.[1] The entire vaginal mucosa is usually treated.[11]
Imiquimod cream 5%, an immune stimulant used to treat genital warts, is an additional topical therapy that has a reported complete clinical response rate of 50% to 86% in small case series of patients with multifocal high-grade HPV-associated VaIN 2 and VaIN 3.[12] However, it is investigational, and it may have only short-lived efficacy.[13]
Women with VaIN 1 can usually be observed carefully without ablative or surgical treatment because the lesions often regress spontaneously. VaIN 2, the intermediate grade, is managed by careful observation or initial treatment. Although the natural history of VaIN is not precisely known because of its rarity, patients with VaIN 3 are presumed to be at substantial risk of progression to invasive cancer and are treated immediately. Lesions with hyperkeratosis respond better to excision or laser vaporization than to 5-FU.[4]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Eifel PJ, Klopp AH, Berek JS, et al.: Cancer of the cervix, vagina, and vulva. In: DeVita VT Jr, Lawrence TS, Rosenberg SA, et al., eds.: DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology. 11th ed. Wolters Kluwer, 2019, pp 1171-1210.
Daling JR, Madeleine MM, Schwartz SM, et al.: A population-based study of squamous cell vaginal cancer: HPV and cofactors. Gynecol Oncol 84 (2): 263-70, 2002. [PUBMED Abstract]
Smith JS, Backes DM, Hoots BE, et al.: Human papillomavirus type-distribution in vulvar and vaginal cancers and their associated precursors. Obstet Gynecol 113 (4): 917-24, 2009. [PUBMED Abstract]
Wright VC, Chapman W: Intraepithelial neoplasia of the lower female genital tract: etiology, investigation, and management. Semin Surg Oncol 8 (4): 180-90, 1992 Jul-Aug. [PUBMED Abstract]
Krebs HB: Treatment of vaginal intraepithelial neoplasia with laser and topical 5-fluorouracil. Obstet Gynecol 73 (4): 657-60, 1989. [PUBMED Abstract]
Cheng D, Ng TY, Ngan HY, et al.: Wide local excision (WLE) for vaginal intraepithelial neoplasia (VAIN). Acta Obstet Gynecol Scand 78 (7): 648-52, 1999. [PUBMED Abstract]
Indermaur MD, Martino MA, Fiorica JV, et al.: Upper vaginectomy for the treatment of vaginal intraepithelial neoplasia. Am J Obstet Gynecol 193 (2): 577-80; discussion 580-1, 2005. [PUBMED Abstract]
Stefanon B, Pallucca A, Merola M, et al.: Treatment with 5-fluorouracil of 35 patients with clinical or subclinical HPV infection of the vagina. Eur J Gynaecol Oncol 17 (6): 534, 1996. [PUBMED Abstract]
Chyle V, Zagars GK, Wheeler JA, et al.: Definitive radiotherapy for carcinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol Phys 35 (5): 891-905, 1996. [PUBMED Abstract]
Graham K, Wright K, Cadwallader B, et al.: 20-year retrospective review of medium dose rate intracavitary brachytherapy in VAIN3. Gynecol Oncol 106 (1): 105-11, 2007. [PUBMED Abstract]
Kang J, Wethington SL, Viswanathan A: Vaginal cancer. In: Halperin EC, Wazer DE, Perez CE, et al., eds.: Perez & Brady’s Principles and Practice of Radiation Oncology. 7th ed. Wolters Kluwer, 2018, pp 1786-1816.
Iavazzo C, Pitsouni E, Athanasiou S, et al.: Imiquimod for treatment of vulvar and vaginal intraepithelial neoplasia. Int J Gynaecol Obstet 101 (1): 3-10, 2008. [PUBMED Abstract]
Haidopoulos D, Diakomanolis E, Rodolakis A, et al.: Can local application of imiquimod cream be an alternative mode of therapy for patients with high-grade intraepithelial lesions of the vagina? Int J Gynecol Cancer 15 (5): 898-902, 2005 Sep-Oct. [PUBMED Abstract]
Treatment of Stage I Vaginal Cancer
The treatment options for stage I vaginal cancer have not been directly compared in randomized trials.[Level of evidence C2] Because of differences in patient selection, expertise in treating local disease, and staging criteria, it is difficult to determine whether there are differences in disease control rates.
Treatment Options for Stage I Squamous Cell Carcinoma (SCC) of the Vagina
These tumors may respond to intracavitary brachytherapy alone,[1] but treatment usually begins with external-beam radiation therapy (EBRT).[2]
EBRT is required for bulky lesions or lesions that encompass the entire vagina.[1] For lesions in the lower third of the vagina, elective radiation therapy is often administered to the patient’s pelvic and/or inguinal lymph nodes.[1,2]
Wide local excision or total vaginectomy with vaginal reconstruction may be performed, especially in lesions of the upper vagina. In cases with close or positive surgical margins, adjuvant radiation therapy is often added.[6]
For lesions in the upper third of the vagina, radical vaginectomy and pelvic lymphadenectomy should be considered. Construction of a neovagina may be performed if feasible and if desired by the patient.[6,7]
In lesions of the lower third, inguinal lymphadenectomy should be considered. In cases with close or positive surgical margins, adjuvant radiation therapy should be considered.[6]
EBRT [2] and/or combination of interstitial and intracavitary radiation therapy may be administered to a dose of at least 75 Gy to the primary tumor.[1,8]
For lesions of the lower third of the vagina, elective radiation therapy of 45 Gy to 50 Gy is given to the pelvic and/or inguinal lymph nodes.[1,2]
Treatment Options for Stage I Adenocarcinoma of the Vagina
Total radical vaginectomy and hysterectomy with lymph node dissection are indicated because the tumor spreads subepithelially.
The deep pelvic lymph nodes are dissected if the lesion invades the upper vagina, and the inguinal lymph nodes are removed if the lesion originates in the lower vagina.
Construction of a neovagina may be performed if feasible and if desired by the patient.[6]
In cases with close or positive surgical margins, adjuvant radiation therapy is often given.[6,7]
Radiation therapy.
Intracavitary and interstitial radiation are administered to a dose of at least 75 Gy to the primary tumor.[1]
For lesions in the lower third of the vagina, elective radiation therapy of 45 Gy to 50 Gy is given to the pelvic and/or inguinal lymph nodes.[1,9]
Combined local therapy in selected cases, which may include wide local excision, lymph node sampling, and interstitial therapy.[10]
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Perez CA, Camel HM, Galakatos AE, et al.: Definitive irradiation in carcinoma of the vagina: long-term evaluation of results. Int J Radiat Oncol Biol Phys 15 (6): 1283-90, 1988. [PUBMED Abstract]
Frank SJ, Jhingran A, Levenback C, et al.: Definitive radiation therapy for squamous cell carcinoma of the vagina. Int J Radiat Oncol Biol Phys 62 (1): 138-47, 2005. [PUBMED Abstract]
Tran PT, Su Z, Lee P, et al.: Prognostic factors for outcomes and complications for primary squamous cell carcinoma of the vagina treated with radiation. Gynecol Oncol 105 (3): 641-9, 2007. [PUBMED Abstract]
Lian J, Dundas G, Carlone M, et al.: Twenty-year review of radiotherapy for vaginal cancer: an institutional experience. Gynecol Oncol 111 (2): 298-306, 2008. [PUBMED Abstract]
Tjalma WA, Monaghan JM, de Barros Lopes A, et al.: The role of surgery in invasive squamous carcinoma of the vagina. Gynecol Oncol 81 (3): 360-5, 2001. [PUBMED Abstract]
Stock RG, Chen AS, Seski J: A 30-year experience in the management of primary carcinoma of the vagina: analysis of prognostic factors and treatment modalities. Gynecol Oncol 56 (1): 45-52, 1995. [PUBMED Abstract]
Rubin SC, Young J, Mikuta JJ: Squamous carcinoma of the vagina: treatment, complications, and long-term follow-up. Gynecol Oncol 20 (3): 346-53, 1985. [PUBMED Abstract]
Andersen ES: Primary carcinoma of the vagina: a study of 29 cases. Gynecol Oncol 33 (3): 317-20, 1989. [PUBMED Abstract]
Chyle V, Zagars GK, Wheeler JA, et al.: Definitive radiotherapy for carcinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol Phys 35 (5): 891-905, 1996. [PUBMED Abstract]
Senekjian EK, Frey KW, Anderson D, et al.: Local therapy in stage I clear cell adenocarcinoma of the vagina. Cancer 60 (6): 1319-24, 1987. [PUBMED Abstract]
Treatment of Stages II, III, and IVa Vaginal Cancer
The treatment options for stages II, III, and IVa vaginal cancer have not been directly compared in randomized trials.[Level of evidence C2] As a result of differences in patient selection, expertise in treating local disease, and staging criteria, it is difficult to determine whether there are differences in disease control rates.
Radiation therapy is the most common treatment for patients with stages II, III, and IVa vaginal cancer.
Treatment Options for Stages II, III, and IVa Squamous Cell Carcinoma (SCC) and Adenocarcinoma of the Vagina
External-beam radiation therapy (EBRT) alone or in combination with interstitial and/or intracavitary brachytherapy.[1–5] For example, EBRT for a period of 5 to 6 weeks (including the pelvic lymph nodes) followed by an interstitial and/or intracavitary implant for a total tumor dose of 75 Gy to 80 Gy and a dose to the lateral pelvic wall of 55 Gy to 60 Gy.[1–3]
For lesions in the lower third of the vagina, elective radiation therapy of 45 Gy to 50 Gy is given to the pelvic and/or inguinal lymph nodes.[1,2,6]
Surgery.
Radical vaginectomy or pelvic exenteration is performed with or without radiation therapy.[7–10]
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Perez CA, Camel HM, Galakatos AE, et al.: Definitive irradiation in carcinoma of the vagina: long-term evaluation of results. Int J Radiat Oncol Biol Phys 15 (6): 1283-90, 1988. [PUBMED Abstract]
Chyle V, Zagars GK, Wheeler JA, et al.: Definitive radiotherapy for carcinoma of the vagina: outcome and prognostic factors. Int J Radiat Oncol Biol Phys 35 (5): 891-905, 1996. [PUBMED Abstract]
Frank SJ, Jhingran A, Levenback C, et al.: Definitive radiation therapy for squamous cell carcinoma of the vagina. Int J Radiat Oncol Biol Phys 62 (1): 138-47, 2005. [PUBMED Abstract]
Tran PT, Su Z, Lee P, et al.: Prognostic factors for outcomes and complications for primary squamous cell carcinoma of the vagina treated with radiation. Gynecol Oncol 105 (3): 641-9, 2007. [PUBMED Abstract]
Lian J, Dundas G, Carlone M, et al.: Twenty-year review of radiotherapy for vaginal cancer: an institutional experience. Gynecol Oncol 111 (2): 298-306, 2008. [PUBMED Abstract]
Andersen ES: Primary carcinoma of the vagina: a study of 29 cases. Gynecol Oncol 33 (3): 317-20, 1989. [PUBMED Abstract]
Rubin SC, Young J, Mikuta JJ: Squamous carcinoma of the vagina: treatment, complications, and long-term follow-up. Gynecol Oncol 20 (3): 346-53, 1985. [PUBMED Abstract]
Stock RG, Chen AS, Seski J: A 30-year experience in the management of primary carcinoma of the vagina: analysis of prognostic factors and treatment modalities. Gynecol Oncol 56 (1): 45-52, 1995. [PUBMED Abstract]
Tjalma WA, Monaghan JM, de Barros Lopes A, et al.: The role of surgery in invasive squamous carcinoma of the vagina. Gynecol Oncol 81 (3): 360-5, 2001. [PUBMED Abstract]
Boronow RC, Hickman BT, Reagan MT, et al.: Combined therapy as an alternative to exenteration for locally advanced vulvovaginal cancer. II. Results, complications, and dosimetric and surgical considerations. Am J Clin Oncol 10 (2): 171-81, 1987. [PUBMED Abstract]
Rajagopalan MS, Xu KM, Lin JF, et al.: Adoption and impact of concurrent chemoradiation therapy for vaginal cancer: a National Cancer Data Base (NCDB) study. Gynecol Oncol 135 (3): 495-502, 2014. [PUBMED Abstract]
Treatment of Stage IVb Vaginal Cancer
Treatment Options for Stage IVb Squamous Cell Carcinoma (SCC) and Adenocarcinoma of the Vagina
Radiation therapy (for palliation of symptoms) with or without chemotherapy.
For patients with stage IVb disease, current therapy is inadequate. No established anticancer drugs have demonstrated clinical benefit, although patients are often treated with regimens used to treat cervical cancer.
Concurrent chemotherapy using fluorouracil or cisplatin-based therapy and radiation therapy is sometimes advocated on the basis of results extrapolated from cervical cancer management strategies.[1–3] Evidence is limited to small case series, and the incremental impact on patient survival and local disease control is not well defined.[Level of evidence C3] For more information, see Cervical Cancer Treatment.
Because stage IVb vaginal cancer is rare, these patients are candidates for clinical trials to improve survival or local control. Information about ongoing clinical trials is available from the NCI website.
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.
Dalrymple JL, Russell AH, Lee SW, et al.: Chemoradiation for primary invasive squamous carcinoma of the vagina. Int J Gynecol Cancer 14 (1): 110-7, 2004 Jan-Feb. [PUBMED Abstract]
Samant R, Lau B, E C, et al.: Primary vaginal cancer treated with concurrent chemoradiation using Cis-platinum. Int J Radiat Oncol Biol Phys 69 (3): 746-50, 2007. [PUBMED Abstract]
Treatment of Recurrent Vaginal Cancer
Patients with recurrent vaginal cancer have a very poor prognosis. Most recurrences occur in the first 2 years after treatment.
Some patients with centrally recurrent vaginal cancers are candidates for pelvic exenteration or radiation therapy. In a large series, only 5 of 50 patients with recurrence were salvaged using surgery or radiation therapy. All five of these salvaged patients originally presented with stage I or II disease and had tumor recurrence in the central pelvis.[1]
No established anticancer drugs have demonstrated clinical benefit, although patients are often treated with regimens used to treat cervical cancer. If patients are eligible, participation in clinical trials should be considered. Information about ongoing clinical trials is available from the NCI website.
Current Clinical Trials
Use our advanced clinical trial search to find NCI-supported cancer clinical trials that are now enrolling patients. The search can be narrowed by location of the trial, type of treatment, name of the drug, and other criteria. General information about clinical trials is also available.
References
Stock RG, Chen AS, Seski J: A 30-year experience in the management of primary carcinoma of the vagina: analysis of prognostic factors and treatment modalities. Gynecol Oncol 56 (1): 45-52, 1995. [PUBMED Abstract]
Latest Updates to This Summary (04/03/2025)
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
Updated statistics with estimated new cases and deaths for 2025 (cited American Cancer Society as reference 3).
This summary is written and maintained by the PDQ Adult Treatment Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® Cancer Information for Health Professionals pages.
About This PDQ Summary
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of vaginal cancer. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the PDQ Adult Treatment Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).
Board members review recently published articles each month to determine whether an article should:
be discussed at a meeting,
be cited with text, or
replace or update an existing article that is already cited.
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Vaginal Cancer Treatment are:
Fumiko Chino, MD (MD Anderson Cancer Center)
Olga T. Filippova, MD (Lenox Hill Hospital)
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website’s Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Adult Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”
The preferred citation for this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Vaginal Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/vaginal/hp/vaginal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389242]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.
Disclaimer
Based on the strength of the available evidence, treatment options may be described as either “standard” or “under clinical evaluation.” These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.
Vaginal cancer is a disease in which malignant (cancer) cells form in the vagina.
Older age and having an HPV infection are risk factors for vaginal cancer.
Signs and symptoms of vaginal cancer include pain or abnormal vaginal bleeding.
Tests that examine the vagina and other organs in the pelvis are used to diagnose vaginal cancer.
Certain factors affect prognosis (chance of recovery) and treatment options.
Vaginal cancer is a disease in which malignant (cancer) cells form in the vagina.
The vagina is the canal leading from the cervix (the opening of the uterus) to the outside of the body. At birth, a baby passes out of the body through the vagina (also called the birth canal).
EnlargeAnatomy of the female reproductive system. The organs in the female reproductive system include the uterus, ovaries, fallopian tubes, cervix, and vagina. The uterus has a muscular outer layer called the myometrium and an inner lining called the endometrium.
Vaginal cancer is not common. There are two main types of vaginal cancer:
Squamous cell carcinoma: Cancer that forms in the thin, flat cells lining the inside of the vagina. Squamous cell vaginal cancer spreads slowly and usually stays near the vagina, but may spread to the lungs, liver, or bone. This is the most common type of vaginal cancer.
Adenocarcinoma: Cancer that begins in glandular cells. Glandular cells in the lining of the vagina make and release fluids such as mucus. Adenocarcinoma is more likely than squamous cell cancer to spread to the lungs and lymph nodes. A rare type of adenocarcinoma (clear cell adenocarcinoma) is linked to being exposed to diethylstilbestrol (DES) before birth. Adenocarcinomas that are not linked with being exposed to DES are most common in women after menopause.
Older age and having an HPV infection are risk factors for vaginal cancer.
Anything that increases a person’s chance of getting a disease is called a risk factor. Not every person with one or more of these risk factors will develop vaginal cancer, and it will develop in people who don’t have any known risk factors. Talk with your doctor if you think you may be at risk. Risk factors for vaginal cancer include the following:
Being exposed to DES while in the mother’s womb. In the 1950s, the drug DES was given to some pregnant women to prevent miscarriage (premature birth of a fetus that cannot survive). This is linked to a rare form of vaginal cancer called clear cell adenocarcinoma. The rates of this disease were highest in the mid-1970s, and it is extremely rare now.
Having had a hysterectomy for tumors that were benign (not cancer) or cancer.
Signs and symptoms of vaginal cancer include pain or abnormal vaginal bleeding.
Vaginal cancer often does not cause early signs or symptoms. It may be found during a routine pelvic exam and Pap test. Signs and symptoms may be caused by vaginal cancer or by other conditions. Check with your doctor if you have any of the following:
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. EnlargePelvic exam. A doctor or nurse inserts one or two lubricated, gloved fingers of one hand into the vagina and presses on the lower abdomen with the other hand. This is done to feel the size, shape, and position of the uterus and ovaries. The vagina, cervix, fallopian tubes, and rectum are also checked.
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. EnlargePap test. A speculum is inserted into the vagina to widen it. Then, a brush is inserted into the vagina to collect cells from the cervix. The cells are checked under a microscope for signs of disease.
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.
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: The removal of cells or tissues from the vagina and cervix so they can be viewed under a microscope by a pathologist to check for signs of cancer. If a Pap test shows abnormal cells in the vagina, a biopsy may be done during a colposcopy.
Certain factors affect prognosis (chance of recovery) and treatment options.
After vaginal cancer has been diagnosed, tests are done to find out if cancer cells have spread within the vagina or to other parts of the body.
There are three ways that cancer spreads in the body.
Cancer may spread from where it began to other parts of the body.
In vaginal intraepithelial neoplasia (VaIN), abnormal cells are found in tissue lining the inside of the vagina.
The following stages are used for vaginal cancer:
Stage I
Stage II
Stage III
Stage IV
Vaginal cancer may recur (come back) after it has been treated.
After vaginal cancer has been diagnosed, tests are done to find out if cancer cells have spread within the vagina or to other parts of the body.
The process used to find out if cancer has spread within the vagina or to other parts of the body is called staging. The information gathered from the staging process determines the stage of the disease. It is important to know the stage in order to plan treatment. The following procedures may be used in the staging process:
Chest x-ray: An x-ray of the organs and bones inside the chest. An x-ray is a type of energy beam that can go through the body and onto film, making a picture of areas inside the body.
CT scan (CAT scan): A procedure that makes a series of detailed pictures of areas inside the body such as the abdomen or pelvis, 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.
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).
PET scan (positron emission tomography scan): A procedure to find malignanttumorcells in the body. A small amount of radioactiveglucose (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.
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. EnlargeCystoscopy. A cystoscope (a thin, tube-like instrument with a light and a lens for viewing) is inserted through the urethra into the bladder. Fluid is used to fill the bladder. The doctor looks at an image of the inner wall of the bladder on a computer monitor to check for abnormal areas.
Proctoscopy: A procedure to look inside the rectum and anus to check for abnormal areas, using a proctoscope. A proctoscope is a thin, tube-like instrument with a light and a lens for viewing the inside of the rectum and anus. It may also have a tool to remove tissue samples, which are checked under a microscope for signs of cancer.
Biopsy: A biopsy may be done to find out if cancer has spread to the cervix. A sample of tissue is removed from the cervix and viewed under a microscope. A biopsy that removes only a small amount of tissue is usually done in the doctor’s office. A cone biopsy (removal of a larger, cone-shaped piece of tissue from the cervix and cervical canal) is usually done in the hospital. A biopsy of the vulva may also be done to see if cancer has spread there.
There are three ways that cancer spreads in the body.
Tissue. The cancer spreads from where it began by growing into nearby areas.
Lymph system. The cancer spreads from where it began by getting into the lymph system. The cancer travels through the lymph vessels to other parts of the body.
Blood. The cancer spreads from where it began by getting into the blood. The cancer travels through the blood vessels to other parts of the body.
Cancer may spread from where it began to other parts of the body.
When cancer spreads to another part of the body, it is called metastasis. Cancer cells break away from where they began (the primary tumor) and travel through the lymph system or blood.
Lymph system. The cancer gets into the lymph system, travels through the lymph vessels, and forms a tumor (metastatic tumor) in another part of the body.
Blood. The cancer gets into the blood, travels through the blood vessels, and forms a tumor (metastatic tumor) in another part of the body.
The metastatic tumor is the same type of cancer as the primary tumor. For example, if vaginal cancer spreads to the lung, the cancer cells in the lung are actually vaginal cancer cells. The disease is metastatic vaginal cancer, not lung cancer.
Many cancer deaths are caused when cancer moves from the original tumor and spreads to other tissues and organs. This is called metastatic cancer. This animation shows how cancer cells travel from the place in the body where they first formed to other parts of the body.
In vaginal intraepithelial neoplasia (VaIN), abnormal cells are found in tissue lining the inside of the vagina.
These abnormal cells are not cancer. Vaginal intraepithelial neoplasia (VaIN) is grouped based on how deep the abnormal cells are in the tissue lining the vagina:
VaIN 1: Abnormal cells are found in the outermost one third of the tissue lining the vagina.
VaIN 2: Abnormal cells are found in the outermost two-thirds of the tissue lining the vagina.
VaIN 3: Abnormal cells are found in more than two-thirds of the tissue lining the vagina. When VaIN 3 lesions are found in the full thickness of the tissue lining the vagina, it is called carcinoma in situ.
VaIN may become cancer and spread into the vaginal wall.
Stage IVB: Cancer has spread to parts of the body that are not near the vagina, such as the lung or bone.
Vaginal cancer may recur (come back) after it has been treated.
The cancer may come back in the vagina or in other parts of the body.
Treatment Option Overview
Key Points
There are different types of treatment for patients with vaginal cancer.
The following types of treatment are used:
Surgery
Radiation therapy
Chemotherapy
New types of treatment are being tested in clinical trials.
Immunotherapy
Radiosensitizers
Treatment for vaginal 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 vaginal cancer.
Different types of treatments are available for patients with vaginal 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.
The following types of surgery may be used to treat VaIN:
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.
Wide local excision: A surgical procedure that takes out the cancer and some of the healthy tissue around it.
The following types of surgery may be used to treat vaginal cancer:
Wide local excision: A surgical procedure that takes out the cancer and some of the healthy tissue around it.
Vaginectomy: Surgery to remove all or part of the vagina. Skin grafts from other parts of the body may be needed to reconstruct the vagina.
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 vaginalhysterectomy. 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. EnlargeHysterectomy. The uterus is surgically removed with or without other organs or tissues. In a total hysterectomy, the uterus and cervix are removed. In a total hysterectomy with salpingo-oophorectomy, (a) the uterus plus one (unilateral) ovary and fallopian tube are removed; or (b) the uterus plus both (bilateral) ovaries and fallopian tubes are removed. In a radical hysterectomy, the uterus, cervix, both ovaries, both fallopian tubes, and nearby tissue are removed. These procedures are done using a low transverse incision or a vertical incision.
Lymph node dissection: A surgical procedure in which lymph nodes are removed and a sample of tissue is checked under a microscope for signs of cancer. This procedure is also called lymphadenectomy. If the cancer is in the upper vagina, the pelvic lymph nodes may be removed. If the cancer is in the lower vagina, lymph nodes in the groin may be removed.
Pelvic exenteration: Surgery to remove the lower colon, rectum, bladder, cervix, vagina, and ovaries. Nearby lymph nodes are also removed. Artificial openings (stoma) are made for urine and stool to flow from the body into a collection bag.
After the doctor removes all the cancer that can be seen at the time of the surgery, some patients may be given radiation therapy after surgery to kill any cancer cells that are left. Treatment given after the surgery, to lower the risk that the cancer will come back, is called adjuvant therapy.
Radiation therapy
Radiation therapy is a cancer treatment that uses high-energy x-rays or other types of radiation to kill cancer cells or keep them from growing. There are two types of radiation therapy:
External radiation therapy uses a machine outside the body to send radiation toward the area of the body with cancer.
The 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 vaginal 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 affect 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.
New types of treatment are being tested in clinical trials.
This summary section describes treatments that are being studied in clinical trials. It may not mention every new treatment being studied. Information about clinical trials is available from the NCI website.
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.
Imiquimod is an immune response modifier that is being studied to treat vaginal lesions and is applied to the skin in a cream.
Radiosensitizers
Radiosensitizers are drugs that make tumor cells more sensitive to radiation therapy. Combining radiation therapy with radiosensitizers may kill more tumor cells.
Treatment for vaginal 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.
Treatment of Vaginal Intraepithelial Neoplasia (VaIN)
Although no anticancer drugs have been shown to help patients with stage IVB vaginal cancer live longer, they are often treated with regimens used for cervical cancer. For more information, see Cervical Cancer Treatment.
Although no anticancer drugs have been shown to help patients with recurrent vaginal cancer live longer, they are often treated with regimens used for cervical cancer. For more information, see Cervical Cancer Treatment.
Use our clinical trial search to find NCI-supported cancer clinical trials that are accepting patients. You can search for trials based on the type of cancer, the age of the patient, and where the trials are being done. General information about clinical trials is also available.
Physician Data Query (PDQ) is the National Cancer Institute’s (NCI’s) comprehensive cancer information database. The PDQ database contains summaries of the latest published information on cancer prevention, detection, genetics, treatment, supportive care, and complementary and alternative medicine. Most summaries come in two versions. The health professional versions have detailed information written in technical language. The patient versions are written in easy-to-understand, nontechnical language. Both versions have cancer information that is accurate and up to date and most versions are also available in Spanish.
PDQ is a service of the NCI. The NCI is part of the National Institutes of Health (NIH). NIH is the federal government’s center of biomedical research. The PDQ summaries are based on an independent review of the medical literature. They are not policy statements of the NCI or the NIH.
Purpose of This Summary
This PDQ cancer information summary has current information about the treatment of vaginal cancer. It is meant to inform and help patients, families, and caregivers. It does not give formal guidelines or recommendations for making decisions about health care.
Reviewers and Updates
Editorial Boards write the PDQ cancer information summaries and keep them up to date. These Boards are made up of experts in cancer treatment and other specialties related to cancer. The summaries are reviewed regularly and changes are made when there is new information. The date on each summary (“Updated”) is the date of the most recent change.
The information in this patient summary was taken from the health professional version, which is reviewed regularly and updated as needed, by the PDQ Adult Treatment Editorial Board.
Clinical Trial Information
A clinical trial is a study to answer a scientific question, such as whether one treatment is better than another. Trials are based on past studies and what has been learned in the laboratory. Each trial answers certain scientific questions in order to find new and better ways to help cancer patients. During treatment clinical trials, information is collected about the effects of a new treatment and how well it works. If a clinical trial shows that a new treatment is better than one currently being used, the new treatment may become “standard.” Patients may want to think about taking part in a clinical trial. Some clinical trials are open only to patients who have not started treatment.
Clinical trials can be found online at NCI’s website. For more information, call the Cancer Information Service (CIS), NCI’s contact center, at 1-800-4-CANCER (1-800-422-6237).
Permission to Use This Summary
PDQ is a registered trademark. The content of PDQ documents can be used freely as text. It cannot be identified as an NCI PDQ cancer information summary unless the whole summary is shown and it is updated regularly. However, a user would be allowed to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks in the following way: [include excerpt from the summary].”
The best way to cite this PDQ summary is:
PDQ® Adult Treatment Editorial Board. PDQ Vaginal Cancer Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: /types/vaginal/patient/vaginal-treatment-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389348]
Images in this summary are used with permission of the author(s), artist, and/or publisher for use in the PDQ summaries only. If you want to use an image from a PDQ summary and you are not using the whole summary, you must get permission from the owner. It cannot be given by the National Cancer Institute. Information about using the images in this summary, along with many other images related to cancer can be found in Visuals Online. Visuals Online is a collection of more than 3,000 scientific images.
Disclaimer
The information in these summaries should not be used to make decisions about insurance reimbursement. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s E-mail Us.
We offer evidence-based supportive and palliative care information for health professionals on the assessment and management of cancer-related symptoms and conditions.