15 Cutaneous T-Cell Lymphoma FRANCINE M. FOSS, MD MARIEFRANCE DEMIERRE, MD, FRCPC Primary cutaneous T-cell lymphomas (CTCLs) comprise a constellation of heterogeneous lymphoproliferative disorders characterized by clonal accumulation of neoplastic T lymphocytes in the skin. Together with primary cutaneous B-cell lymphomas, they are the second most common group of extranodal non-Hodgkin’s lymphomas, following primary gastrointestinal lymphomas. Cutaneous T-cell lymphomas differ clinically and histologically from nodal lymphomas of the same histology. Before treating a CTCL, it is essential to determine that the patient does not have cutaneous manifestations of a primary nodal lymphoma.1 EPIDEMIOLOGY Cutaneous T-cell lymphoma comprises approximately 80 percent of all primary cutaneous lymphomas (Table 15–1). The majority of C-TCL consist of mycosis fungoides (MF), which takes its name from the mushroom-shaped tumor nodules resulting from the vertical proliferation of infiltrating cells, and Sézary syndrome (SS). Anaplastic large cell lymphomas (CD30+) comprise 25 percent, and CD30- peripheral T-cell lymphomas comprise 10 percent. The National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) program reports an increase in the overall incidence of CTCL in the United States from 1973 to 1984, with a rise from approximately 0.2 cases per 100,000 population in 1973 to 0.4 cases per 100,000 population in 1984.2 However, there was no evidence of increasing incidence rate during the period of 1983 through 1992.3 These data included follow-up through 1994. Overall, the incidence rate from 1973 through 1992 was 0.36 per 105 person-years. These changes in incidence are not well understood, but it is probable that modern diagnostic techniques, including immunohistochemistry, electron microscopy, and molecular analysis, contribute to the initial apparent increase in incidence by identifying patients earlier in the course of the disease. Survival was noted to improve substantially over the 20-year period of study and was unrelated to variations of age, race, gender, or marital status.4 Increased survival may be due to improvements in treatment, changes in the biology of the disease, or the inclusion of a proportion of biologically benign disorders, such as lymphomatoid papulosis or large plaque parapsoriasis, which might be misclassified as MF. Recent reports suggest a nearly normal life expectancy for patients with very early-stage disease.5,6 Although MF/SS has been reported in young individuals, the incidence increases with age. Median age at the time of diagnosis is 55 to 60 years. The incidence is highest in African Americans, followed by Caucasians and Asian Americans. Table 15–1. LYMPHOMAS PRESENTING IN THE SKIN Type of Lymphoma Mycosis fungoides/SS CD30+ ALCL Peripheral T-cell CD30– B cell (FSC, DLC) MALT type Incidence (%) 45 25 10 15 <5 SS = Sézary syndrome; ALCL = anaplastic large cell lymphomas; FSC = follicular small cell; DLC = diffuse large cell; MALT = mucosa-associated lymphoid tissue. 259 260 MALIGNANT LYMPHOMAS Both incidence and mortality are higher among men than among women. Mycosis fungoides and SS are classified in the revised European-American lymphoma (REAL) classification as low-grade T-cell lymphomas. Other less common types of CTCLs or lymphoproliferative disorders include the CD30+ lymphoproliferative disorders (lymphomatoid papulosis, primary anaplastic large cell lymphoma of the skin), pagetoid reticulosis (Woringer-Kolopp disease), granulomatous slack skin, large plaque parapsoriasis, and T-γ lymphoproliferative disorder. The intermediateor high-grade variants of T-cell lymphoma are human T-cell lymphotropic virus type I (HTLV-I)associated adult T-cell leukemia/lymphoma, peripheral T-cell lymphoma, and CD8+ epidermotropic cytotoxic T-cell lymphoma. This chapter addresses only the biology and treatment of MF/SS, except where specifically noted to the contrary. ETIOLOGY Many studies have attempted to demonstrate the etiology of MF/SS, but the results are contradictory and generally inconclusive. Extensive studies of chronic antigenic stimulation resulting from exposure to industrial chemicals have failed to implicate these agents conclusively.7,8 Epidermal Staphylococcus aureus colonization and bacteremia were noted in one series of patients, but the study design was unable to distinguish between microbial pathogenesis and infection secondary to immunosuppression.9 Early attempts to demonstrate a viral etiology of MF/SS, based on identification of the HTLV-I in a single patient and observation of similarities between MF/SS and HTLV-I-associated adult leukemia/lymphoma, produced no serologic consistency. More recently, however, molecular analysis has revealed the presence of portions of the HTLV-I genome in a subset of patients with MF/SS.10–13 Nevertheless, epidemiologic data to date fail to support a role for HTLV-I or HTLV-II in MF/SS.14 Other investigations are focusing on a possible role for HTLV-V.15 Cytokine-driven proliferation of T cells has been implicated in the etiology of CTCL. Interleukin (IL)-7 is a growth factor for Sézary cells isolated from patients with SS. The proliferative response to IL-7 can be abrogated by an anti-IL-7 monoclonal antibody.16 In addition, IL-2 and IL-7 co-stimulate CTCL cells potentially by an autocrine or paracrine growth-stimulatory mechanism since IL-7 is secreted by normal keratinocytes.17 These findings suggest that cytokine mediation may be involved in the development of SS and other CTCLs. This hypothesis is supported by the demonstration that transgenic mice carrying the gene for IL-7 develop a progressive cutaneous disorder involving T-lymphocyte infiltration.18 Chromosomal abnormalities occur commonly in CTCL,19 but they have not been shown to be specific. Chromosome 1 is affected most frequently, followed by chromosome 6. The region between 1p22 and 1p36 has been proposed as the site containing a gene that encodes a protein important for either the malignant transformation or progression in MF/SS.20 A recent study found that epidermotropic T cells from patients with MF/SS expressed high levels of telomerase, an enzyme that replaces terminal chromosome sequences lost during serial replication.21 Telomerase has been reported to be overexpressed in many tumors but not in normal tissue,22 suggesting that telomerase expression may be necessary for tumorigenesis. It is possible that telomerase inhibition may limit tumor cell growth.23,24 CLINICAL MANIFESTATIONS Mycosis fungoides is characterized by infiltration of the skin by atypical small T lymphocytes with hyperconvoluted cerebriform nuclei (mycosis cells) that usually express a mature peripheral T-cell (CD4+) phenotype. A suppressor phenotype (CD8+) is seen infrequently. Expression of an IL-2 receptor (CD25) is seen in approximately half of MF/SS cases. Epidermotropism or mononuclear exocytosis into the epidermis and Pautrier’s microabscesses in the epidermis are characteristic histopathologic findings (Figure 15–1). Cutaneous lesions can occur as patches, plaques, or tumors, but patients may have lesions of two or three stages simultaneously. The patch stage consists of multiple scaly erythematous patches that occur most frequently over the trunk and extremities and often are associated with severe pruritis. The patch stage resembles psoriasis or eczema Cutaneous T-Cell Lymphoma 261 Figure 15–2. Plaque-stage cutaneous T-cell lymphoma. Lesions are raised, scaling, and erythematous and may resemble eczema or parapsoriasis. Figure 15–1. Skin biopsy from a patient with stage IA cutaneous T-cell lymphoma showing exocytosis of lymphocytes into the epidermis, forming of Pautrier’s microabscesses (×40 original magnification). and may persist for months or even years with a waxing-and-waning clinical picture before progressing to the plaque stage. In the plaque stage, lesions are oval or circular, well demarcated, and elevated (Figure 15–2). Patches may regress spontaneously or coalesce into larger plaques. Dermal thickening of the skin by T-cell infiltration may lead to an infrequent but classic appearance on the face known as leonine facies. The differential diagnosis of plaque-stage disease is shown in Table 15–2 and includes nonmalignant conditions such as eczema, psoriasis, and parapsoriasis. Although MF is usually an indolent disease, the tumor stage can be very aggressive as neoplastic cells enter a vertical growth phase that presents clinically as expanding nodules (Figure 15–3). These are seen most commonly on the face and in major skin folds: the axilla, groin, and inframammary areas. Nodules may experience central necrosis with ulceration that may lead to secondary bacterial infection Table 15–2. DIFFERENTIAL DIAGNOSIS OF PATCHES AND PLAQUES Differential Diagnosis Distribution Pathology Atopic dermatitis Flexural areas, hands Spongiosis, exocytosis Contact dermatitis Areas of contact Spongiosis, exocytosis, occasional eosinophilia Fungal infection Intertriginous areas, nails Hyphae within stratum corneum Psoriasis Elbows, knees, nails Epidermal hyperplasia, absence of granular cell layer, dilated capillaries in papillary dermis, and PMNs in dermal papillae and epidermis Parapsoriasis Flanks, submammary area Perivascular lymphocytic infiltrate with occasional epidermotropism PMN = polymorphonuclear neutrophil leukocyte. 262 MALIGNANT LYMPHOMAS percent have cutaneous tumors, and 12 percent have generalized erythroderma. Palpable adenopathy is infrequent in patients with limited plaque, but it occurs in approximately 50 percent of patients with extensive plaques, tumors, or erythroderma. Sézary syndrome, the leukemic variant of MF seen in approximately 5 percent of newly reported cases of CTCL, shares many of the clinical and immunohistologic features of MF. It is distinguished from MF, however, by its characteristic triad of diffuse erythroderma, generalized lymphadenopathy, and the presence of hyperconvoluted neoplastic T lymphocytes (CD4+CD7-) in skin, lymph nodes, and peripheral blood. In patients with SS, 20 percent or more of the circulating lymphocytes are Sézary cells. Circulating Sézary cells typically bear the same immunophenotype as skin infiltrates, but there may be antigen discordance, especially for the CD25 antigen, which often is absent from circulating Sézary cells. Patients with SS may demonstrate manifestations of scaling or fissuring of the palms and soles, alopecia, ectropion, or loss of nails (Fig- Figure 15–3. Skin biopsy of tumor-stage cutaneous T-cell lymphoma showing extensive dermal and epidermal infiltration with atypical lymphoid cell infiltrate with marked pleomorphism (× 40 original magnification). (Figure 15–4). The differential diagnosis for tumorstage CTCL includes lymphomatoid papulosis and B-cell lymphoproliferative disorders, including pseudolymphoma (Table 15–3). Biopsy and immunophenotypic staining are required to make a definitive diagnosis. Recent evidence suggests that progression to the tumor stage may involve a loss of Fas receptor expression, enabling malignant cells to escape apoptotic signals.25 Other studies suggest a possible relationship promoter hypermethylation of p16INK4a and both disease progression and prognosis.26 Mutations and functional loss of the p53 tumor suppressor gene have been reported in MF, as have mutations and overexpression of the myc, ras, and lck oncogenes. At diagnosis, 42 percent of MF patients have limited plaques involving 10 percent or less of total body surface, 30 percent have extensive plaques, 16 Figure 15–4. Patient with extensive tumor-stage cutaneous T-cell lymphoma. Cutaneous tumors may ulcerate and become secondarily infected. Cutaneous T-Cell Lymphoma 263 Table 15–3. DIFFERENTIAL DIAGNOSIS OF SKIN TUMORS Differential Diagnosis Pseudolymphoma Lymphomatoid papulosis Leukemia cutis Metastatic disease Distribution Pathology Face, neck, mammary areas, groin Trunk and extremities Face, trunk, extremities Scalp, skin overlying primary tumor or lymphatic drainage Infiltrate mixed, in dermis, nonclonal Wedge-shaped infiltrate, mild perivascular interstitial infiltrate Grenz zone between dermis and epidermis Malignant cells infiltrating dermis and/or subcutaneous tissue ure 15–5). Ankle edema, malaise, and weight loss are common, as are chills and fever resulting from impaired thermoregulation. IMMUNOPATHOGENESIS The immunopathogenesis of MF/SS has been reviewed.27 Early stages of MF/SS are characterized by epidermotropism, that is, localization of the malignant T cell (memory helper T cell CD3+CD4+CD45RO+) to the skin. The mechanisms for this epidermotropism are likely multifactorial. The memory T cells that home to the skin express both cutaneous lymphocyte antigen (CLA), a skin-homing receptor not commonly expressed on T cells, and lymphocyte function–associated antigen 1 (LFA-1). The former interacts with E-selectin, an adhesion molecule found on endothelial cells of cutaneous venules. The latter, expressed by all T cells, interacts with intercellular adhesion molecule 1, which is produced by keratinocytes in response to interferon-γ (IFN-γ). It appears that an IFN-γ inducible protein (IP-10), chemotactic for CD4+ lymphocytes, tumor necrosis factor-α, and a monokine induced by IFN-γ , could be important in the events leading to epidermotropism.28,29 The lesions in MF/SS have been shown to be Th2 cells since they secrete IL-4, IL-5, IL-6, and IL-10.27 An excess of Th2 cytokine production could account for the histopathologic changes seen in the evolution of early plaque to tumor or erythroderma. Several immune abnormalities also have been noted in MF/SS:27 decreased cell-mediated cytotoxicity, decreased natural killer cell and lymphokine-activated killer cell activity, eosinophilia, increased levels of immunoglobulin E and A, and decreased cutaneous hypersensitivity. These underlying immunologic defects contribute to the high infection rate seen in patients with advanced MF/SS. HISTOPATHOLOGY The diagnosis of MF/SS is based on a combination of clinical and histopathologic findings. In patients with a chronic nonspecific dermatitis, generalized erythroderma, or poikiloderma suspected to have MF, skin biopsies are indicated. Characteristic histopathologic findings are infiltration of the epidermis with cytologically atypical T lymphocytes (epidermotropism) and the presence of large hyperchromatic “mycosis cells” within the dermis and epidermis.30 Clusters of these cells in the epidermis are Figure 15–5. Hyperkeratosis of soles in a patient with Sézary syndrome. Clinical manifestations include erythroderma, extensive pruritis, and hyperkeratosis and cracking of palms and soles. 264 MALIGNANT LYMPHOMAS known as Pautrier’s microabscesses, whereas in the upper dermis these cells are present in bandlike infiltrates. In the early stages of MF, the histology may be indistinguishable from benign or borderline conditions, such as parapsoriasis. If clinical suspicion of MF cannot be corroborated by biopsy, follow-up skin biopsies are warranted. In the tumor stage of MF, the epidermis may be ulcerated due to extensive infiltration. Often these tumors clinically resemble lesions of peripheral Tcell lymphoma. However, in the latter, the epidermis often is spared, and the infiltrate is confined to the deep dermis. Although Pautrier’s microabscesses may be present in the epidermis in SS, more commonly, the upper dermis shows a dense infiltrate of lymphocytes, histiocytes, and Sézary cells.31 Immunophenotypic analysis of Sézary cells demonstrates that they express pan-T-cell markers CD2, CD3, and CD5 but may demonstrate loss of CD7 in two-thirds of cases.32–34 The majority of Sézary cells are memory cells (CD45RO+) and also express the skin-homing protein CLA. Clonality has been demonstrated for the T-cell receptor gene, but this may not be diagnostic in early-stage lesions, where the number of infiltrating cells may be small.35,36 Monoclonal T-cell receptor rearrangements may be demonstrated in cases of large plaque parapsoriasis as well as in lymphomatoid papulosis and are not therefore diagnostic of MF/SS.37 PROGNOSIS AND STAGING The staging system for MF/SS is based on the type and extent of skin involvement in the form of plaques, tumors, or erythroderma, the presence of palpable lymph node involvement, and visceral disease (Table 15–4). Lymphadenopathy is present in approximately 47 percent of all patients, with the highest frequency occurring in tumor-state MF, and in 80 to 90 percent of patients with erythroderma. Visceral involvement is found most frequently in the liver and bone marrow. At autopsy, pulmonary involvement and bone involvement are found in 60 percent and 40 percent of cases, respectively. Skin stage is the most important prognostic factor. A recent longitudinal study of over 450 patients with MF demonstrated that median survival among Table 15–4. CUTANEOUS T-CELL LYMPHOMA TNM STAGING Stage Characteristics Early IA IB IIA < 10% BSA patch or plaque (T1) ≥ 10% BSA patch or plaque (T2) T1–2, palpable adenopathy (node biopsy negative) Intermediate IIB III IVA Advanced IVB Cutaneous tumors (T3) Erythroderma (T4) T1–4, node biopsy positive T1–4, visceral involvement TNM = tumor staging according to primary tumor (T), regional nodes (N), and metastasis (M); BSA = body surface area. patients with limited skin plaques is the same as that of age-matched controls (Table 15–5).38 A recent analysis of published epidemiologic data on MF concluded that 90 percent of patients with 10 percent or less of skin involvement survive for 15 years or more.39 In a study of 122 patients with limited cutaneous involvement, only 2 percent of patients had died after 32 years, and 9 percent showed disease progression.40 A multivariate analysis of data from 309 Dutch patients with MF found that the 5year disease-specific survival rates for individuals with limited skin involvement and skin tumors were 100 percent and 80 percent, respectively, but that the rate for individuals with lymph node involvement dropped to 40 percent.41 Older data indicate that the median survival for patients with visceral involvement is 24 to 30 months. Five-year survival for patients with SS is generally reported as 10 percent. In a multivariate analysis of 51 patients with SS, the overall 5-year survival was 33.5 percent. When patients were stratified for risk based on three variables found to be adverse Table 15–5. CUTANEOUS T-CELL LYMPHOMA PROGNOSIS BY SKIN STAGE Skin Stage at Diagnosis 10-Year Relative Survival (%)* p Value T1 T2 T3 T4 100 67 39 41 NS .002 < .001 < .001 *Observed ÷ expected survival) × 100, in controls matched for age, sex, and race. Reprinted with permission from Zackheim HS et al. J Am Acad Dermatol 1999;40:418. Cutaneous T-Cell Lymphoma independent prognostic indicators (PAS-positive cytoplasmic inclusions in circulating Sézary cells, CD7- phenotype, and the presence of large circulating Sézary cells), those with zero or one of the indicators had a 5-year survival rate of 58 percent, whereas those with two or three indicators had a 5year survival rate of 5 percent.42 In addition to the risk for disease progression in MF/SS patients, there is a well-established risk for cytologic transformation to CD30+ large cell lymphoma,43,44 the diagnosis of which is established by the presence of at least 25 percent large cells on biopsy.45 In one series of 150 CTCL patients, the risk of transformation was 12 percent, with a median time from diagnosis to transformation of 21.5 months and median survival following transformation of 2 months.43 In another study of 115 MF/SS patients, 23 percent underwent transformation, with a median time from diagnosis to transformation of 12 months and median survival subsequent to transformation of 19 months. The investigators calculated a 39 percent cumulative probability of transformation at 12 years following initial diagnosis.44 Analysis by the French Study Group on Cutaneous Lymphomas determined that the median time from diagnosis to transformation was 6.5 years and that mean survival from transformation to death was 22 months.45 A recent review article that summarized all of the published epidemiologic literature concluded that the cumulative rate per year and per patient of transformation from MF/SS to large cell lymphoma is 8 to 23 percent, and that transformation is associated with poor prognosis evidenced by mean survival ranging from 2 to 10 months. 265 Several staging algorithms have been proposed since 1978 for establishing prognosis in individual CTCL patients. All are based on the principal risk factors for disease progression (extent of skin, nodal, and visceral involvement) and on histopathologic information from lymph node and bone marrow biopsies. In 1979, the Committee on Staging and Classification of Cutaneous T-Cell Lymphoma proposed a system in which low, intermediate, and high risks were determined by differentiating four levels of skin involvement, five levels of nodal involvement, the presence or absence of palpable adenopathy, and the presence or absence of visceral involvement.46 Subsequently, Sausville and colleagues proposed a simplified version based on a multivariate analysis that demonstrated that skin stage, histopathologic lymph node involvement, and visceral involvement were significant prognostic factors.47 Based on this classification system, three survival groups were identified (Figure 15–6): lowrisk patients with skin patches or plaques and histopathologically uninvolved lymph nodes (stages IA and IB) had an excellent prognosis, whereas patients with visceral disease (stage IVB) had a poor prognosis, irrespective of skin stage. Patients with skin tumors, histopathologically involved lymph nodes, and erythroderma (stages IIB, III, and IVA) had an intermediate prognosis, similar to that of patients with follicular B-cell lymphoma.48 Bone marrow involvement was defined further in a retrospective study in which histopathologic staging of bone marrow based on presence of lymphoid aggregates or infiltrating lymphoma was performed. Figure 15–6. Survival based on prognostic category. (Reproduced with permission from Sausville E, et al. Ann Intern Med 1988;109:372. 266 MALIGNANT LYMPHOMAS In this study, the presence of lymphoid aggregates had no independent prognostic significance, whereas diffuse lymphomatous infiltrates in the marrow were associated with an inferior outcome.49,50 CLINICAL MANAGEMENT Initial management of CTCL depends on the extent of clinical manifestations and the impact of symptoms on quality of life. For more advanced disease, the primary emphases are palliation and prevention or deceleration of disease progression.51 Most patients experience pruritus, skin pain, progressive physical limitation, sleep deprivation, and infectious complications. Furthermore, because stigmatizing skin lesions may occur in places that are not easily concealed, many patients with CTCL resort to reclusive or other antisocial behavior or may exhibit other signs of extreme psychological distress. Quality-of-life issues are matters of serious long-term concern, as is the expense of treatment that may continue for decades. Because progressive skin deterioration is characteristic of MF/SS, infection and sepsis from S. aureus, S. epidermidis, and Pseudomonas aeruginosa are the principal causes of morbidity and mortality. Chronic or recurrent infection require prolonged or repeat antimicrobial therapy. Immune deficiency inherent to CTCL also exposes patients to potentially life-threatening fungal and viral infections. Almost all patients with MF/SS require treatment directed at the skin. Systemic therapies are used in MF/SS when patients become refractory to topical therapies or when they demonstrate signs of advanced systemic disease in the form of lymphadenopathy or visceral involvement. The therapeutic approaches to MF based on clinical stage are outlined in Table 15–6. Skin-Directed Therapies Topical corticosteroids frequently achieve good responses, particularly in early-stage disease, providing symptomatic relief. Zackheim and colleagues reported the largest experience with topical steroids in 79 patients with patch/plaque MF. High-potency steroids were used in all T1 patients. With a median follow-up of 9 months, 63 percent of the T1 patients achieved complete remission and 31 percent achieved partial remission.51 Topical chemotherapy using mechlorethamine (nitrogen mustard, HN2) has proven effective for cutaneous MF since its use was first reported in 1973.52,53 It is dissolved in tap water and applied to the entire body surface. One topical HN2 trial reported a 51 percent complete response rate and an 88 percent overall response rate in 43 patients with stage I patch disease, and complete and overall response rates of 26 percent and 70 percent, respectively, in 58 patients with stage II plaque disease.53 A subsequent trial involving 41 patients with T1 disease and 76 patients with T2 disease reported complete remission rates after 1 year of 67 percent in the T1 group (median time to remission = 4.4 months) and 40 percent in the T2 group (median time to remission = 20.4 months).54 In a long-term follow-up study in which data interpretation was confounded by multiple therapies administered to patients, 11 percent of 331 patients treated with topical HN2 experienced complete remissions lasting more than 8 years.52 For patients with tumor-stage disease, topical therapy with HN2 may not be as effective and may be supplemented with local irradiation or systemic therapies. Topical chemotherapy using carmustine (BCNU) yields similar results to those with HN2. Like HN2, carmustine is mixed with water and applied daily to the full body surface except uninvolved areas of the face, hands, genitals, and skinfolds. In a trial involving 89 patients with patch-stage disease, there was an overall response rate of 92 percent after 3 years, a 5-year survival rate of 97 percent, and a median treatment time to complete response of 9 weeks. In Table 15–6. MANAGEMENT OF CUTANEOUS T-CELL LYMPHOMA BY STAGE Stage Management IA/IB Topical chemotherapy, PUVA, local EBRT, topical retinoids PUVA, topical chemotherapy, local EBRT, oral retinoids, IFN, methotrexate, ONTAK (refractory) EBRT, methotrexate, IFN, ONTAK, chemotherapy, oral retinoids Photopheresis, IFN, ONTAK, pentostatin, chemotherapy, retinoids ONTAK, pentostatin, chemotherapy, retinoids, investigational agents IIA IIB III IV PUVA = psoralen plus ultaviolet A; EBRT = electron beam radiation therapy; IFN = interferon; ONTAK = denileukin diftitox. Cutaneous T-Cell Lymphoma the same series, 83 patients with plaque-stage MF had an overall response rate of 64 percent, a 5-year survival of 79 percent, and a median treatment time to remission of 12 weeks.55 Cutaneous hypersensitivity reactions occur in approximately 30 percent of patients treated with topical HN2, although this can be reduced to approximately 5 percent by switching from the aqueous solution to an ointment-based preparation. Longterm HN2 therapy, especially in combination with multiple other skin-based therapies, has been associated with an increased risk for squamous and basal cell carcinomas.56 There is no systemic absorption of HN2. Carmustine, in contrast, does not induce cutaneous hypersensitivity reaction, but it is absorbed systemically to some degree. Secondary skin cancers are a minor risk with carmustine. Phototherapy Phototherapy using ultraviolet A (UVA) with psoralen or ultraviolet B (UVB) has been shown to be effective for treating early-stage disease.57,58 In a retrospective study of 30 patients, 25 patients (83%) achieved complete remission after a median of 5 months of UVB treatment three times per week. The median duration of remission was 22 months.57 On the basis of a small trial (N = 8), Clark and colleagues have shown that narrow-band TL-01 phototherapy may be as effective as UVB therapy and may reduce the risk of long-term adverse effects.59 Photochemotherapy, which combines UVA with a psoralen-based photosensitizing agent, is the most commonly used form of phototherapy in MF/SS patients. Known as PUVA therapy, this treatment consists of oral administration of 8-methoxypsoralen (8-MOP), which is activated by exposure to UV light in the range of 330 to 340 nm given 2 hours later. During a clearance phase of up to 6 months, treatments typically are given two or three times per week depending on skin type, severity of skin reaction, and response to treatment. Subsequently, treatments are reduced to one to three times per month, although the frequency may be increased in the event of disease recurrence during the maintenance regimen. The efficacy of this treatment for patch and plaque MF has been well established over a period of more than 20 years, with a response rate of 267 approximately 59 percent.60–63 This has been improved to as much as 80 percent with the addition of low-dose IFN.64 The long-term cure rate from treating early-stage MF patients with PUVA is 15 to 20 percent. Patients with SS may require supplementation of PUVA with systemic therapies including corticosteroids, IFN, and cytotoxic chemotherapy. Potential side effects from PUVA therapy include nausea due to psoralen ingestion, erythema, intensified pruritis, and chronic dry skin. Psoralen with UVA is associated with an increased incidence of cutaneous carcinomas, especially squamous carcinomas of the skin and male genitalia. Radiation Therapy Radiation therapy has been the most important modality for treating both early and advanced MF/SS. External beam photon irradiation is effective for controlling refractory plaque areas and reducing tumors, but the penetration of radiation by this method is associated with myelosuppression and radiation dermatitis. Electron beam therapy, in contrast, can be delivered at an energy of 4 to 9 MeV with less than 5 percent of the dose penetrating beyond 2 cm. The target volume for most patients does not exceed a depth of 5 mm.65 Hence, internal organ toxicity is uncommon. Techniques involving the scattering of electrons generated by a linear accelerator are capable of achieving total-skin electron beam therapy (TSEBT). Treatment consists of a total of 30 to 36 Gy administered three or four times per week over an 8- to 12-week period. This regimen is based on evidence that patients who receive more than 20 Gy have significantly better 5-year survival and longer complete responses than do patients treated with 20 Gy or less.66 Total-skin EBT is used most commonly in patients with diffuse plaque involvement that is refractory to other skin-directed therapies, tumorstage MF, or SS. Complete response rates to TSEBT range from 56 to 96 percent in patients with stages IA to IIA disease. Patients with stage IA disease have a relapse-free survival rate of 33 to 52 percent at 10 years, whereas the same rate for patients with stage IB or worse disease is 16 percent.67,68 Toxicities associated with TSEBT include erythema, 268 MALIGNANT LYMPHOMAS desquamation, temporary depilation, and temporary loss of fingernails, toenails, and sweat gland function. Ocular complications are possible theoretically because lead shields are not completely effective against electron beam irradiation.69 Several clinical trials have explored the efficacy of supplementing TSEBT with adjuvant therapies. In a trial in which 103 patients with early-stage MF, all of whom underwent TSEBT, were randomized to either conservative (topical) therapy or aggressive (multiagent systemic chemotherapy) treatment, patients treated aggressively had significantly higher complete response rates and significantly higher toxicities. At a median follow-up of 75 months, however, there was no significant difference between the treatment groups in disease-free or overall survival, suggesting that aggressive early treatment does not improve prognosis.70 A study in which 14 patients with T1 and T2 disease underwent PUVA therapy following TSEBT reported significant improvement in disease-free survival in the PUVA group and a trend toward improved overall survival.68 An evaluation of 44 patients with erythrodermic (T4) MF concluded that extracorporeal photopheresis (ECP) concurrent with or immediately following TSEBT significantly improves both progression-free survival and causespecific survival.71 A French study evaluated combination therapy using TSEBT for plaque-stage disease and photon beam irradiation for thick plaques, tumors, and nodal involvement in 45 patients with advanced MF. The overall response rates were 81 percent for T3 patients, 61 percent for T4 patients, 79 percent for N1 patients (63% of which were complete responses), and 70 percent for N3 patients (41%, complete responses). The 5-year overall survival rates were 37 percent for T3 patients, 44 percent for T4 patients, and 32 percent for N3 patients.72 Extracorporeal Photopheresis The use of extracorporeal photopheresis, which was first reported for treating CTCL in 1987,73 is a technique in which leukopheresis mononuclear cells are exposed to a psoralen photoactivating agent (eg, 8MOP) and ultraviolet light (UVA) ex vivo and reinfused into the patient. In essence, ECP is systemic photochemotherapy or a technique for administering PUVA systemically. Initially, patients are treated on 2 consecutive days once monthly until minimal clearance occurs; then they are tapered to monthly treatment for 6 months followed by treatment at 2-month intervals before discontinuation. More recently, therapy has been administered at 2-week intervals, and response times have shortened. The mechanism by which ECP elicits a response is not known, but the most probable hypothesis is that psoralen-photoactivated circulating tumor cells are damaged by UVA and therefore stimulate an immune-mediated antitumor response following re-infusion. This hypothesis is supported by studies that demonstrate that response to ECP is associated with the presence of circulating tumor cells and with increase in CD8+ cytotoxic Tcell populations during therapy. It is also possible that photopheresis has a direct cytotoxic or antiproliferative effect to induce apoptosis in the circulating tumor cells.74–76 Extracorporeal photophoresis has been most effective in patients with erythrodermic CTCL.77 In the largest series of erythrodermic (T4) patients reported, the overall response rate was 83 percent, with 20 percent being complete responses. Forty-one percent of patients experienced at least a 50 percent improvement in their skin disease.74 In a series of patients whose CTCL was refractory to other therapies and most of whom had SS, ECP produced a 73 percent response rate.75 However, in a retrospective study that required presence of a monoclonal T-cell population in the peripheral blood of treated patients, the median survival among patients treated with ECP was not significantly different from that of patients not treated with ECP.78 The addition of IFN or retinoids to ECP has improved response rates and response durations in some patients.79–81 In a recent pilot study, immune activation of lymphocytes and natural killer cells using ultra-low doses of interleukin-2 along with ECP has been implemented and is associated in a small group of patients with a rapid decrease in circulating tumor cells and improvement in skin disease. Ongoing studies are exploring other potential adjuvants to ECP. Systemic Therapies Cytotoxic Chemotherapy Single-agent systemic cytotoxic chemotherapy has been used to treat patients with refractory MF. Initial Cutaneous T-Cell Lymphoma therapy may consist of pulse steroids, alkylating agents, or methotrexate. Single-agent therapy can induce complete responses in up to 30 percent of patients, but remissions are typically of short duration.82 Methotrexate given orally twice weekly by intravenous infusion has been reported to have activity in up to 72 percent of patients. Low-dose pulse dexamethasone is effective in inducing symptomatic relief in patients with severe SS.83 Gemcitabine has demonstrated activity in a recent phase II trial involving 44 patients with pretreated refractory or relapsed CTCL.84 Pentostatin (deoxycoformycin), a purine nucleoside analogue, is a potent adenosine deaminase inhibitor that is highly lymphocytotoxic.85 Encouraging clinical results in patients treated with this agent for hairy cell leukemia led to interest in using it to treat CTCL as well. Subsequently, there have been multiple phase I and phase II trials using pentostatin as a single agent and in combination with antineoplastic agents (Table 15–7). Recently, Dearden and colleagues reported clinical data for 145 cases of mature B- and T-cell malignancies, covering 15 years of pentostatin treatment. They concluded that pentostatin is an effective single-agent therapy for patients with SS (62 percent response rate).86–87 A phase II trial conducted by the Leukemia Cooperative Group and the European Organization for Research and Treatment of Cancer (EORTC) demonstrated 33.4 percent complete remission and a 22.7 percent partial remission in patients treated with single-agent pentostatin for SS and refractory MF.88 Other purine analogues have demonstrated similar activity in CTCL, albeit with more significant toxicities. Fludarabine and 2-chlorodeoxyadenosine have been used as single agents in phase II studies and have demonstrated response rates ranging from 20 to 40 percent in previously treated patients. Myelosuppression generally has been more severe with these agents, and remission durations have been shorter than those with pentostatin.89–91 Prednisone and chlorambucil, alone and in combination (the Winkelmann regimen), have been proven effective in treating patients with SS. In a recent uncontrolled pilot study, 13 patients with erythrodermic cutaneous CTCL including SS were treated with a daily regimen of chlorambucil and 269 steroids. The mean duration of remissions was 16.5 months (median, 12 months). After a median followup of 27 months, 6 patients remained in complete remission and 3 had stable partial remission. Four patients had died, 2 of them from lymphoma.90 Combination systemic chemotherapy has been associated with high response rates, but responses generally are not durable. In a recent phase II evaluation of the EPOCH regimen (infusional etoposide, vincristine, and doxorubicin, bolus cyclophosphamide, and oral prednisone) in 15 patients with advanced refractory CTLC, including 6 patients with SS, the complete response rate was 27 percent, and the partial response rate was 53 percent. The nonresponders were 3 patients with visceral involvement: 2 of three patients with anaplastic large-cell morphology and 1 patient with human T-cell lymphoma virus leukemia/lymphoma. Two of 6 patients with SS had complete disappearance of circulating Sézary cells.92 To date, however, no phase III trial has demonstrated a significant survival benefit for the majority of patients from combination chemotherapy (see Table 15–7). Interferon-α Interferon-α is one of the most active systemic biologic therapies for MF/SS. In the first clinical trial in these diseases in 1984,93 20 patients with advanced disease, most heavily pretreated, had complete and partial response rates of 15 percent and 30 percent, respectively.94 Other trials have reported stagedependent complete response rates as high as 40 percent and partial response rates as high as 60 per- Table 15–7. THERAPIES AND RESPONSE DURATIONS FOR ADVANCED CUTANEOUS T-CELL LYMPHOMA Treatment Median Response Duration TSEBT + CAPO70 IFN high dose93 Fludarabine122 2-CDA123 Nipent single agent85–88 Nipent + high-dose IFN102 Fludarabine + low-dose IFN99 ONTAK106 Targretin 300 mg/m2/d113 13.7 mo 8 mo 3 mo 4 mo, 3 mo 1.3–8.3 mo 13.1 mo 6.5 mo 7.3 mo 59 wk TSEBT = total-skin electron beam therapy; IFN = interferon; CDA = chlorodeoxyadenosine; ONTAK = denileukin diftitox. 270 MALIGNANT LYMPHOMAS cent.95 Objective response rates as high as 88 percent have been reported in previously untreated patients with stage I or II disease. A study of 53 patients determined that dose escalation improves response rates in some patients who do not respond to low doses.94,96,97 The optimal induction dose is 3 million U three times per week, but doses up to 12 million U three times per week usually are tolerable. Toxicities are dose dependent and include fever, chills, myalgia, malaise, and anorexia. Very high doses may induce mild reversible cytopenias and hypothyroidism.95 Olsen and Bunn reported that the mean time to an objective response to IFN-2α was 5.4 months, but maximum response may take much longer, as is indicated by a range of 0.6 to 14.8 months.95,97 Treatment by subcutaneous or intramuscular injection often is continued in responding patients for years because relapse generally occurs when dose reduction precedes the maximum response. There is no evidence supporting the hypothesis that combining interferons improves the efficacy of IFN- 2α. Based on negative findings in trials involving other malignancies such as chronic myelogenous leukemia, the concept has been abandoned. Several efforts have been made to assess the clinical benefit of combining IFN with other established therapies for MF/SS, with inconsistent results. For example, two trials in which IFN was combined with ECP yielded uncompelling results.95,98 Combination treatment using IFN-2α and fludarabine phosphate in 35 patients with advanced or refractory MF or SS demonstrated a 51 percent overall response rate. Three of four complete responders were in unmaintained remission after 18 to 35 months, suggesting that this treatment is efficacious in a subset of patients.99 However, in three trials totaling 31 patients treated with IFN-2α plus PUVA, all but 1 patient responded, and 22 patients had complete responses.100,101 In general, patients enrolled in these three studies were at earlier stages of disease and less heavily pretreated than were patients in singleagent IFN-α studies.95 In 1992, Foss and colleagues reported results of a phase II study that supported the activity of a regimen in which IFN-2α and pentostatin were alternated in patients with advanced or refractory MF or SS, except in patients with visceral involvement.102 In this study, the response rate was 41 percent, the median response duration was 13 months, and several patients remained in unmaintained response for over 8 years. Fusion Toxins Denileukin diftitox (DAB389IL-2, ONTAK) is a fusion toxin that combines the active moiety of diphtheria toxin and the full-length sequence of the IL-2 gene. This chimeric protein specifically binds to cells bearing high-affinity IL-2 receptors, thus sparing normal cells. The fusion toxin enters the cell by receptor-mediated endocytosis, and the active fragment of diphtheria toxin is liberated in the cytosol, where it adenosine diphosphate (ADP)-ribosylates elongation factor 2, thus inhibiting protein synthesis. Cytotoxicity by denileukin diftitox is predicated on the presence of high-affinity IL-2 receptors on neoplastic T lymphocytes. Immunohistochemical analysis of > 250 skin biopsies from CTCL patients demonstrates that IL-2 receptor expression, as measured by CD25 immunostaining, is present in 50 to 60 percent of cases.103,104 In phase I trials of denileukin diftitox in patients with IL-2 receptor expressing hematologic malignancies, 35 CTCL patients were enrolled, all of whom had refractory disease. Thirty-seven percent of patients had objective responses, and 5 patients (14%) had complete responses. The complete responses occurred in patients with extensive erythroderma and tumor-stage MF.105 A subsequent phase III trial of denileukin diftitox enrolled 71 patients with biopsy-proven CTCL that expressed CD25 on at least 20 percent of lymphocytes. Patients were assigned to one of two dose levels (9 or 18 µg/kg/d) of denileukin diftitox administered for 5 consecutive days every 3 weeks for up to eight cycles. Twenty percent of patients had partial responses, and 10 percent experienced complete responses. The response rate and duration of response (median, 6.9 months; range, 2.7 to 46.1 months) did not differ significantly by dose. Flulike symptoms and acute infusion-related events, including hypotension, dyspnea, and chest and back pain, all were unrelated to dose, as were capillary leak syndrome, transient elevation of hepatic transaminase levels (61% of patients with 17 percent grades 3 and 4), and hypoalbumine- Cutaneous T-Cell Lymphoma mia (79% of patients with 15 percent grades 3 and 4). There was no evidence of cumulative toxicity at either dose.106 It remains to be determined if denileukin diftitox has a role in the treatment of early-stage CTCL. Steroid premedication has been shown to reduce the frequency and severity of hypersensitivity reaction and capillary leak in a study of 20 patients with CTCL treated with denileukin diftitox.107 In this study, the overall response rate was 60 percent, significantly higher than that observed in the prior phase III study. Studies are under way to attempt to enhance the response to denileukin diftitox by immunomodulating the expression of high-affinity IL-2 receptor on the tumor cells. A number of biologic agents including IL-2 and retinoids have been shown to upregulate IL-2 receptor components and sensitize cells to killing by denileukin diftitox.108 Retinoids The retinoids have been used for CTCL since the 1980s. 13-cis retinoic acid has been reported as inducing overall response rates of 44 percent in 25 patients with at least T2 disease109 and 68 percent in a study of 28 patients with MF/SS.110 In a Mexican study in which the antipsoriatic retinoid etretinate was combined with IFN-2a, 10 of 12 patients with heavily pretreated refractory CTCL achieved complete responses after 1 year. After a median followup of 5 years, 7 patients (58%) remained in complete remission.111 Retinoids have been combined with other therapies such as PUVA, IFN, and TSEBT, but such combinations have not been found to be significantly better than IFN or TSEBT alone. Patients on retinoids plus PUVA require fewer treatments and a significantly lower dose of UVA than do patients receiving PUVA alone. Whereas 9-cis retinoic acid, 13-cis retinoic acid, and all-trans retinoic acid act by binding to intracellular retinoid A receptors (RARs) that regulate transcriptional activity of target genes, a family of novel retinoids targets the the retinoid “X” receptors (RXRs).112 Retinoid X receptors can form heterodimers with other receptors such as RARs, vitamin D receptor, thyroid receptor, and peroxisome proliferator activator receptors (PPARs). Bexarotene, an RXR ligand that selectively binds and activates 271 retinoid X receptor subtypes (RXRα, RXRβ, RXRγ), has demonstrated activity both orally and topically in the treatment of CTCL. The exact mechanism of action of bexarotene is unknown, but it has been shown to induce apoptosis in selected epithelial tumor cell lines. To date, two phase II–III trials have reported activity of bexarotene in CTCL. The first trial was a randomized open-label multicenter trial to evaluate the safety and efficacy of bexarotene in 58 patients with histologically confirmed early-stage (TNM IA, IB, and IIA) CTCL who were refractory to prior therapy.113 Doses ranged from 6.5 to 650 mg per m2 per day of oral bexarotene. A dose relationship was seen in the overall response rates and in the time to disease progression (30 weeks at 300 mg/m2/d and 74 weeks at > 300 mg/m2/d). In the second trial, 94 patients with advancedstage (TNM IIB–IVB) CTCL were treated at doses ranging from 300 to 650 mg per m2 per d.114,115 A dose relationship was again seen in the overall response rate, complete clinical response, median time to response, and time to progression. Significant responses were reported in patients with Sézary syndrome, with > 70 percent clearing of skin by week 12. Adverse events included hyperlipidemia, hypercholesterolemia, and hypothyroidism, requiring dose reductions in most patients. Bexarotene Gel A phase I–II clinical trial program of bexarotene (Targretin) gel 1 percent was conducted in 67 patients with early-stage CTCL.116 Clinical response, defined as ≥ 50 percent clearing of cutaneous lesions without requirement for biopsy confirmation, was documented in 63 percent (42 of 67) of the study subjects. Twenty-one percent achieved complete resolution of their lesions. The median time to response (eg, ≥ 50 percent improvement) was 15 months. The median duration of treatment was 10 months, with a maximum duration of 55 weeks. A subsequent phase III trial was conducted in 50 patients with refractory or persistent early-stage CTCL. All patients had failed at least two prior CTCL treatments, and most (68%) were either refractory to two or more prior therapies or were refractory to one therapy and intolerant to at least 272 MALIGNANT LYMPHOMAS one therapy. The overall response rate (complete and partial remissions) was 26 percent (13 of 50) and 28 percent (13 of 47) in stage IA and IB patients, respectively, and the time from onset of therapy to response ranged from 36 to 154 days. Among responding subjects, 23 percent (3 of 13) experienced a relapse of the disease, with a median time to relapse of 149 days (range, 56 to 342 days). Toxicities included rash in 56 percent and pruritis in 18 percent of patients. ative therapies, a number of systemic treatments are available. However, response durations are short for most therapies, and no survival benefit has been established for any treatment modality. A better understanding of the pathophysiology of the disease has directed development of novel therapeutic approaches including immunomodulatory cytokine therapies and targeted cytotoxics. Vaccine strategies are under development, as are nonmyeloablative transplant regimens, to establish a platform for longterm adoptive immunotherapy. Other Novel Therapies Several experimental treatment regimens for CTCL remain under investigation. Among these are immunomodulatory cytokine therapies. Continuous infusion of high-dose IL-2 was associated with a 67 percent response rate in 6 MF/SS patients. Subsequent studies using low-dose IL-2 regimens have demonstrated activity and are ongoing.117 Interleukin-12 also has demonstrated activity in patients with early-stage MF, with a 50-percent objective response rate reported in a phase II study.118 The benefit of IL-12 use appears to be its potent stimulation of IFN-γ.118 In a phase II trial using IFN-γ, 31 percent of CTCL patients experienced objective responses.119 CAMPATH-1H, an anti-CD52 monoclonal antibody, has demonstrated activity in a number of lymphomas expressing CD52. In one pilot study, 2 of 8 patients with MF had complete responses, and 2 others had partial responses.120 Another targeted therapy, 90yttrium-T101, a radioimmunoconjugate, demonstrated activity in 3 of 8 patients with CTCL who had partial responses.121 Other T cell targeted therapies that have demonstrated activity in CTCL include anti-CD3 antibodies and both unmodified and 90yttrium anti-CD25 antibody. CONCLUSION Because CTCL is a relatively indolent disease in its early stages, the goals of early treatment are symptom relief, with minimum discomfort and inconvenience for patients, and with consideration for their quality of life. Efforts should be made to prevent or slow disease progression. When the disease becomes more advanced or is refractory to conserv- REFERENCES 1. Siegel RS, Pandolfino T, Guitart J, et al. Primary cutaneous T-cell lymphoma: review and current concepts. J Clin Oncol 2000;18:2908–25. 2. Weinstock MA, Horm JW. Mycosis fungoides in the United States: increasing evidence and descriptive epidemiology. JAMA 1988;260:42–6. 3. Weinstock MA, Gardstein B. Twenty-year trends in the reported incidence of mycosis fungoides and associated mortality. Am J Public Health 1999;89:1240–4. 4. Weinstock MA, Reynes JF. The changing survival of patients with mycosis fungoides: a population-based assessment of trends in the United States. Cancer 1999;85:208–12. 5. Kim YH, Hoppe RT. Mycosis fungoides and the Sézary syndrome. Semin Oncol 1999;26:276–89. 6. Kim YH, Jensen RA, Watanabe GL, et al. Clinical stage IA (limited patch and plaque) mycosis fungoides. A long-term outcome analysis. Arch Dermatol 1996;132:1309–13. 7. Whittemore AS, Holly EA, Lee IM, et al. Mycosis fungoides in relation to environmental exposures and immune response: a case-control study. J Natl Cancer Inst 1989;81:1560–7. 8. Tuyp E, Burgoyne A, Aitchison T, et al. A case-control study of possible causative factors in mycosis fungoides. Arch Dermatol 1987;123:196–200. 9. Jackow CM, Cather JC, Hearne V, et al. Association of erythrodermic cutaneous T-cell lymphoma, superantigenpositive Staphylococcus aureus and oligoclonal T-cell receptor V-beta gene expression. Blood 1997;89:32–40. 10. Bunn P, Schechter G, Blayner D, et al. Clinical course of retrovirus-associated adult T-cell lymphoma. N Engl J Med 1983;309:257–64. 11. Hall W, Liu C, Schneewind O, et al. Deleted HTLV-1 provirus in blood and cutaneous lesions of patients with mycosis fungoides. Science 1991;253:317–20. 12. Whitaker S, Ng Y, Rustin M, et al. HTLV-1-associated cutaneous disease: a clinicopathologic and molecular study of patients from the UK. Br J Dermatol 1993;128:483–92. 13. Bunn PA, Foss FM. T-cell lymphoma cell lines (HUT102 and HUT78) established at the National Cancer Institute: history and importance to understanding the biology, clinical features, and therapy of cutaneous T-cell lymphomas (CTCL) and adult T-cell leukemia-lymphomas (ATLL). J Cell Biochem Suppl 1996;24:12–23. 14. Manzari V, Gismondi A, Barillari G, et al. HTLV-1: a new Cutaneous T-Cell Lymphoma 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. human retrovirus isolated in Tac-negative T-cell lymphoma/leukemia. Science 1987;238:1591–2. Demierre M, Halachmi S. Mycosis fungoides in the work place. Clin Occup Environ Med 2001. [In Press] Dalloul A, Laroche L, Bagot M, et al. Interleukin-7 is a growth factor for Sézary lymphoma cells. J Clin Invest 1992;90:1054–60. Foss FM, Koc Y, Stetler-Stevenson MA, et al. Costimulation of cutaneous T-cell lymphoma cells by interleukin-7 and interleukin-2: potential autocrine or paracrine effectors in the Sézary syndrome. J Clin Oncol 1994;12:326–35. Rich BE, Campos-Torres J, Tepper RI, et al. Cutaneous lymphoproliferation and lymphomas on interleukin-7 transgenic mice. J Exp Med 1993;177:305–16. Shapiro PE, Warburton D, Berger CL, Edelson RL. Clonal chromosomal abnornalities in cutaneous T-cell lymphoma. Cancer Genet Cytogenet 1987;28:267–76. Thangavelu M, Finn WG, Yelevarthi KK, et al. Recurring structural chromosome abnormalities in peripheral blood lymphocytes of patients with mycosis fungoides/Sézary syndrome. Blood 1997;89:3371–7. Wu K, Lund M, Bang K, et al. Telomerase activity and telomere length in lymphocytes from patients with cutaneous T-cell lymphoma. Cancer 1999;86:1056–63. Buys CH. Telomeres, telomerase, and cancer. N Engl J Med 2000;342:1282–3. Hahn WC, Counter CM, Lundberg S, et al. Creation of human tumor cells with defined genetic elements. Nature 1999;400:464–8. Hahn, WC, Stewart SA, Brooks MW, et al. Inhibition of telomerase limits the growth of human cancer cell. Nat Med 1999;5:1164–70. Zoi-Toli O, Vermeer MH, De Vries E, et al. Expression of Fas and Fas-ligand in primary cutaneous T-cell lymphoma (CTLC): association between lack of Fas expression and aggressive types of CTCL. Br J Dermatol 2000;143:313–9. Navas IC, Ortiz-Romero PL, Villuendas R, et al. P16(INK4a) gene alterations are frequent lesions of mycosis fungoides. Am J Pathol 2000;156:1565–72. Rook AH, Heald P. The immunopathogenesis of cutaneous Tcell lymphoma. Hematol Oncol Clin North Am 1995;9:997–1010. Tensen CP, Vermeer MH, vander Stoop PM, et al. Epidermal interferon-γ inducible protein-10 (IP10) and monokine induced by γ interferon (Mig) but not IL-8 mRNA expression is associated with epidermotropism in cutaneous Tcell lymphomas. J Invest Dermatol 1998;111:222–6. Daliani D, Ulmer RA, Jackow C, et al. Tumor necrosis factorα and interferon-γ, but not HTLV-1 tax are likely factors in the epidermotropism of cutaneous T-cell lymphoma via induction of interferon-inducible protein-10. Leuk Lymphoma 1998;29:315–28. Burg G, Drummer R, Dommann S, et al. Pathology of cutaneous T-cell lymphoma. Oncol Clin North Am 1995;9: 961–95. Buechner SA, Winkelmann RK. Sézary syndrome: a clinicopathologic study of 39 cases. Arch Dermatol 1983;119: 979–86. Harris NL, Jaffe ES, Stein H, et al. A revised EuropeanAmerican classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood 1994;84:1361–92. Michie SA, Abel EA, Hoppe RT, et al. Expression of T-cell 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 273 receptor antigens in mycosis fungoides and inflammatory skin lesions. J Invest Dermatol 1989;93:116–20. Wood WG, Weiss LM, Warneke RA, et al. The immunopathology of cutaneous lymphomas: immunophenotypic and immunogenotypic characteristics. Semin Dermatol 1986;5:334–7. Wood GS, Tung RM, Haeffner AC, et al. Detection of clonal T-cell receptor gamma gene rearrangements in early mycosis fungoides/Sézary syndrome by polymerase chain reaction and denaturing gradient gel electrophoresis (PCR/DGGE). J Invest Dermatol 1994;103:34–41. Wood GS, Liao S, Crooks CF, et al. Cutaneous lymphoid infiltrates: analysis by polymerase chain reaction and denaturing gradient gel electrophoresis (PCR/DGGE). J Invest Dermatol 1992;98:553–6. Wood GS. The benign and malignant cutaneous lymphoproliferative disease including mycosis fungoides. In: Knowles GM, editor. Neoplastic hematopathology. Baltimore: Williams and Wilkins: 1992. p. 917. Zackheim HS, Amin S, Kashani-Sabet M, McMillan A. Prognosis in cutaneous T-cell lymphoma by skin stage: long term survival in 489 patients. J Am Acad Dermatol 1999;40:418–25. Morales Suarez-Varela MM, Llopis Gonzalez A, Marquina Vila A, Bell J. Mycosis fungoides: review of epidemiological observations. Dermatology 2000;201:21–8. Kim YH, Jensen RA, Watanabe GL, et al. Clinical stage 1A (limited patch and plaque) mycosis fungoides: a long-term outcome analysis. Arch Dermatol 1996;132:1309–13. Van Doorn R, van Haselen CW, van Voorst Vader PC, et al. Mycosis fungoides: disease evolution and prognosis of 309 Dutch patients. Arch Dermatol 2000;136:504–10. Bernengo MG, Quaglino P, Novelli M, et al. Prognostic factors in Sézary syndrome: a multivariate analysis of clinical, hematological and immunologic factors. Ann Oncol 1998;9:857–63. Dmitrovsky E, Matthews MJ, Bunn PA, et al. Cytologic transformation in cutaneous T-cell lymphoma: a clinicopathologic entity associated with poor prognosis. J Clin Oncol 1987;5:208–15. Diamandidou E, Colome-Grimmer M, Fayad L, et al. Transformation of mycosis fungoides/Sézary syndrome: clinical characteristics and prognosis. Blood 1998;92:1150–9. Vergier B, de Muret A, Beylot-Barry, M, et al. Transformation of mycosis fungoides: clinicopathological and prognostic features of 45 cases. French Study Group of Cutaneous Lymphomas. Blood 2000;95:2212–8. Bunn P, Lamberg S. Report of the Committee on Staging and Classification of Cutaneous T-Cell Lymphoma. Cancer Treat Rep 1979;63:725–8. Sausville E, Worsham G, Matthews M, et al. Histologic assessment of lymph nodes in mycosis fungoides/Sézary syndrome: clinical correlations and prognostic importance of a new classification system. Hum Pathol 1985;16:1098–109. Sausville E, et al. Ann Intern Med 1988:109:372. Graham SJ, Sharpe RW, Steinberg SM, et al. Prognostic implications of a bone marrow histopatholigic classification system in mycosis fungoides and the Sézary syndrome. Cancer 1993;72:726–34. Foss FM, Sausville EA. Prognosis and staging of cutaneous T-cell lymphoma. Hematol Oncol Clin North Am 1995;9:1017. 274 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. MALIGNANT LYMPHOMAS Zackheim HS, Kashani-Sabet M, Amin S. Topical corticosteroids for mycosis fungoides: experience in 79 patients. Arch Dermatol 1998;134:949–54. Vonderheid E, Tan E, Kantor A, et al. Long-term efficacy, curative potential, and carcinogenicity of topical mechlorethamine chemotherapy in cutaneous T-cell lymphoma. J Acad Dermatol 1989;20:416–28. Hoppe RT, Abel EA, Deneau DG, et al. Mycosis fungoides: management with topical nitrogen mustard. J Clin Oncol 1987;5:1796–803. Ramsey DL, Ed M, Halperin PS, et al. Topical mechlorethamine therapy for early stage mycosis fungoides. J Am Acad Dermatol 1988;19:684–91. Zackheim HS. Topical carmustine (BCNU) for patch/plaque mycosis fungoides. Semin Dermatol 1994;13:202–6. Abel E, Sendagorta E, Hoppe R. Cutaneous malignancies and metastatic squamous cell carcinoma following local therapy for mycosis fungoides. J Am Acad Dermatol 1986;14:1029–38. Ramsey DL, Lish KM, Yalowitz CB, et al. Ultraviolet-B phototherapy for early-stage cutaneous T-cell lymphoma. Arch Dermatol 1992;128:931–3. Resnik K, Vonderheid E. Home UV phototherapy of early mycosis fungoides: long-term follow-up observations in thirty-one patients. J Am Acad Dermatol 1993;29:73–7. Clark C, Dawe RS, Evans AT, et al. Narrowband TL-01 phototherapy for patch-stage mycosis fungoides. Arch Dermatol 2000;136:748–52. Molin L, Thomas K, Volden K, et al. Photochemotherapy (PUVA) in the pretumor stage of mycosis fungoides: a report from the Scandinavian Mycosis Fungoides Study Group. Acta Derm Venereol 1980;61:47–51. Lowe NJ, Cripps DJ, Dufton PA, et al. Photochemotherapy for mycosis fungoides: a clinical and histological study. Arch Dermatol 1979;115:50–3. Vella-Briffa D, Warin AP, Harrington CI, et al. Photochemotherapy in mycosis fungoides: a study of 73 patients. Lancet 1980;2:49–53. Honigsmann H, Brenner W, Rauschmeier W, et al. Photochemotherapy for cutaneous T-cell lymphoma. J Am Acad Dermatol 1984;10:238–45. Kuzel TM, Gilyon K, Springer E, et al. Interferon alfa-2a combined with phototherapy in the treatment of cutaneous T-cell lymphoma. J Natl Cancer Inst 1990;82:203–7. Jones GW, Hoppe RT, Glatstein E. Electronic beam treatment for cutaneous T-cell lymphoma. Hematol Oncol Clin North Am 1995;9:1057–76. Reddy S, Parker CM, Shidnia H, et al. Total skin electron beam radiation therapy for mycosis fungoides. Am J Clin Oncol 1992;15:119–24. Hoppe R, Cox RS, Fuks Z, et al. Electron-beam therapy for mycosis fungoides. The Stanford University Experience. Cancer Treat Rep 1979;63:691–700. Quiros PA, Ones GW, Kacinski BM, et al. Total skin electron beam therapy followed by adjuvant psoralen/ultraviolet-A light in the management of patients with T1 and T2 cutaneous T-cell lymphoma. Int J Radiat Oncol Biol Phys 1997;38:1027–35. Amdur RJ, Kalbaugh KJ, Ewald LM, et al. Radiation therapy for skin cancer near the eye: kilovoltage x-rays versus electrons. Int J Radiat Oncol Biol Phys 1992;23:769–79. Kaye FJ, Bunn PA Jr, Steinberg SM, et al. A randomized trial comparing combination electron-beam radiation and 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. chemotherapy with topical therapy in the initial treatment of mycosis fungoides. N Engl J Med 1989;321:1784–90. Wilson LD, Jones GW, Kim D, et al. Experience with total skin electron beam therapy in combination with extracorporeal photopheresis in the management of patients with erythrodermic (T4) mycosis fungoides. J Am Acad Dermatol 2000;43:54–60. Maingon P, Truc G, Dalac S, et al. Radiotherapy of advanced mycosis fungoides: indications and results of total skin electron beam and photon beam irradiation. Radiother Oncol 2000;54:73–8. Edelson R, Berger C, Gasparro F, et al. Treatment of cutaneous T-cell lymphoma by extracorporeal photochemotherapy. N Engl J Med 1987;316:297–303. Heald P, Rook A, Perez M, et al. Treatment of erythrodermic cutaneous T-cell lymphoma with extracorporeal photochemotherapy. J Am Acad Dermatol 1992;27:427–33. Edelson R, Heald P, Perez M, et al. Photopheresis update. Prog Dermatol 1991;25:1–6. Marks D, Rockman S, Oziemski M, et al. Mechanisms of lymphocytotoxicity induced by extracorporeal photochemotherapy for cutaneous T-cell lymphoma. J Clin Invest 1990;86:2080–5. Lim HW, Edelson RL. Photopheresis for the treatment of cutaneous T-cell lymphoma. Hematol Oncol Clin North Am 1995;9:1117–26. Fraser-Andrews E, Seed P, Whitaker S, et al. Extracorporeal photopheresis in Sézary syndrome: no significant effect in the survival of 44 patients with a peripheral blood T-cell clone. Arch Dermatol 1998;134:1001–5. Bisaccia E, Gonzalez J, Palangio, et al. Extracorporeal photochemotherapy alone or with adjuvant therapy in the treatment of cutaneous T-cell lymphoma: a 9-year retrospective study at a single institution. J Am Acad Dermatol 2000;43:263–71. Wollina U, Liebold K, Kaatz M, et al. Survival of patients with cutaneous T-cell lymphoma after treatment with extracorporeal photochemotherapy. Oncol Rep 2000;7:1197–201. Knobler R. Extracorporeal photochemotherapy—present and future. Vox Sang 2000;78 Suppl:197–201. Rosen ST, Foss FM. Chemotherapy for mycosis fungoides and the Sézary syndrome. Hematol Oncol Clin North Am 1995;9:1109–16. Zackheim HS, Epstein EH. Low-dose methotrexate for the Sézary syndrome. J Am Acad Dermatol 1989;21:757–62. Zinzani PL, Baliva G, Magagnoli M, et al. Gemcitabine treatment in pretreated cutaneous T-cell lymphoma: experience in 44 patients. J Clin Oncol 2000;18:2603–6. Foss FM. Activity of pentostatin (Nipent) in cutaneous T-cell lymphoma: single-agent and combination studies. Semin Oncol 2000;27(2 Suppl 5):58–63. Dearden CE, Matutes E, Catovsky D. Clinical overview of pentostatin (Nipent) use in lymphoid malignancies. Semin Oncol 2000;27(2Suppl 5):22–6. Dearden CE, Matutes E, Catovsky D. Pentostatin treatment of cutaneous T-cell lymphoma. Oncology (Huntingt) 2000;14(6 Suppl 2):37–40. Ho AD, Suciu S, Strychmans P, et al. Pentostation (Nipent) in T-cell malignancies. Leukemia Cooperative Group and the European Organization for Research and Treatment of Cancer. Semin Oncol 2000;27(2 Suppl 5):52–7. Foss F. Combination therapy with purine analogs. Oncology 2000;14(Suppl 2):31–5. Cutaneous T-Cell Lymphoma 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. Coors EA, von den Driesch P. Treatment of erythrodermic cutaneous T-cell lymphoma with intermittent chlorambucil and fluocortolone therapy. Br J Dermatol 2000;143:127–31. Kuzel TM, Hurria A, Samuelson E, et al. Phase II trial of 2chlorodeoxyadenosine for the treatment of cutaneous Tcell lymphoma. Blood 1996;87:906–11. Akpek G, Koh HK, Bogen S, et al. Chemotherapy with etoposide, vincristine, doxorubicin, bolus cyclophosphamide, and oral prednisone in patients with refractory cutaneous T-cell lymphoma. Cancer 1999;86:1368–76. Bunn PA, Foon KA, Ihde DC, et al. Recombinant leukocyte A interferon: an active agent in advanced cutaneous T-cell lymphomas. Ann Intern Med 1984;101:484–7. Bunn PA, Ihde DC, Foon KA, et al. The role of recombinant interferon alfa-2a in the therapy of cutaneous T-cell lymphomas. Cancer 1986;57:1689–95. Olsen EA, Bunn PA. Interferon in the treatment of cutaneous T-cell lymphoma. Hematol Oncol Clin North Am 1995;9: 1089–107. Olsen EAQ, Rosen ST, Vollmer RT, et al. Interferon alfa-2a in the treatment of cutaneous T-cell lymphoma. J Am Acad Dermatol 1989;20:395–407. Mughal TI. The role of interferon alfa-2b in the management of patients with advanced cutaneous T-cell lymphoma. Eur J Cancer 1991;279 Suppl 4: 539–40. Vonderheid EC, Bigler RD, Greenberg AS, et al. Extracorporeal photopheresis and recombinant interferon alfa-2b in Sézary syndrome. Am J Clin Oncol 1994;17:255–63. Foss FM, Inhde DC, Linnoila IR, et al. Phase II trial of fludarabine phosphate and interferon alfa-2a in advanced mycosis fungoides/Sézary syndrome. J Clin Oncol 1994;12:2051–9. Mostow EN, Nechel SL, Oberhelman L, et al. Complete remissions in psoralen and UVA (PUVA)-refractory mycosis fungoides-type cutaneous T-cell lymphoma with combined interferon alfa and PUVA. Arch Dermatol 1993;129:747–52. Otte HG, Herges A, Stadler R. Kombinations Therapri mit Interferon alfa 2a and PUVA bei kutanen T-Zell Lymphomen. Hautarzt 1992;43:695–9. Foss FM, Breneman DL, Phelps RM, et al. Phase II study of pentostatin and intermittent high-dose recombinant interferon alfa-2a in advanced mycosis fungoides/Sézary syndrome. J Clin Oncol 1992;10:1907–13. Foss FM, Borkowski TA, Gilliom M, et al. Chimeric fusion protein toxin DAB486IL-2 in advanced mycosis fungoides and the Sézary syndrome: correlation of activity and interleukin-2 receptor expression in a phase II study. Blood 1994;84:1765–74. Nichols J, Foss FM, Kuzel TM, et al. Interleukin-2 fusion protein: an investigational therapy for interleukin-2 receptor expressing malignancies. Eur J Cancer 1997;33:S34–6. Saleh MN, LeMaistre CF, Kuzel TM, et al. Antitumor activity DAB389IL-2 fusion toxin in mycosis fungoides. J Am Acad Dermatol 1998;39:63–73. Olsen EA, Duvic M, Frankel A, et al. Pivotal phase III trial of two dose levels of denileukin diftitox for the treatment of cutaneous T-cell lymphoma. J Clin Oncol 2001;19:376–88. Foss FM, Bacha P, Kuzel TM. Biological correlates of acute hypersensitivity events with DAB2389IL-2 in non- 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 275 Hodgkin’s lymphoma: decreased frequency and severity with steroid premedication [abstract]. Clinical Lymphoma 2001;1(4):298–302. Gorgun G, Urbano A, Foss FM. Enhanced cytotoxicity to DAB389IL-2 (ONTAK) in human T-cell lines by retinoic acid [abstract]. American Society of Hematology, annual meeting, December 3–7, 1999. Blood 1999;94(10). Kessler JF, Jones SE, Levine N, et al. Isotretinoin and cutaneous helper T-cell lymphoma (mycosis fungoides). Arch Dermatol 1987;123:201–4. Molin L, Thomson K, Volden G, et al. 13-cis retinoic acid in mycosis fungoides. In: Saurat JH, editor. Retinoids: new trends in research and therapy. Basel (Switzerland): Karger; 1985. p. 341–4. Aviles A, Guzman R, Garcia EL, Diaz-Maqueo JC. Biological modifiers (etretinate and interferon alfa 2a) in the treatment of refractory T-cell lymphoma. Cancer Biother Radiopharm 1996;11:21–4. Boehm M, Zhang L, Badea B, et al. Synthesis and structureactivity relationships of novel retinoid X receptor-selective retinoids. J Med Chem 1994;37:2930–42. Duvic M, Martin A, Kim Y, et al. Oral bexarotene is safe and effective in a phase II–III clinical trial in refractory or persistent early stage CTCL [abstract]. Oral presentation at the 41st annual meeting and exposition of the American Society of Hematology, New Orleans, Louisiana, December 3–7, 1999. Blood 1999;94 10Suppl 1 (Pt 1) A2927:659a. Duvic M, Martin A, Kim Y, et al and the Worldwide Bexarotene Study Group. Phase II–III clinical trial of bexarotene capsules demonstrated efficacy and safety for patients with refractory or persistent early stage CTCL [abstract]. Poster presented at the 58th annual meeting of the American Academy of Dermatology, San Francisco, CA, Mar 10–15, 2000. Hymes K, Duvic M, Heald P, et al. Oral bexarotene benefits patients with refractory advanced stage CTCL [abstract]. Poster presented at the 41st annual meeting and exposition of the American Society of Hematology, New Orleans, Louisiana, December 3–7, 1999. Blood 1999;94 10 Suppl 1 (Pt 1) A425:97a. Heald P, Mehlmauer M, Martin AG, et al. The benefits of topical bexarotene in patients with refractory or persistent early stage cutaneous T-cell lymphoma: results of the phase 3 clinical trial. In press. Marolleau JP, Baccard M, Flageul B, et al. High-dose recombinant interleukin-2 in advanced cutaneous T-cell lymphoma. Arch Dermatol 1995;131:574–9. Rook AH, Kubin M, Fox FE, et al. The potential role of interleukin-12 in cutaneous T-cell lymphoma. Ann N Y Acad Sci 1996;795:310–8. Kaplan EH, Rosen ST, Norris DB, et al. Phase II study of recombinant human interferon gamma for treatment of cutaneous T-cell lymphomas. J Natl Cancer Inst 1990;82:208–12. Lundin J, Osterborg A, Brittinger G, et al. CAMPATH-1H monoclonal antibody in therapy for previously treated low-grade non-Hodgkin’s lymphomas: a phase II multicenter study. J Clin Oncol 1998;16:3257–63. Foss FM, Rubitscheck A, Mulshine JL, et al. Phase I study of pharmacokinetics of a radioimmunoconjugate, 90Y-T101, in patients with CD5-expressing leukemia and lymphoma. Clin Cancer Res 1998;4:2691–700.
© Copyright 2024