The ISCL/EORTC recommends revisions to the Mycosis Fungoides Cooperative Group classification and staging system for cutaneous T-cell lymphoma (CTCL). These revisions are made to incorporate advances related to tumor cell biology and diagnostic techniques as pertains to mycosis fungoides (MF) and Sézary syndrome (SS) since the 1979 publication of the original guidelines, to clarify certain variables that currently impede effective interinstitution and interinvestigator communication and/or the development of standardized clinical trials in MF and SS, and to provide a platform for tracking other variables of potential prognostic significance. Moreover, given the difference in prognosis and clinical characteristics of the non-MF/non-SS subtypes of cutaneous lymphoma, this revision pertains specifically to MF and SS. The evidence supporting the revisions is discussed as well as recommendations for evaluation and staging procedures based on these revisions.

When it became recognized that mycosis fungoides (MF), Sézary syndrome (SS), and other cutaneous T-cell lymphomas arising in the skin were part of a broader spectrum of cutaneous T-cell lymphoma (CTCL),1  the Mycosis Fungoides Cooperative Group (MFCG) developed a staging system for CTCL2  aimed at the specific findings in the MF/SS subtypes and based on the TNM (tumor-node-metastasis) classification advocated by the International Union Against Cancer (UICC)3  and American Joint Committee on Cancer.4  This classification and staging system was modified in conjunction with the National Cancer Institute (NCI) and the Veteran's Administration (VA) Hospital and published in 1979 (Tables 1,2).5  The MFCG system has proved to be an extremely useful tool in the management of patients with MF/SS and is the standard for the staging and classification of MF/SS patients today.

Table 1

Original Mycosis Fungoides Cooperative Group TNM classification of cutaneous T-cell lymphoma (CTCL)

ClassificationDescription
T: Skin*  
    T0 Clinically and/or histopathologically suspicious lesions 
    T1 Limited plaques, papules, or eczematous patches covering < 10% of the skin surface 
    T2 Generalized plaques, papules, or erythematous patches covering ≥ 10% or more of the skin surface 
    T3 Tumors, one or more 
    T4 Generalized erythroderma 
N: Lymph nodes  
    N0 No clinically abnormal peripheral lymph nodes; pathology negative for CTCL 
    N1 Clinically abnormal peripheral lymph nodes; pathology negative for CTCL 
    N2 No clinically abnormal peripheral lymph nodes; pathology positive for CTCL 
    N3 Clinically abnormal peripheral lymph nodes; pathology positive for CTCL 
B: Peripheral blood  
    B0 Atypical circulating cells not present (< 5%) 
    B1 Atypical circulating cells present (> 5%); record total white blood count and total lymphocyte counts, and number of atypical cells/100 lymphocytes 
M: Visceral organs  
    M0 No visceral organ involvement 
    M1 Visceral involvement (must have pathology confirmation and organ involved should be specified) 
ClassificationDescription
T: Skin*  
    T0 Clinically and/or histopathologically suspicious lesions 
    T1 Limited plaques, papules, or eczematous patches covering < 10% of the skin surface 
    T2 Generalized plaques, papules, or erythematous patches covering ≥ 10% or more of the skin surface 
    T3 Tumors, one or more 
    T4 Generalized erythroderma 
N: Lymph nodes  
    N0 No clinically abnormal peripheral lymph nodes; pathology negative for CTCL 
    N1 Clinically abnormal peripheral lymph nodes; pathology negative for CTCL 
    N2 No clinically abnormal peripheral lymph nodes; pathology positive for CTCL 
    N3 Clinically abnormal peripheral lymph nodes; pathology positive for CTCL 
B: Peripheral blood  
    B0 Atypical circulating cells not present (< 5%) 
    B1 Atypical circulating cells present (> 5%); record total white blood count and total lymphocyte counts, and number of atypical cells/100 lymphocytes 
M: Visceral organs  
    M0 No visceral organ involvement 
    M1 Visceral involvement (must have pathology confirmation and organ involved should be specified) 
*

Pathology of T1-4 is diagnostic of a CTCL. When more than 1 T exists, both are recorded and the highest is used for staging (eg, T4(3)).

Record number of sites of abnormal nodes (eg, cervical; left + right), axillary (left + right), inguinal (left + right), epitrochlear, and submandibular/submaxillary.

Table 2

Original Mycosis Fungoides Cooperative Group staging system for cutaneous T-cell lymphoma (CTCL)

Clinical stageTNM
IA 
IB 
IIA 1,2 
IIB 0,1 
III 0,1 
IVA 1-4 2,3 
IVB 1-4 0-3 
Clinical stageTNM
IA 
IB 
IIA 1,2 
IIB 0,1 
III 0,1 
IVA 1-4 2,3 
IVB 1-4 0-3 

The “B” classification does not alter clinical stage.

Since the publication of the MFCG classification and staging system, there have been steady advances in the areas of molecular biology, immunohistochemistry, and imaging as well as new data on prognostic variables in MF and SS that affect staging. In addition, it has become clear that the non-MF/non-SS subtypes of CTCL neither share the same T or N stages nor have the same prognosis as MF and SS.6,7  The purpose of revising the MFCG staging and classification system now is to incorporate these advances and new data; to exclude the non-MF/non-SS variants; to provide for identification, subsequent tracking, and validation of certain variables that appear to have prognostic importance; to provide clear definitions of certain variables necessary to carry out the staging and classification that in the current system have been open to variations in interpretation; and to incorporate blood (B) involvement, a major prognostic factor for patients with MF and SS.

To address these issues, the International Society for Cutaneous Lymphomas (ISCL), which includes key leaders of the EORTC, sponsored a series of workshops in 2002 to 2006 on the classification and staging of MF and SS, and the resulting revision to the MFCG classification and staging represents a consensus of both groups. The updated ISCL/EORTC staging and classification applies specifically and solely to MF and SS and has maintained the primary components of the MFCG system to allow for continued comparison of patient outcomes within both systems.

MF and SS represent approximately 65% of the cases of CTCL.6  Both are characterized by a monoclonal proliferation of predominantly CD4+/CD45R0+ helper T cells and the loss of mature T-cell antigens in the skin and other involved organs.6,8,9  SS is currently defined by the ISCL as a distinctive erythrodermic CTCL (albeit potentially lacking the diagnostic histologic features in the skin10 ) with hematologic evidence of leukemic involvement.11  The WHO/EORTC considers SS to be a separate entity from cases that otherwise meet the criteria for SS but have been preceded by clinically typical MF.6,9  Such latter cases have been designated as “SS preceded by MF” and also as “secondary” SS.12 

In some instances, the diagnosis of MF can be rendered with confidence on a skin biopsy specimen based on typical light microscopic changes, that is, marked epidermotropism of cytologically atypical T lymphocytes, clusters of these cells in the epidermis (Pautrier microabscesses), or a bandlike infiltrate containing abnormal lymphocytes in the upper dermis.13–16  However, a definitive histopathologic diagnosis by light microscopy alone may be difficult to make in early MF17,18  or in erythroderma in which inflammatory cells often predominate.19  The ISCL recently proposed a diagnostic algorithm for early MF (Table 3).15  No patient with clinically suspect patch- or plaque-stage disease who does not at least fulfill this algorithm nor any patient with tumor-stage disease with histologic findings only “suggestive of MF” should be entered into MF/SS databases or into therapeutic trials for MF/SS. In the case of erythrodermic CTCL, multiple skin biopsies may be necessary to establish a firm diagnosis or a definitive diagnosis may be made by blood studies and/or by biopsy of an enlarged lymph node or other tissue.19,20 

Table 3

Algorithm for the diagnosis of early MF15 

CriteriaMajor (2 points)Minor (1 point)
Clinical   
    Persistent and/or progressive patches and plaques plus Any 2 Any 1 
        (1) Non–sun-exposed location   
        (2) Size/shape variation   
        (3) Poikiloderma   
Histopathologic   
    Superficial lymphoid infiltrate plus Both Either 
        (1) Epidermotropism without spongiosis   
        (2) Lymphoid atypia*   
Molecular/biologic: clonal TCR gene rearrangement NA Present 
Immunopathologic   
        (1) CD2,3,5 less than 50% of T cells NA Any 1 
        (2) CD7 less than 10% of T cells   
        (3) Epidermal discordance from expression of CD2,3,5 or CD7 on dermal T cells   
CriteriaMajor (2 points)Minor (1 point)
Clinical   
    Persistent and/or progressive patches and plaques plus Any 2 Any 1 
        (1) Non–sun-exposed location   
        (2) Size/shape variation   
        (3) Poikiloderma   
Histopathologic   
    Superficial lymphoid infiltrate plus Both Either 
        (1) Epidermotropism without spongiosis   
        (2) Lymphoid atypia*   
Molecular/biologic: clonal TCR gene rearrangement NA Present 
Immunopathologic   
        (1) CD2,3,5 less than 50% of T cells NA Any 1 
        (2) CD7 less than 10% of T cells   
        (3) Epidermal discordance from expression of CD2,3,5 or CD7 on dermal T cells   

— indicates not applicable.

*

Lymphoid atypia is defined as cells with enlarged hyperchromatic nuclei and irregular or cerebriform nuclear contours.

Not applicable since it cannot fulfill any major criteria.

For patients presenting with tumors, it is important to differentiate tumor-stage MF from non-MF subtypes of CTCL. In classic MF, the tumor lesions generally develop in the presence of patch or plaque disease and not de novo. In the past, this latter presentation was classified as MF tumor d'emblee, but many of these cases would now, with immunophenotypic markers, likely be classified as various types of non-MF T-cell lymphoma or even B-cell lymphoma of the skin.6,7  Given the large number of neoplastic lymphocytes in tumoral lesions of CTCL, molecular analysis of tumor lesions will usually, but not invariably,21  demonstrate evidence of a T-cell clone; however, if this is lacking, chromosomal analysis22  and/or additional molecular studies to rule out a T-cell–rich B-cell lymphoma are in order. Histologic evaluation of an enlarged node can also be used to confirm the type of non-MF CTCL and/or to confirm (and stage) MF.

The original MFCG skin scoring system for CTCL (Table 1) included a T0 category for “clinically and/or histopathologically suspicious lesions.” While T0 may be a useful category for tracking disorders with malignant potential, current practice dictates that clinical staging be applied only to cases in which a diagnosis of cancer has been established. Therefore, the T0 category has been eliminated in the ISCL/EORTC updated staging and classification scheme (Table 4) that requires all staged patients have a definitive diagnosis of MF/SS and/or algorithmic diagnosis of early MF.

Table 4

ISCL/EORTC revision to the classification of mycosis fungoides and Sézary syndrome

TNMB stages
Skin  
    T1 Limited patches,* papules, and/or plaques covering < 10% of the skin surface. May further stratify into T1a (patch only) vs T1b (plaque ± patch). 
    T2 Patches, papules or plaques covering ≥ 10% of the skin surface. May further stratify into T2a (patch only) vs T2b (plaque ± patch). 
    T3 One or more tumors (≥ 1-cm diameter) 
    T4 Confluence of erythema covering ≥ 80% body surface area 
Node  
    N0 No clinically abnormal peripheral lymph nodes§; biopsy not required 
    N1 Clinically abnormal peripheral lymph nodes; histopathology Dutch grade 1 or NCI LN0-2 
        N1a Clone negative# 
        N1b Clone positive# 
    N2 Clinically abnormal peripheral lymph nodes; histopathology Dutch grade 2 or NCI LN3 
        N2a Clone negative# 
        N2b Clone positive# 
    N3 Clinically abnormal peripheral lymph nodes; histopathology Dutch grades 3-4 or NCI LN4; clone positive or negative 
    Nx Clinically abnormal peripheral lymph nodes; no histologic confirmation 
Visceral  
    M0 No visceral organ involvement 
    M1 Visceral involvement (must have pathology confirmation and organ involved should be specified) 
Blood  
    B0 Absence of significant blood involvement: ≤ 5% of peripheral blood lymphocytes are atypical (Sézary) cells 
        B0a Clone negative# 
        B0b Clone positive# 
    B1 Low blood tumor burden: > 5% of peripheral blood lymphocytes are atypical (Sézary) cells but does not meet the criteria of B2 
        B1a Clone negative# 
        B1b Clone positive# 
    B2 High blood tumor burden: ≥ 1000/μL Sézary cells with positive clone# 
TNMB stages
Skin  
    T1 Limited patches,* papules, and/or plaques covering < 10% of the skin surface. May further stratify into T1a (patch only) vs T1b (plaque ± patch). 
    T2 Patches, papules or plaques covering ≥ 10% of the skin surface. May further stratify into T2a (patch only) vs T2b (plaque ± patch). 
    T3 One or more tumors (≥ 1-cm diameter) 
    T4 Confluence of erythema covering ≥ 80% body surface area 
Node  
    N0 No clinically abnormal peripheral lymph nodes§; biopsy not required 
    N1 Clinically abnormal peripheral lymph nodes; histopathology Dutch grade 1 or NCI LN0-2 
        N1a Clone negative# 
        N1b Clone positive# 
    N2 Clinically abnormal peripheral lymph nodes; histopathology Dutch grade 2 or NCI LN3 
        N2a Clone negative# 
        N2b Clone positive# 
    N3 Clinically abnormal peripheral lymph nodes; histopathology Dutch grades 3-4 or NCI LN4; clone positive or negative 
    Nx Clinically abnormal peripheral lymph nodes; no histologic confirmation 
Visceral  
    M0 No visceral organ involvement 
    M1 Visceral involvement (must have pathology confirmation and organ involved should be specified) 
Blood  
    B0 Absence of significant blood involvement: ≤ 5% of peripheral blood lymphocytes are atypical (Sézary) cells 
        B0a Clone negative# 
        B0b Clone positive# 
    B1 Low blood tumor burden: > 5% of peripheral blood lymphocytes are atypical (Sézary) cells but does not meet the criteria of B2 
        B1a Clone negative# 
        B1b Clone positive# 
    B2 High blood tumor burden: ≥ 1000/μL Sézary cells with positive clone# 
*

For skin, patch indicates any size skin lesion without significant elevation or induration. Presence/absence of hypo- or hyperpigmentation, scale, crusting, and/or poikiloderma should be noted.

For skin, plaque indicates any size skin lesion that is elevated or indurated. Presence or absence of scale, crusting, and/or poikiloderma should be noted. Histologic features such as folliculotropism or large-cell transformation (> 25% large cells), CD30+ or CD30, and clinical features such as ulceration are important to document.

For skin, tumor indicates at least one 1-cm diameter solid or nodular lesion with evidence of depth and/or vertical growth. Note total number of lesions, total volume of lesions, largest size lesion, and region of body involved. Also note if histologic evidence of large-cell transformation has occurred. Phenotyping for CD30 is encouraged.

§

For node, abnormal peripheral lymph node(s) indicates any palpable peripheral node that on physical examination is firm, irregular, clustered, fixed or 1.5 cm or larger in diameter. Node groups examined on physical examination include cervical, supraclavicular, epitrochlear, axillary, and inguinal. Central nodes, which are not generally amenable to pathologic assessment, are not currently considered in the nodal classification unless used to establish N3 histopathologically.

For viscera, spleen and liver may be diagnosed by imaging criteria.

For blood, Sézary cells are defined as lymphocytes with hyperconvoluted cerebriform nuclei. If Sézary cells are not able to be used to determine tumor burden for B2, then one of the following modified ISCL criteria along with a positive clonal rearrangement of the TCR may be used instead: (1) expanded CD4+ or CD3+ cells with CD4/CD8 ratio of 10 or more, (2) expanded CD4+ cells with abnormal immunophenotype including loss of CD7 or CD26.

#

A T-cell clone is defined by PCR or Southern blot analysis of the T-cell receptor gene.

The definition and differentiation of patch versus plaque23  versus tumor lesions24,25  is more than of just semantic importance because both prognosis and choice of and response to treatment are linked to these different designations. Currently, the distinction between patch and thin plaque lesions and between thick plaque and tumor lesions in MF is quite subjective4,26  (personal communication, E. Vonderheid, ISCL Workshop San Francisco, 1999). The ISCL/EORTC have attempted to add some clarity to the situation by suggesting definitions for the skin lesions (Table 4). Because in patch/plaque disease, histology has been shown to offer an objective means of defining each subtype,27,28  be a validated surrogate for the clinical classification of MF lesions,29  and have prognostic implications,30  there is a provision in the classification system for characterizing exclusively patch-stage disease with the subscript of “a” (T1a and T2a) versus combined patch/plaque disease with the subscript of “b” (T1b and T2b) in order to gather additional longitudinal data on this distinction (Table 4). However, the derivation of all T stages remains a clinical determination.

In both the original MFCG5  and the revised staging system, the T1 skin rating is defined as papules, patches, and/or plaques covering less than 10% body surface area (BSA) and T2 skin rating is defined as patches and/or plaques covering 10% or more BSA. In the classification published by the MFCG in 1979, 1% BSA was defined as equal to the “palmar surface of the hand.”5  However, the area of the palm and digits together is actually slightly less than 1% BSA (∼ 0.8%),31–34  and mathematically and reliably, the palm at 0.5% BSA31,34  may be the easiest and most reliable measure to use in assigning BSA of lesions of MF or SS. Another method of determining BSA is to estimate the percentage of skin involvement in each of 12 regions of the body (each with a relative assigned percent BSA35  [Figure 1]), multiplying this number by the percentage of the BSA for that particular region and adding up the regional percentages to obtain the total BSA involved with MF/SS.

Figure 1

Regional percent body surface area (BSA) in the adult. Adapted from Lund and Browder35  with permission.

Figure 1

Regional percent body surface area (BSA) in the adult. Adapted from Lund and Browder35  with permission.

Close modal

Although ulceration, which may occur in plaques as well as tumors, is generally a poor prognostic sign,4,36,37  ulceration may be caused by infection as well as tumor necrosis and, by multivariable analysis, does not alter the prognosis once the extent of the T rating is known.38  Therefore, the ISCL/EORTC does not recommend using ulceration as the sole criterion to move a patient from plaque (T1 or T2)- to tumor (T3)-stage MF.

Whether any given number of lesions, aggregate volume, size of largest lesion, number of lesions, or specific body regions involved has any predictive prognostic value in tumor-stage MF is unknown at this time. The MFCG originally required that a diagnosis of tumor-stage disease include at least 3 tumors,26  but this was changed to one or more tumors in the final MFCG staging system.5  The proposed ISCL/EORTC classification revision retains the requirement of at least one tumor (≥ 1.5 cm in diameter) for the definition of T3. Whether there should be a minimum histologic depth of infiltrate to distinguish plaque from tumor in order to corroborate this important assignment of T stage based on a single lesion has not been yet been determined.

The skin of erythrodermic CTCL may show some degree of clinically apparent infiltration that may be caused by either dermal infiltration with tumor cells or an inflammatory reaction with or without edema. There is currently no distinction in the updated staging system for subclassifying T4 based on varying degrees of induration, erythema, or scale. Specific grading systems for erythroderma that do include these variables have been published elsewhere39,40  and may be of value for use in clinical trials.

When more than one T rating might apply, the highest is used for staging purposes. In the situations where both tumors and erythroderma exist simultaneously, both T ratings should be recorded (eg, T4(3)).5  This latter nuance of the T staging suggested by the MFCG continues to offer a way of tracking a variable that may impact on the poor prognosis of T4 disease, but would otherwise be buried in the classification hierarchy.

There are at least 2 histologic findings on skin biopsies in MF that appear to have prognostic importance but require further data before modifications to the staging system are justified. Folliculotropic MF is characterized histologically by atypical CD4+ T lymphocytes that surround and infiltrate the hair follicles (folliculotropism), usually without evidence of epidermotropism and with frequent concomitant follicular mucinosis.41–43  Clinically, folliculotropic MF is typically classified under the T1 or T2 skin rating even though the infiltrate extends histologically along the hair follicles deeper than is typical for plaque-stage disease.41,44  Folliculotropic MF has been shown to be associated with a worse prognosis than expected for clinical stage41–43,45 : the 5-year survival is similar and the 10-year survival is worse than in patients with tumor-stage MF.45  Large-cell transformation, defined as a biopsy specimen showing large cells (≥ 4 times the size of a small lymphocyte) in 25% or more of the dermal infiltrate,46–49  is a poor prognostic sign, seen most commonly in tumor-stage MF and less commonly in plaque-stage and erythrodermic MF. Based on molecular analysis, large-cell transformation in MF or SS represents evolution of the original malignant clone.50,51  These large cells may or may not be CD30+.48  The possibility that a patient with CD30+ nodules might have primary cutaneous CD30+ anaplastic large-cell lymphoma coexisting with MF must also be considered, although the coexistence of typical patches and plaques of MF would normally indicate that such lesions represent large-cell transformation (CD30+) of MF rather than a separate primary cutaneous lymphoma. The ISCL/EORTC recommends tracking folliculotropic MF and large-cell transformation to determine if either warrants an advance in stage.

Defining peripheral adenopathy

The negative impact on survival of “palpable adenopathy” in MF has long been appreciated.14,24,26,36,37,52,53  In the MFCG classification, “N” represented only peripheral lymph nodes. Although there was no size designation for “abnormal” nodes, this was not problematic as all patients, even those without palpable nodes, were to have node biopsies as part of the staging evaluation since each N rating for staging was based on both clinical and histopathologic findings. Although Sausville et al reported that 9 (32%) of 28 patients with MF or SS without adenopathy had either frank lymphoma or advanced dermatopathic findings on histopathologic review of “blind” nodal biopsies,13  biopsies of nonpalpable nodal groups are rarely done in clinical practice today.

The updated ISCL/EORTC classification eliminates biopsies of lymph nodes for staging purposes that are not enlarged on physical examination or imaging studies. In doing so, however, the size of a peripheral lymph node designated to be “clinically abnormal” takes on more importance because a lymph node biopsy is recommended only of such nodes. The ISCL/EORTC revision defines clinically abnormal peripheral nodes as those 1.5 cm or larger in the longest transverse diameter or any palpable peripheral node, regardless of size, that on physical examination is firm, irregular, clustered, or fixed. The 1.5-cm size is different from the 1-cm diameter node designated as abnormal both by the International Workshop on Response Criteria for Non-Hodgkin Lymphoma54  and the ISCL/EORTC staging for non-MF/SS primary cutaneous lymphomas55  since reactive or dermatopathic peripheral lymph nodes commonly occur in MF, SS, and benign inflammatory skin disorders56  but are uncommon in these other lymphomas involving the skin. These clinically enlarged or abnormal nodes should be corroborated by an imaging study (computed tomography [CT] ± 18F-fluorodeoxyglucose positron emission tomography [FDG-PET] or by magnetic resonance imaging (MRI) (in cases where patients may be allergic to contrast dye) or ultrasound prior to biopsy.

Pathology of lymph nodes

The original N rating did not include guidelines for the distinction between nodes judged pathologically “negative” or “positive” for CTCL nor did it give any weight to increasing magnitude of tumor involvement based on light microscopy, flow cytometry, or molecular genetic results. The 2 main histopathologic grading systems for lymph nodes in MF/SS in use today are the NCI/VA classification system,13  first proposed by Matthews and Gazdar,57  and the Dutch System58  (Table 5). The major difference between these classification systems resides in the criteria used to define “abnormal” lymphocytes.58,60  Specifically, the NCI/VA system,13  although it defines abnormal (neoplastic) cells as small (6-10 μm) or large (> 11.5 μm) cells with cerebriform, irregularly folded, hyperconvoluted nuclei (ie, Sézary cells), does not use the size of cells but instead uses the relative numbers of such cells in the paracortex of the lymph node for the LN0-2 definition (Table 5). Conversely, the Dutch system uses the diameter of the cerebriform cells (> 7.5 μm) to define abnormal (neoplastic) cells, and if present, this constitutes early involvement (grade 2).58 

Table 5

Histopathologic staging of lymph nodes in mycosis fungoides and Sézary syndrome

Updated ISCL/EORTC classificationDutch system58 NCI-VA classification13,57,59 
N1 Grade 1: dermatopathic lymphadenopathy (DL) LN0: no atypical lymphocytes 
  LN1: occasional and isolated atypical lymphocytes (not arranged in clusters) 
  LN2: many atypical lymphocytes or in 3-6 cell clusters 
N2 Grade 2: DL; early involvement by MF (presence of cerebriform nuclei > 7.5 μm) LN3: aggregates of atypical lymphocytes; nodal architecture preserved 
N3 Grade 3: partial effacement of LN architecture; many atypical cerebriform mononuclear cells (CMCs) Grade 4: complete effacement LN4: partial/complete effacement of nodal architecture by atypical lymphocytes or frankly neoplastic cells 
Updated ISCL/EORTC classificationDutch system58 NCI-VA classification13,57,59 
N1 Grade 1: dermatopathic lymphadenopathy (DL) LN0: no atypical lymphocytes 
  LN1: occasional and isolated atypical lymphocytes (not arranged in clusters) 
  LN2: many atypical lymphocytes or in 3-6 cell clusters 
N2 Grade 2: DL; early involvement by MF (presence of cerebriform nuclei > 7.5 μm) LN3: aggregates of atypical lymphocytes; nodal architecture preserved 
N3 Grade 3: partial effacement of LN architecture; many atypical cerebriform mononuclear cells (CMCs) Grade 4: complete effacement LN4: partial/complete effacement of nodal architecture by atypical lymphocytes or frankly neoplastic cells 

Prognosis in MF/SS is clearly related to partially or completely effaced nodal architecture as defined by either the NCI-VA (LN4) or Dutch (grade 3/4) grading system,60  and continues to define the N3 node rating in the updated ISCL/EORTC staging system. However, the prognostic importance of lesser degrees of involvement as defined by light microscopy alone is less clear.24,59,61,62  Some investigators advocate that the LN3 rating should be considered equivalent to LN4 for staging purposes.13  Although in the context of MF and SS, the survival of patients with an NCI/VA LN3 node rating is worse than patients with LN0-2,24,62  it must be remembered that findings comparable with LN3 have been found in nodes from patients with benign disorders56,60,63  and that the survival of patients with LN4 is worse than for those with LN3.24,60 

Prognostic significance of T-cell clonality in lymph nodes

As early as 1988, Sausville et al recognized the potential prognostic significance of a clonal rearrangement of the TCR in nodal staging.24  In 3 studies that have reported on the use of Southern blot technique, which has a 5% detection threshold to show clonal T cells in lymph nodes,62,64,65  none of the lymph nodes classified histologically as uninvolved (NCI/VA LN0-1 or Dutch grade 1) had evidence of a clone. However, 13% of LN2 nodes, 83% of LN3 nodes, and 100% of patients with LN4 nodes had clonal rearrangements in one of the studies.62  These studies indicate that Southern blot analysis is useful as an adjunct study for lymph nodes that show higher histologic grades of involvement, especially given that the prognosis of such patients with evidence of a clone was worse than patients without a clone.62 

However, the Southern blot technique has largely been replaced by the technically easier polymerase chain reaction (PCR)–based methods, with much more sensitive detection thresholds.66  Although not significant in a multivariate analysis, Fraser-Andrews et al67  have reported detection of T-ell clones by PCR in 6 of 19 N0-2 lymph nodes and Assaf et al68  in 7 of 14 dermatopathic lymph nodes in patients with MF, and in none of the lymph nodes obtained from patients with benign conditions. In a comparison of Southern blot and PCR determination of clonality of TCR rearrangement in both palpable and nonpalpable lymph nodes in patients with MF/SS, Juarez et al concurred that in a univariate analysis, both were predictive of a poor outcome but that only Southern blot analysis was predictive of a poor prognosis in a multivariate analysis that included skin stage, presence or absence of lymphadenopathy, and histologic lymph node score.69 

For staging purposes, the ISCL recommends that nodal rating still be based on histopathology until new molecular markers have been validated, and that uneffaced nodes exhibiting an NCI/VA grade LN3 or Dutch grade 2 be classified as an N2 rating with further division into 2 subgroups: N2a (clone negative) and N2b (clone positive). It is hoped that by capturing this information longitudinally, it can be determined if there is a similar prognosis of patients with N2b and N3 node ratings.

Clinical-only staging of nodes

Because a biopsy of a clinically abnormal node is not always done at initial staging, the revised ISCL/EORTC classification has added a new category, the Nx node rating, to facilitate capture of at least this clinical information.

Lymph node biopsy

The ISCL/EORTC recommends excisional biopsy as the preferred method to evaluate abnormal lymph nodes in MF/SS. In addition to routine histologic examination, a portion of the node can be processed for immunohistochemistry, flow cytometry, and/or molecular genetic or cytogenetic analysis. Since excisional lymph node biopsies put the patient at risk for sepsis, especially in erythrodermic patients whose skin is often colonized with Staphylococcus, alternative methods to obtain nodal tissue (core biopsy or fine needle aspiration [FNA]) have been suggested as potential substitutes particularly if combined with flow cytometry.70  However, these alternate methods may have inadequate or poorly representative sampling71  or incomplete concordance with excisional biopsy results,72,73  and they do not provide the histopathologic assessment of nodal architecture necessary for N staging.

In general, the largest peripheral lymph node draining an area of involved skin and/or one that shows intense uptake on an FDG-PET scan should be selected for biopsy. If there are multiple enlarged nodes, the order of preference for biopsy by location remains cervical, axillary, then inguinal5  since cervical nodes have a higher chance of showing lymphomatous involvement than other sites.74  A biopsy of multiple nodes of a single nodal group may show different LN histopathologic ratings, raising the issue of “sampling error” and whether more than one node should be sampled at the time of excision.75  However, the need to study multiple lymph nodes from one region may be obviated if ancillary studies are performed on the specimen.76 

Potential involvement of central nodes in MF or SS was not addressed in the original MFCG classification.5  Central adenopathy may be secondary to a second malignancy (especially a second lymphoma), infection, a reactive process, or MF. The ISCL/EORTC recommends that central enlarged nodes be excluded from the determination of “N” status except in cases where an excisional biopsy of a central node has proven lymphomatous (N3) involvement with MF.

Revision to M classification

Visceral involvement with MF/SS is a well-documented, independently significant prognostic factor in MF/SS.24,77  But what constitutes “visceral disease” has not been well defined. It is generally asymptomatic and therefore prone to be underdiagnosed, especially in those with more advanced skin involvement. Visceral involvement should be questioned in the absence of node or blood involvement. To be considered as having visceral disease (stage IVb), documentation of involvement by only one organ outside the skin, nodes, or blood is needed.

Splenomegaly in MF patients is generally proven to be either diffuse or nodular involvement with MF14  and is uncommon in healthy persons or in those with benign skin disease. The ISCL/EORTC considers splenomegaly as visceral disease, even without biopsy confirmation, when it is (a) unequivocally present on physical exam and (b) documented radiographically by either enlargement or multiple focal defects that are neither cystic nor vascular (a more rigorous modification of the Cotswolds meeting recommendations on definition of lymphomatous involvement of the spleen in Hodgkin lymphoma).78 

Liver disease may be suspected by physical examination, abnormal liver function tests, or radiologic tests (CT, FDG-PET, liver/spleen scan) but should be confirmed by liver biopsy.79  In agreement with the Cotswolds meeting on Hodgkin lymphoma, multiple focal hepatic defects, which are neither cystic nor vascular, on at least 2 imaging techniques may be considered indicative of tumor involvement.78 

Bone marrow biopsy confirmation of frank lymphoma is a low-yield procedure in MF unless there is evidence of blood or nodal disease24,38,47,52,61,80  Although it has been suggested that the finding of cytologic atypical lymphoid aggregates in the bone marrow of patients with MF correlates with shorter survival,24,81–83  multivariate analysis has not demonstrated the independent prognostic value of bone marrow involvement.23,80,84,85  The ISCL/EORTC recommends performance of a bone marrow biopsy in patients with MF and SS who have B2 blood involvement (as described in paragraph 4 of “Revisions to the B (blood) rating”) or unexplained hematologic abnormalities. Tracking and recording the specific cytologic findings and whether clonality of the TCR gene rearrangement is present will provide further data on whether bone marrow involvement has independent prognostic significance and should be considered as evidence of visceral disease.

If lung abnormalities or other suggestions of extracutaneous lymphomatous involvement besides splenomegaly are found on radiographic examination, pathological assessment is recommended before ascribing this to visceral involvement with MF/SS. Visceral abnormalities seen radiologically in MF could be secondary to either another unrelated cancer (second malignancies are not uncommon in MF/SS86–88 ) or to an infectious disorder and not to MF/SS.

Following the lead of Clendenning et al,89  the original MFCG classification of blood involvement in MF (B1 rating) was defined as more than 5% of the total lymphocytes that exhibited an atypical convoluted appearance by light microscopy.5  Low numbers of atypical cerebriform mononuclear cells have been reported in both benign skin conditions90–94  and in healthy donors,92,95  and can be generated in vivo by incubating lymphocytes with cellular mitogens96  or stimulating peripheral T cells via CD3 or CD2 in the presence of phorbol esters.97  Because the prognostic importance of this criterion was unclear, the B rating was not used for staging purposes.5  Later studies at the NCI and elsewhere indicated that more than 20% atypical (Sézary) lymphocytes was associated with an adverse prognosis in MF,24,98  although no formal revision to the staging classification was done because this more rigorous B1 rating was not found to be an independent prognostic variable.13,24  However, Kim et al have subsequently shown that “significant” blood involvement (> 1000 Sézary cells/mm3 and/or > 20% Sézary cells) has prognostic significance regardless of T or N rating,25  and others have corroborated that blood involvement has independent prognostic significance.61,99,100 

The assessment of blood tumor burden in CTCL based on morphologic features of the neoplastic cells alone (eg, Sézary cell counts) is subjective and prone to interobserver variability, although absolute counts of Sézary cells continue to be used in staging at centers where such counts are routinely performed.100,101  Blood flow cytometry offers an alternate objective means of identifying and quantifying these neoplastic lymphocytes in the blood. There has been an increasing awareness that neoplastic T cells in CTCL often have altered surface expression of normal markers such as CD3, CD4, CD7, and CD26.102–108  Deletion of one or more of these markers on the surface of CD4+ cells is typical of Sézary cells, but the blood of many patients with benign inflammatory dermatoses may also show CD7 deletion.107,109,110  Loss of CD26 may be a more specific phenotype for the neoplastic lymphocytes.104,108  Identification of neoplastic cells by flow cytometry is complicated, however, by the fact that all neoplastic lymphocytes may not have the same phenotypic features and several clones may be present in a given patient with SS.111  The correlation of the percentage of abnormal cells determined by flow cytometry and that by Sézary cell preparation is inexact and may offer differing results in some cases.

Clonal expansion of TCR gene rearrangement in the blood is extremely common in early-stage disease even without a significant population of morphologically or immunophenotypically abnormal cells.99,112  It is not synonymous with blood involvement by MF/SS since benign lymphoproliferative disorders113,114  and some healthy elderly volunteers115,116  may have clonal TCR gene rearrangements of the blood T cells. Using spectratyping, MF patients even at early stages demonstrate loss of their T-cell repertoire with emergence of one of more clones.117  The presence of a peripheral blood clone in MF patients, if the same as that in the skin, has been found to have prognostic significance independent of skin stage.99 

Previously, the ISCL has categorized blood involvement into prognostically significant B ratings based on the degree of involvement, that is, B0 = absence of significant blood involvement; B1 = aleukemic, low blood tumor burden; and B2 = leukemic, high blood tumor burden.11  The ISCL/EORTC has simplified and clarified the definitions of B0 to B2. B0 remains 5% or less Sézary cells. B2 is now defined as a clonal rearrangement of the TCR in the blood and either 1.0 K/μL or more Sézary cells or one of the 2 criteria outlined by the ISCL,11  that is, (1) increased CD4+ or CD3+ cells with CD4/CD8 of 10 or more or (2) increase in CD4+ cells with an abnormal phenotype (≥ 40% CD4+/CD7 or ≥ 30% CD4+/CD26 has been suggested118 ). B1 is defined as more than 5% Sézary cells but either less than 1.0 K/μL absolute Sézary cells or absence of a clonal rearrangement of the TCR or both.

The staging of MF/SS according to the TNMB system implies that an appropriate evaluation of the 4 TNMB systems has been performed. The recommended workup is detailed in Table 6.

Table 6

Recommended evaluation/initial staging of the patient with mycosis fungoides/Sézary syndrome

Complete physical examination including 
    Determination of type(s) of skin lesions 
        If only patch/plaque disease or erythroderma, then estimate percentage of body surface area involved and note any ulceration of lesions 
        If tumors are present, determine total number of lesions, aggregate volume, largest size lesion, and regions of the body involved 
    Identification of any palpable lymph node, especially those ≥ 1.5 cm in largest diameter or firm, irregular, clustered, or fixed 
    Identification of any organomegaly 
Skin biopsy 
    Most indurated area if only one biopsy 
    Immunophenotyping to include at least the following markers: CD2, CD3, CD4, CD5, CD7, CD8, and a B-cell marker such as CD20. CD30 may also be indicated in cases where lymphomatoid papulosis, anaplastic lymphoma, or large-cell transformation is considered. 
    Evaluation for clonality of TCR gene rearrangement 
Blood tests 
    CBC with manual differential, liver function tests, LDH, comprehensive chemistries 
    TCR gene rearrangement and relatedness to any clone in skin 
    Analysis for abnormal lymphocytes by either Sézary cell count with determination absolute number of Sézary cells and/or flow cytometry (including CD4+/CD7 or CD4+/CD26
Radiologic tests 
    In patients with T1N0B0 stage disease who are otherwise healthy and without complaints directed to a specific organ system, and in selected patients with T2N0B0 disease with limited skin involvement, radiologic studies may be limited to a chest X-ray or ultrasound of the peripheral nodal groups to corroborate absence of adenopathy 
    In all patients with other than presumed stage IA disease, or selected patients with limited T2 disease and the absence of adenopathy or blood involvement, CT scans of chest, abdomen, and pelvis alone ± FDG-PET scan are recommended to further evaluate any potential lymphadenopathy, visceral involvement, or abnormal laboratory tests. In patients unable to safely undergo CT scans, MRI may be substituted. 
Lymph node biopsy 
    Excisional biopsy is indicated in those patients with a node that is either ≥ 1.5 cm in diameter and/or is firm, irregular, clustered, or fixed 
    Site of biopsy 
        Preference is given to the largest lymph node draining an involved area of the skin or if FDG-PET scan data are available, the node with highest standardized uptake value (SUV). 
        If there is no additional imaging information and multiple nodes are enlarged and otherwise equal in size or consistency, the order of preference is cervical, axillary, and inguinal areas. 
    Analysis: pathologic assessment by light microscopy, flow cytometry, and TCR gene rearrangement. 
Complete physical examination including 
    Determination of type(s) of skin lesions 
        If only patch/plaque disease or erythroderma, then estimate percentage of body surface area involved and note any ulceration of lesions 
        If tumors are present, determine total number of lesions, aggregate volume, largest size lesion, and regions of the body involved 
    Identification of any palpable lymph node, especially those ≥ 1.5 cm in largest diameter or firm, irregular, clustered, or fixed 
    Identification of any organomegaly 
Skin biopsy 
    Most indurated area if only one biopsy 
    Immunophenotyping to include at least the following markers: CD2, CD3, CD4, CD5, CD7, CD8, and a B-cell marker such as CD20. CD30 may also be indicated in cases where lymphomatoid papulosis, anaplastic lymphoma, or large-cell transformation is considered. 
    Evaluation for clonality of TCR gene rearrangement 
Blood tests 
    CBC with manual differential, liver function tests, LDH, comprehensive chemistries 
    TCR gene rearrangement and relatedness to any clone in skin 
    Analysis for abnormal lymphocytes by either Sézary cell count with determination absolute number of Sézary cells and/or flow cytometry (including CD4+/CD7 or CD4+/CD26
Radiologic tests 
    In patients with T1N0B0 stage disease who are otherwise healthy and without complaints directed to a specific organ system, and in selected patients with T2N0B0 disease with limited skin involvement, radiologic studies may be limited to a chest X-ray or ultrasound of the peripheral nodal groups to corroborate absence of adenopathy 
    In all patients with other than presumed stage IA disease, or selected patients with limited T2 disease and the absence of adenopathy or blood involvement, CT scans of chest, abdomen, and pelvis alone ± FDG-PET scan are recommended to further evaluate any potential lymphadenopathy, visceral involvement, or abnormal laboratory tests. In patients unable to safely undergo CT scans, MRI may be substituted. 
Lymph node biopsy 
    Excisional biopsy is indicated in those patients with a node that is either ≥ 1.5 cm in diameter and/or is firm, irregular, clustered, or fixed 
    Site of biopsy 
        Preference is given to the largest lymph node draining an involved area of the skin or if FDG-PET scan data are available, the node with highest standardized uptake value (SUV). 
        If there is no additional imaging information and multiple nodes are enlarged and otherwise equal in size or consistency, the order of preference is cervical, axillary, and inguinal areas. 
    Analysis: pathologic assessment by light microscopy, flow cytometry, and TCR gene rearrangement. 

The updated ISCL staging classification (Table 7) takes into account the B stage and also differentiates levels of blood involvement (B1 and B2). B1 is used to separate erythrodermic patients without overt lymph node involvement (T4N0-2M0) into 2 subgroups, IIIA (T4N0-2M0B0) and IIIB (T4N0-2M0B1), which will allow determination of the prognostic significance of low blood tumor burden in the setting of erythrodermic CTCL. The ISCL blood rating of B2 is considered comparable with nodal involvement (N3 nodal rating). Stage IVA is now defined as any skin stage and either blood involvement (B2 [IVA1]) or nodal lymphoma (N3 [IVA2]) that allows for independent tracking of these 2 important prognostic indicators.

Table 7

ISCL/EORTC revision to the staging of mycosis fungoides and Sézary syndrome

TNMB
IA 0,1 
IB 0,1 
II 1,2 1,2 0,1 
IIB 0-2 0,1 
III 0-2 0,1 
IIIA 0-2 
IIIB 0-2 
IVA1 1-4 0-2 
IVA2 1-4 0-2 
IVB 1-4 0-3 0-2 
TNMB
IA 0,1 
IB 0,1 
II 1,2 1,2 0,1 
IIB 0-2 0,1 
III 0-2 0,1 
IIIA 0-2 
IIIB 0-2 
IVA1 1-4 0-2 
IVA2 1-4 0-2 
IVB 1-4 0-3 0-2 

What has not been dealt with adequately in this updated staging is the continued classification of tumor-stage MF at a stage below that of erythroderma with the data from several centers now demonstrating that the survival curves for T3 patients are similar24,25,29  or even worse than T4 patients53,52,61,84,119,120  However, it remains unclear to what degree other important prognostic factors such as lymph node or blood involvement or large-cell transformation are influencing these observations. Ideally, comparison of the survival curves of patients with T3 and T4 skin ratings who have comparable N and now B ratings should be undertaken before concluding that the hierarchy of T3 and T4 should be modified. It is also possible that erythrodermic patients with coexisting tumors may have this important prognostic variable lost in the final staging because only the highest skin T rating is used for staging purposes. For these reasons, the ISCL/EORTC have elected to retain the existing staging parameters until additional information is available.

In oncology, the stage assigned to a patient with malignancy at the initial diagnosis and workup is the primary prognostic indicator, and although the condition may go into complete or partial remission, relapse, or progress, the “clinical stage” does not change thereafter.121  In keeping with this tradition, the formal stage of a patient with MF/SS refers to the overall tumor status at initial diagnosis. However, as with other malignancies, changes in the tumor burden in patients with MF/SS often occur during the course of disease, which affects treatment choices and response to treatment. Moreover, it is important to have a means to indicate both the maximum and the current disease status at the time of enrollment into clinical trials. Therefore, the ISCL/EORTC recommends that, in addition to “clinical stage” at diagnosis of MF/SS patients, that TNMB ratings without the corresponding stage be used to indicate the maximum tumor burden and the current tumor burden. These distinctions will provide a means of communicating the initial, maximum, and current level and type of tumor burden for an individual patient.

The ISCL/EORTC recommended revisions to the MFCG classification and staging system for CTCL are made both to incorporate advances since 1979 related to tumor-cell biology and diagnostic techniques as pertains to MF and SS and to clarify certain variables that currently impede effective interinstitution and interinvestigator communication and/or the development of standardized clinical trials in MF and SS. The ISCL/EORTC recognizes that although this revision to the staging and classification of MF and SS further narrows and defines the variables involved, it does not provide a finite staging system that inherently incorporates all potential prognostic factors. There are several current factors, primarily related either to histopathology of lesions (folliculotropic MF, large-cell transformation, patch vs plaque disease) or TCR gene rearrangement in tissue and/or blood, that require further data on their prognostic importance before making formal revisions to the staging system of MF and SS related to them. The revisions to the classification outlined here provide a framework on which to gather data and facilitate validation efforts regarding these variables, all of which can be addressed in an optional fashion for any given MF or SS patient without affecting his/her overall staging. As additional clinical, genetic, or molecular information becomes available, it is anticipated that there will be further revisions to the classification and staging guidelines for MF and SS.

An Inside Blood analysis of this article appears at the front of this issue.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Contribution: E.O. wrote the draft of the paper, participated in paper preparation, and was an active participant in the meetings where revisions to the staging and classification were generated; E.V., N.P., R.W., Y.K., R.K., H.Z., M.D., T.E., S.L., G.W., R.D., A.R., P.H., M.P., M.-G.B., and S.W. participated in paper preparation and were active participants in the meetings where revisions to the staging and classification were generated; G.B. and W.S. participated in paper preparation; L.L. and F.T. were active participants in the meetings where revisions to the staging and classification were generated.

Complete lists of the active members of the International Society for Cutaneous Lymphomas (Document S1) and the European Organization of Research and Treatment of Cancer (Document S2) are available on the Blood website; see the Supplemental Appendices link at the top of the online article.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Elise A. Olsen, Box 3294 DUMC, Durham, NC 27516; e-mail: olsen001@mc.duke.edu.

1
Lutzner
 
AM
Edelson
 
R
Schein
 
P
Green
 
I
Kirkpatrick
 
C
Ahmed
 
A
Cutaneous T-cell lymphomas: the Sézary syndrome, mycosis fungoides, and related disorders.
Ann Intern Med
1975
, vol. 
83
 (pg. 
534
-
552
)
2
Lamberg
 
SI
Diamond
 
EL
Lorincz
 
AL
, et al. 
Mycosis fungoides cooperative study.
Arch Dermatol
1975
, vol. 
111
 (pg. 
457
-
459
)
3
Harmer
 
MH
TNM Classification of Malignant Tumours.
1978
3rd ed.
Geneva, Switzerland
UICC–International Union Against Cancer
4
Oliver
 
HB
Carr
 
DT
Rubin
 
P
, et al. 
American Joint Committee on Cancer: AJCC Cancer Staging Manual.
1978
1st ed.
Chicago, IL
Whiting Press
5
Bunn
 
PA
Lamberg
 
SI
Report of the committee on staging and classification of cutaneous T-cell lymphomas.
Cancer Treat Rep
1979
, vol. 
63
 (pg. 
725
-
728
)
6
Willemze
 
R
Jaffe
 
ES
Burg
 
G
, et al. 
WHO-EORTC classification for cutaneous lymphomas.
Blood
2005
, vol. 
105
 (pg. 
3768
-
3785
)
7
Burg
 
G
Jaffe
 
ES
Kempf
 
W
, et al. 
LeBoit
 
P
Burg
 
G
Weedon
 
D
Sarasin
 
A
WHO/EORTC classification of cutaneous lymphomas.
WHO Books: Tumors of the Skin
, vol. 
Vol X
 
Lyon, France
WHO IARC
(pg. 
166
-
168
)
8
Wood
 
GS
Weiss
 
LM
Warnke
 
RA
Sklar
 
J
The immunopathology of cutaneous lymphomas: immunophenotypic and immunogenotypic characteristics.
Semin Dermatol
1986
, vol. 
5
 (pg. 
334
-
345
)
9
Burg
 
G
Kempf
 
W
Cozzio
 
A
, et al. 
WHO/EORTC classification of cutaneous lymphomas 2005: histological and molecular aspects.
J Cutan Pathol
2005
, vol. 
32
 (pg. 
647
-
674
)
10
Sentis
 
HJ
Willemze
 
R
Scheffer
 
E
Histopathologic studies in Sézary syndrome and erythrodermic mycosis fungoides: a comparison with benign forms of erythroderma.
J Am Acad Dermatol
1986
, vol. 
15
 (pg. 
1217
-
1226
)
11
Vonderheid
 
EC
Bernengo
 
MG
Burg
 
G
, et al. 
Update on erythrodermic cutaneous T-cell lymphoma: report of the International Society for Cutaneous Lymphomas.
J Am Acad Dermatol
2002
, vol. 
46
 (pg. 
95
-
106
)
12
Diwan
 
AH
Prieto
 
VG
Herling
 
M
Duvic
 
M
Jones
 
D
Primary Sézary syndrome commonly shows low-grade cytologic atypia and an absence of epidermotropism.
Am J Clin Pathol
2005
, vol. 
123
 (pg. 
510
-
515
)
13
Sausville
 
EA
Worsham
 
GF
Matthews
 
MJ
, et al. 
Histologic assessment of lymph nodes in mycosis fungoides/Sézary syndrome (cutaneous T-cell lymphoma): clinical correlations and prognostic import of a new classification system.
Human Pathol
1985
, vol. 
16
 (pg. 
1098
-
1109
)
14
Rappaport
 
H
Thomas
 
LB
Mycosis fungoides: the pathology of extracutaneous involvement.
Cancer
1974
, vol. 
34
 (pg. 
1198
-
1229
)
15
Pimpinelli
 
N
Olsen
 
EA
Santucci
 
M
, et al. 
Defining early mycosis fungoides.
J Am Acad Dermatol
2005
, vol. 
53
 (pg. 
1053
-
1063
)
16
Guitart
 
J
Kennedy
 
J
Ronan
 
S
Chmiel
 
JS
Hsiegh
 
Y-C
Variakojis
 
D
Histologic criteria for the diagnosis of mycosis fungoides: proposal for a grading system to standardize pathology reporting.
J Cutan Pathol
2001
, vol. 
28
 (pg. 
174
-
183
)
17
Santucci
 
M
Biggeri
 
A
Feller
 
A
Massi
 
D
Burg
 
G
Efficacy of histologic criteria for diagnosing early mycosis fungoides: an EORTC cutaneous lymphoma study group investigation: European Organization for Research and Treatment of Cancer.
Am J Surg Pathol
2000
, vol. 
24
 (pg. 
40
-
50
)
18
Massone
 
C
Kodama
 
K
Kerl
 
H
Cerroni
 
L
Histopathologic features of early (patch) lesions of mycosis fungoides: a morphologic study on 745 biopsy specimens from 427 patients.
Am J Surg Pathol
2005
, vol. 
29
 (pg. 
550
-
560
)
19
Vonderheid
 
EC
On the diagnosis of erythrodermic cutaneous T-cell lymphoma.
J Cutan Pathol
2006
, vol. 
33
 
suppl 1
(pg. 
27
-
42
)
20
Russell-Jones
 
R
Diagnosing erythrodermic cutaneous T-cell lymphoma.
Br J Dermatol
2005
, vol. 
153
 (pg. 
1
-
5
)
21
Vega
 
F
Luthra
 
R
Medeiros
 
J
Dunmire
 
V
, et al. 
Clonal heterogeneity in mycosis fungoides and its relationship to clinical course.
Blood
2002
, vol. 
100
 (pg. 
3369
-
3373
)
22
Karenko
 
L
Sarna
 
S
Kähkönen
 
M
Ranki
 
A
Chromosomal abnormalities in relation to clinical disease in patients with cutaneous T-cell lymphoma: a 5-year follow-up study.
Br J Dermatol
2003
, vol. 
148
 (pg. 
55
-
64
)
23
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
, vol. 
40
 (pg. 
418
-
425
)
24
Sausville
 
EA
Eddy
 
JL
Makuch
 
RW
, et al. 
Histopathologic staging at initial diagnosis of mycosis fungoides and the Sézary syndrome: definition of three distinctive prognostic groups.
Ann Int Med
1988
, vol. 
109
 (pg. 
372
-
382
)
25
Kim
 
YH
Liu
 
HL
Mraz-Gernhard
 
S
Varghese
 
A
Hoppe
 
RT
Long term outcome of 525 patients with mycosis fungoides and Sézary Syndrome: clinical prognostic factors and risk for disease progression.
Arch Dermatol
2003
, vol. 
139
 (pg. 
857
-
866
)
26
Lamberg
 
SI
Green
 
SB
Byar
 
DP
, et al. 
Clinical staging for cutaneous T-cell lymphoma.
Ann Int Med
1984
, vol. 
100
 (pg. 
187
-
192
)
27
Zackheim
 
HS
Kashani-Sabet
 
M
Amin
 
S
Topical corticosteroids for mycosis fungoides: experience in 79 patients.
Arch Dermatol
1998
, vol. 
134
 (pg. 
949
-
954
)
28
LeBoit
 
PE
McCalmont
 
TH
Elder
 
D
Cutaneous lymphomas and leukemias.
Lever's Histopathology of the Skin
1977
Philadelphia, PA
Lippincott-Raven
(pg. 
805
-
846
)
29
Kashani-Sabet
 
M
McMillan
 
A
Zackheim
 
HS
A modified staging classification for cutaneous T-cell lymphoma.
J Am Acad Dermatol
2001
, vol. 
45
 (pg. 
700
-
706
)
30
Martí
 
RM
Estrach
 
T
Reverter
 
JC
Mascaró
 
JM
Prognostic clinicopathologic factors in cutaneous T-cell lymphoma.
Arch Dermatol
1991
, vol. 
127
 (pg. 
1511
-
1516
)
31
Rossiter
 
ND
Chapman
 
P
Haywood
 
IA
How big is a hand?
Burns
1996
, vol. 
22
 (pg. 
230
-
231
)
32
Amirsheybani
 
HR
Crecelius
 
GM
Timothy
 
NH
Pfeiffer
 
M
Saggers
 
GC
Manders
 
EK
The natural history of the growth of the hand: I, hand area as a percentage of body surface area.
Plast Reconstr Surg
2001
, vol. 
107
 (pg. 
726
-
733
)
33
Perry
 
RJ
Moore
 
CA
Morgan
 
BDG
Plummer
 
DL
Determining the approximate area of a burn: an inconsistency investigated and re-evaluated [letter].
Br Med J
1996
, vol. 
312
 pg. 
1338
 
34
Sheridan
 
RL
Petras
 
L
Basha
 
G
, et al. 
Planimetry study of the percent of body surface represented by the hand and palm: sizing irregular burns is more accurately done with the palm.
J Burn Care Rehabil
1995
, vol. 
16
 (pg. 
605
-
606
)
35
Lund
 
CC
Browder
 
NC
The estimation of areas of burns.
Surg Gynecol Obstet
1944
, vol. 
79
 (pg. 
352
-
358
)
36
Epstein
 
EH
Levin
 
DL
Croft
 
JD
Lutzner
 
MA
Mycosis fungoides: survival, prognostic features, response to therapy, and autopsy findings.
Medicine
1972
, vol. 
51
 (pg. 
61
-
72
)
37
Lamberg
 
SI
Green
 
SB
Byar
 
DP
, et al. 
Status report of 376 mycosis fungoides patients at four years: Mycosis Fungoides Cooperative Group.
Cancer Treat Rep
1979
, vol. 
63
 (pg. 
701
-
707
)
38
Green
 
SB
Byar
 
DP
Lamberg
 
SI
Prognostic variables in mycosis fungoides.
Cancer
1981
, vol. 
47
 (pg. 
2671
-
2677
)
39
Edelson
 
RL
Berger
 
C
Gasparro
 
F
, et al. 
Treatment of cutaneous T-cell lymphoma by extracorporeal photochemotherapy: preliminary results.
N Engl J Med
1987
, vol. 
316
 (pg. 
297
-
303
)
40
Olsen
 
E
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
, vol. 
19
 (pg. 
376
-
388
)
41
Klemke
 
C-D
Dippel
 
E
Assaf
 
C
, et al. 
Follicular mycosis fungoides.
Br J Dermatol
1999
, vol. 
141
 (pg. 
137
-
140
)
42
Bonta
 
MD
Tannous
 
ZS
Demierre
 
M-F
Gonzalez
 
E
Harris
 
NL
Duncan
 
LM
Rapidly progressing mycosis fungoides presenting as follicular mucinosis.
J Am Acad Dermatol
2000
, vol. 
43
 (pg. 
635
-
640
)
43
Gilliam
 
AC
Lessin
 
SR
Wilson
 
DM
Salhany
 
KE
Folliculotropic mycosis fungoides with large cell transformation presenting as dissecting cellulitis of the scalp.
J Cutan Pathol
1997
, vol. 
24
 (pg. 
169
-
175
)
44
Sangüeza
 
OP
Mycosis fungoides: new insights into an old problem.
Arch Dermatol
2002
, vol. 
138
 (pg. 
244
-
246
)
45
van Doorn
 
R
Scheffer
 
E
Willemze
 
R
Follicular mycosis fungoides, a distinct disease entity with or without associated follicular mucinosis.
Arch Dermatol
2002
, vol. 
138
 (pg. 
191
-
198
)
46
Diamandidou
 
E
Colome-Grimmer
 
M
Fayad
 
L
Duvic
 
M
Kurzrock
 
R
Transformation of mycosis fungoides/Sézary Syndrome: clinical characteristics and prognosis.
Blood
1998
, vol. 
92
 (pg. 
1150
-
1159
)
47
Salhany
 
KE
Cousar
 
JB
Greer
 
JP
Casey
 
TT
Fields
 
JP
Collins
 
RD
Transformation of cutaneous T cell lymphoma to large cell lymphoma: a clinicopathologic and immunologic study.
Am J Pathol
1988
, vol. 
132
 (pg. 
265
-
277
)
48
Vergier
 
B
de Muret
 
A
Beylot-Barry
 
M
, et al. 
Transformation of mycosis fungoides: clinicopathological and prognostic features of 45 cases.
Blood
2000
, vol. 
95
 (pg. 
2212
-
2218
)
49
Cerroni
 
L
Rieger
 
E
Hödl
 
S
Kerl
 
H
Clinicopathologic and immunologic features associated with transformation of mycosis fungoides to large cell lymphoma.
Am J Surg Path
1992
, vol. 
16
 (pg. 
543
-
552
)
50
Wood
 
GS
Bahler
 
DW
Hoppe
 
RT
Warnke
 
RA
Sklar
 
JL
Levy
 
R
Transformation of mycosis fungoides: T-cell receptor â gene analysis demonstrates a common clonal origin for plaque-type mycosis fungoides and CD30+ large-cell lymphoma.
J Invest Dermatol
1993
, vol. 
101
 (pg. 
296
-
300
)
51
Wolfe
 
JT
Chooback
 
L
Finn
 
DT
Jaworsky
 
C
Rook
 
AH
Lessin
 
SR
Large-cell transformation following detection of minimal residual disease in cutaneous T-cell lymphoma: molecular and in situ analysis of a single neoplastic T-cell clone expressing the identical T-cell receptor.
J Clin Oncol
1995
, vol. 
13
 (pg. 
1751
-
1757
)
52
Hoppe
 
RT
Cox
 
RS
Fuks
 
Z
Price
 
NM
Bagshaw
 
MA
Farber
 
EM
Electron beam therapy for mycosis fungoides: the Stanford University experience.
Cancer Treat Rep
1979
, vol. 
63
 (pg. 
691
-
700
)
53
Fuks
 
ZY
Bagshaw
 
MA
Farber
 
EM
Prognostic signs and the management of the mycosis fungoides.
Cancer
1973
, vol. 
32
 (pg. 
1385
-
1395
)
54
Cheson
 
BD
Horning
 
SJ
Coiffier
 
B
, et al. 
Report of an international workshop to standardize response criteria for non-Hodgkin's lymphomas.
J Clin Oncol
1999
, vol. 
17
 (pg. 
1244
-
1253
)
55
Kim
 
Y
Willemze
 
R
Pimpinelli
 
N
, et al. 
TNM classification system for primary cutaneous lymphomas other than mycosis fungoides and Sézary syndrome: a proposal of the International Society for Cutaneous Lymphomas (ISCL) and the Cutaneous Lymphoma Task Force of the European Organization of Research and Treatment of Cancer (EORTC).
Blood
2007
, vol. 
110
 (pg. 
479
-
484
)
56
Burke
 
JS
Colby
 
TV
Dermatopathic lymphadenopathy: comparison of cases associated and unassociated with mycosis fungoides
Am J Surg Pathol
1981
, vol. 
5
 (pg. 
343
-
352
)
57
Clendenning
 
WE
Rappaport
 
HW
Report of the committee on pathology of cutaneous T cell lymphomas.
Cancer Treat Report
1979
, vol. 
63
 (pg. 
719
-
724
)
58
Scheffer
 
E
Meijer
 
CJLM
van Vloten
 
WA
Dermatopathic lymphadenopathy and lymph node involvement in mycosis fungoides.
Cancer
1980
, vol. 
45
 (pg. 
137
-
148
)
59
Colby
 
TV
Burke
 
JS
Hoppe
 
RT
Lymph node biopsy in mycosis fungoides.
Cancer
1981
, vol. 
47
 (pg. 
351
-
359
)
60
Vonderheid
 
EC
Diamond
 
LW
van Vloten
 
WA
, et al. 
Lymph node classification systems in cutaneous T-cell lymphoma: evidence for the utility of the working formulation of non-Hodgkin's lymphomas for clinical usage.
Cancer
1994
, vol. 
73
 (pg. 
207
-
218
)
61
Toro
 
JR
Stoll
 
HL
Stomper
 
PC
Oseroff
 
AR
Prognostic factors and evaluation of mycosis fungoides and Sézary syndrome.
J Am Acad Dermatol
1997
, vol. 
37
 (pg. 
58
-
67
)
62
Kern
 
DE
Kidd
 
PG
Moe
 
R
Hanke
 
D
Olerud
 
JE
Analysis of T-cell receptor gene rearrangement in lymph nodes of patients with mycosis fungoides.
Prognostic implications Arch Dermatol
1998
, vol. 
134
 (pg. 
158
-
164
)
63
Wechsler
 
J
Diebold
 
J
Gerard-Marchant
 
R
Aspects histopathologiques ganglionnaires au cours des lymphomas cutanés de type mycosis fungoïde ou syndrome de Sézary: etude rétrospective de 98 biopsies.
Ann Pathol
1987
, vol. 
7
 (pg. 
122
-
129
)
64
Lynch
 
JW
Linoilla
 
I
Sausville
 
EA
, et al. 
Prognostic implications of evaluation for lymph node involvement by T-cell antigen receptor gene rearrangement in mycosis fungoides.
Blood
1992
, vol. 
79
 (pg. 
3293
-
3299
)
65
Bakels
 
V
van Oostveen
 
JW
Gordijn
 
RLJ
Walboomers
 
JMM
Meijer
 
CJLM
Willemze
 
R
Diagnostic value of T-cell receptor beta gene rearrangement analysis on peripheral blood lymphocytes of patients with erythroderma.
J Invest Dermatol
1991
, vol. 
97
 (pg. 
782
-
786
)
66
Wood
 
GS
Haeffner
 
A
Dummer
 
R
Crooks
 
CF
Molecular biology techniques for the diagnosis of cutaneous T-cell lymphoma.
Dermatol Clin
1994
, vol. 
12
 (pg. 
231
-
241
)
67
Fraser-Andrews
 
EA
Mitchell
 
T
Ferreira
 
S
, et al. 
Molecular staging of lymph nodes from 60 patients with mycosis fungoides and Sézary syndrome: correlation with histopathology and outcome suggests prognostic relevance in mycosis fungoides.
Br J Dermatol
2006
, vol. 
155
 (pg. 
756
-
762
)
68
Assaf
 
C
Hummel
 
M
Steinhoff
 
M
, et al. 
Early TCR-β and TCR-γ PCR detection of T-cell clonality indicates minimal tumor disease in lymph nodes of cutaneous T-cell lymphoma: diagnostic and prognostic implications.
Blood
2005
, vol. 
105
 (pg. 
503
-
510
)
69
Juarez
 
T
Isenbath
 
SN
Polissar
 
NL
, et al. 
Analysis of T-cell receptor gene rearrangement for predicting clinical outcome in patients with cutaneous T-cell lymphoma: a comparison of Southern blot and polymerase chain reaction methods.
Arch Dermatol
2005
, vol. 
141
 (pg. 
1107
-
1113
)
70
Galindo
 
LM
Garcia
 
FU
Hanau
 
CA
, et al. 
Fine needle aspiration biopsy in the evaluation of lymphadenopathy associated with cutaneous T-cell lymphoma (Mycosis fungoides/Sézary syndrome).
Am J Clin Pathol
2000
, vol. 
113
 (pg. 
865
-
871
)
71
Papa
 
VI
Hussain
 
HK
Reznek
 
RH
, et al. 
Role of image-guided core-needle biopsy in the management of patients with lymphoma.
J Clin Oncol
1996
, vol. 
14
 (pg. 
2427
-
2430
)
72
Demharter
 
J
Müller
 
P
Wagner
 
T
Schlimok
 
G
Haude
 
K
Bohndorf
 
K
Percutaneous core-needle biopsy of enlarged lymph nodes in the diagnosis and subclassification of malignant lymphomas.
Eur Radiol
2001
, vol. 
11
 (pg. 
276
-
283
)
73
Schoengen
 
A
Binder
 
T
Fembacher
 
P
Zeelen
 
U
The fine-needle aspiration cytology of lymph nodes suspected of malignancy: its diagnostic value and capacity to predict the histogenesis.
Dtsch Med Wochenschr
1995
, vol. 
120
 (pg. 
549
-
554
)
74
Vonderheid
 
EC
Diamond
 
LW
Lai
 
S-M
Au
 
F
Dellavecchia
 
MA
Lymph node histopathologic findings in cutaneous T-cell lymphoma: a prognostic classification system based on morphologic assessment.
Am J Clin Pathol
1992
, vol. 
97
 (pg. 
121
-
129
)
75
Breneman
 
DL
Raju
 
US
Breneman
 
JC
, et al. 
Lymph node grading for staging of mycosis fungoides may benefit from examination of multiple excised lymph nodes.
J Am Acad Dermatol
2003
, vol. 
48
 (pg. 
702
-
706
)
76
Fraser-Andrews
 
E
Whittaker
 
S
Russell-Jones
 
R
Should multiple lymph node biopsies be taken [letter]?
J Am Acad Dermatol
2004
, vol. 
51
 pg. 
483
 
77
Bunn
 
PA
Huberman
 
MS
Whang-Peng
 
J
, et al. 
Prospective staging evaluation of patients with cutaneous T-cell lymphomas: demonstration of a high frequency of extracutaneous dissemination.
Ann Int Med
1980
, vol. 
93
 (pg. 
223
-
230
)
78
Lister
 
TA
Crowther
 
D
Sutcliffe
 
SB
, et al. 
Report of a committee convened to discuss the evaluation and staging of patients with Hodgkin's disease: Cotswolds meeting.
J Clin Oncol
1989
, vol. 
7
 (pg. 
1630
-
1636
)
79
Huberman
 
MS
Bunn
 
PA
Matthews
 
MJ
, et al. 
Hepatic involvement in the cutaneous T-cell lymphomas: results of percutaneous biopsy and peritoneoscopy.
Cancer
1980
, vol. 
45
 (pg. 
1683
-
1688
)
80
Sibaud
 
V
Beylot-Barry
 
M
Thiébaut
 
R
, et al. 
Bone marrow histopathologic and molecular staging in epidermotropic T-cell lymphomas.
Am J Clin Pathol
2003
, vol. 
119
 (pg. 
414
-
423
)
81
Graham
 
SJ
Sharpe
 
RW
Steinberg
 
SM
Cotelingam
 
JD
Sausville
 
EA
Foss
 
FM
Prognostic implications of a bone marrow histopathologic classification system in mycosis fungoides and the Sézary syndrome.
Cancer
1993
, vol. 
72
 (pg. 
726
-
734
)
82
Foss
 
FM
Sausville
 
EA
Prognosis and staging of cutaneous T-cell lymphoma.
Hem Oncolog Clin NA
1995
, vol. 
9
 (pg. 
1011
-
1019
)
83
Salhany
 
KE
Greer
 
JP
Cousar
 
JB
Collins
 
RD
Marrow involvement in cutaneous T-cell lymphoma: a clinicopathologic study of 60 cases.
Am J Clin Pathol
1989
, vol. 
92
 (pg. 
747
-
754
)
84
Diamandidou
 
E
Colome
 
M
Fayad
 
L
Duvic
 
M
Kurzrock
 
R
Prognostic factor analysis in mycosis fungoides/Sézary syndrome.
J Am Acad Dermatol
1999
, vol. 
40
 (pg. 
914
-
924
)
85
Beylot-Barry
 
M
Parrens
 
M
Delaunay
 
M
, et al. 
Is bone marrow biopsy necessary in patients with mycosis fungoides and Sézary syndrome? a histological and molecular study at diagnosis and during follow-up.
Br J Dermatol
2005
, vol. 
152
 (pg. 
1378
-
1379
)
86
Olsen
 
EA
Delzell
 
E
Jegasothy
 
BV
Second malignancies in cutaneous T-cell lymphoma.
J Am Acad Dermatol
1984
, vol. 
10
 (pg. 
197
-
204
)
87
Väkevä
 
L
Pukkala
 
E
Ranki
 
A
Increased risk of secondary cancers in patients with primarycutaneous T cell lymphoma.
J Invest Dermatol
2000
, vol. 
115
 (pg. 
62
-
65
)
88
Huang
 
KP
Weinstock
 
MA
Clarke
 
CA
Hoppe
 
R
Kim
 
Y
Increased risk for second lymphomas in patients with mycosis fungoides and Sézary syndrome [abstract].
J Invest Dermatol
2005
, vol. 
124
 pg. 
A48
 
89
Clendenning
 
WE
Brecher
 
G
Van Scott
 
EJ
Mycosis fungoides: relationship to malignant cutaneous reticulosis and the Sézary syndrome.
Arch Dermatol
1964
, vol. 
89
 (pg. 
785
-
792
)
90
Lutzner
 
MA
Jordan
 
HW
The utrastructure of an abnormal cell in Sézary's syndrome.
Blood
1968
, vol. 
31
 (pg. 
719
-
726
)
91
Duncan
 
SC
Winkelmann
 
RK
Circulating Sézary cells in hospitalized dermatology patients.
Br J Dermatol
1978
, vol. 
99
 (pg. 
171
-
178
)
92
Meijer
 
CJLM
van Leeuwen
 
AWFM
van der Loo
 
EM
van de Putte
 
LBA
van Vloten
 
WA
Cerebriform (Sézary like) mononuclear cells in healthy individuals: a morphologically distinct population of T cells: relationship with mycosis fungoides and Sézary's Syndrome.
Virchows Arch. B Cell Path
1977
, vol. 
25
 (pg. 
95
-
104
)
93
Rosas-Uribe
 
A
Variakojis
 
D
Molnar
 
Z
Rappaport
 
H
Mycosis fungoides: an ultrastructural study.
Cancer
1974
, vol. 
34
 (pg. 
634
-
645
)
94
Lutzner
 
MA
Hobbs
 
JW
Horvath
 
P
Ultrastructure of abnormal cells in Sézary syndrome, mycosis fungoides and parapsoriasis en plaque.
Arch Dermatol
1971
, vol. 
103
 (pg. 
375
-
386
)
95
van der Loo
 
EM
Cnossen
 
J
Meijer
 
CJLM
Morphological aspects of T-cell subpopulations in human blood: characterization of the cerebriform mononuclear cells in healthy individuals.
Clin Exp Immunol
1981
, vol. 
43
 (pg. 
506
-
516
)
96
Yeckley
 
JA
Weston
 
WL
Thorne
 
EG
Krueger
 
GC
Production of Sézary-like cells from normal human lymphocytes.
Arch Dermatol
1975
, vol. 
111
 (pg. 
29
-
32
)
97
Reinhold
 
U
Herpertz
 
M
Kukel
 
S
Oltermann
 
I
Uerlich
 
M
Kreysel
 
H-W
Induction of nuclear contour irregularity during T-cell activation via the T-cell receptor/CD3 complex and CD2 antigens in the presence of phorbol esters.
Blood
1994
, vol. 
83
 (pg. 
703
-
706
)
98
Schecter
 
GP
Sausville
 
EA
Fischmann
 
AB
, et al. 
Evaluation of circulating malignant cells provides prognostic information in cutaneous T cell lymphoma.
Blood
1987
, vol. 
69
 (pg. 
841
-
849
)
99
Fraser-Andrews
 
E
Woolford
 
AJ
Russell-Jones
 
R
Seed
 
PT
Whittaker
 
SJ
Detection of a peripheral blood T cell clone is an independent prognostic marker in Sézary syndrome.
J Inv Dermatol
2000
, vol. 
114
 (pg. 
117
-
121
)
100
Scarisbrick
 
JJ
Whittaker
 
S
Evans
 
AV
, et al. 
Prognostic significance of tumor burden in the blood of patients with erythrodermic primary cutaneous T-cell lymphoma.
Blood
2001
, vol. 
97
 (pg. 
624
-
630
)
101
Vonderheid
 
EC
Pena
 
J
Nowell
 
P
Sézary cell counts in erythrodermic cutaneous T cell lymphoma: implications for prognosis and staging.
Leuk Lymphoma
2006
, vol. 
47
 (pg. 
1841
-
1856
)
102
Kuchnio
 
M
Sausville
 
EA
Jaffe
 
ES
, et al. 
Flow cytometric detection of neoplastic T cells in patients with mycosis fungoides based on levels of T-cell receptor expression.
Am J Clin Pathol
1994
, vol. 
102
 (pg. 
856
-
860
)
103
Ginaldi
 
L
Matutes
 
E
Farahat
 
N
De Martinis
 
M
Morilla
 
R
Catovsky
 
D
Differential expression of CD3 and CD7 in T-cell malignancies: a quantitative study by flow cytometry.
Br J Haematol
1996
, vol. 
93
 (pg. 
921
-
927
)
104
Jones
 
D
Dang
 
NH
Duvic
 
M
Washington
 
LT
Huh
 
YO
Absence of CD26 expression is a useful marker for diagnosis of T-cell lymphoma in peripheral blood.
Am J Clin Pathol
2001
, vol. 
115
 (pg. 
885
-
892
)
105
Haynes
 
B
Mann
 
DL
Hemler
 
ME
, et al. 
Characterization of a monoclonal antibody which defines an immunoregulatory T cell subset for immunoglobulin synthesis in man.
Proc Natl Acad Sci U S A
1980
, vol. 
77
 (pg. 
2914
-
2918
)
106
Borowitz
 
MJ
Weidner
 
A
Olsen
 
EA
Picker
 
LJ
Abnormalities of circulating T-cell subpopulations in patients with cutaneous T-cell lymphoma: cutaneous lymphoma-associated antigen expression on T cells correlates with extent of disease.
Leukemia
1993
, vol. 
7
 (pg. 
859
-
863
)
107
Harmon
 
CB
Witzig
 
TE
Katzmann
 
JA
Pittelkow
 
MR
Detection of circulating T cells with CD4+CD7- immunophenotype in patients with benign and malignant lymphoproliferative dermatoses.
J Am Acad Dermatol
1996
, vol. 
35
 (pg. 
404
-
410
)
108
Bernengo
 
MG
Novelli
 
M
Quaglino
 
P
, et al. 
The relevance of the CD4+CD26- subset in the identification of circulating Sézary cells.
Br J Dermatol
2001
, vol. 
144
 (pg. 
125
-
135
)
109
Wood
 
GS
Hong
 
SR
Sasaki
 
DT
, et al. 
Leu-8/CD7 antigen expression of CD3+ T cells: comparative analysis of skin and blood in mycosis fungoides/Sézary syndrome relative to normal blood values.
J Am Acad Dermatol
1990
, vol. 
22
 (pg. 
602
-
607
)
110
Abel
 
EA
Lindae
 
ML
Hoppe
 
RT
Wood
 
G
Benign and malignant forms of erythroderma: cutaneous immunophenotypic characteristics.
J Am Acad Dermatol
1988
, vol. 
9
 (pg. 
1089
-
1095
)
111
Jackow
 
CM
Cather
 
JC
Hearne
 
V
Asano
 
AT
Musser
 
JM
Duvic
 
M
Association of erythrodermic cutaneous T-cell lymphoma, superantigen-positive Staphylococcus aureus and oligoclonal T-cell receptor Vâ gene expansion.
Blood
1997
, vol. 
89
 (pg. 
32
-
40
)
112
Muche
 
J
Lukowsky
 
A
Asadullah
 
K
Gellrich
 
S
Sterry
 
W
Demonstration of frequent occurrence of clonal T cells in the peripheral blood of patients with primary cutaneous T-cell lymphoma.
Blood
1997
, vol. 
90
 (pg. 
1636
-
1642
)
113
Bakels
 
V
van Ostveen
 
JW
Geerts
 
M-L
Gordijn
 
RLJ
, et al. 
Diagnostic and prognostic significance of clonal T-cell receptor beta gene rearrangements in lymph nodes of patients with mycosis fungoides.
J Pathol
1993
, vol. 
170
 (pg. 
249
-
255
)
114
Weinberg
 
JM
Jaworsky
 
C
Benoit
 
M
Telegan
 
B
Rook
 
AH
Lessin
 
SR
The clonal nature of circulating Sézary cells.
Blood
1995
, vol. 
86
 (pg. 
4257
-
4262
)
115
Posnett
 
DN
Sinha
 
R
Kabak
 
S
Russo
 
C
Clonal populations of T cells in normal elderly humans: the T cell equivalent to “benign monoclonal gammopathy.”
J Exp Med
1994
, vol. 
179
 (pg. 
609
-
618
)
116
Muche
 
JM
Sterry
 
W
Gellrich
 
S
Rzany
 
B
Audring
 
H
Lukowsky
 
A
Peripheral blood T-cell clonality in mycosis fungoides and nonlymphoma controls.
Diagn Mol Pathol
2003
, vol. 
12
 (pg. 
142
-
150
)
117
Yawalkar
 
N
Ferenczi
 
K
Jones
 
DA
, et al. 
Profound loss of T-cell receptor repertoire complexity in cutaneous T-cell lymphoma.
Blood
2003
, vol. 
102
 (pg. 
4059
-
4066
)
118
Vonderheid
 
EC
Bernengo
 
MG
The Sézary syndrome: hematologic criteria.
Hematol Oncol Clin N Amer
2003
, vol. 
17
 (pg. 
1367
-
1389
)
119
de Coninck
 
EC
Kim
 
YH
Varghese
 
A
Hoppe
 
RT
Clinical characteristics and outcome of patients with extracutaneous mycosis fungoides.
J Clin Oncol
2001
, vol. 
19
 (pg. 
779
-
784
)
120
Vonderheid
 
EC
Van Scott
 
EJ
Wallner
 
PE
Johnson
 
WC
A 10-year experience with topical mechlorethamine for mycosis fungoides: comparison with patients treated by total-skin electron-beam radiation therapy.
Cancer Treat Rep
1979
, vol. 
63
 (pg. 
681
-
689
)
121
Sobin
 
LH
Wittekind
 
Ch
UICC International Union Against Cancer: TNM Classification of Malignant Tumours.
2002
6th ed.
New York, NY
Wiley-Liss
Sign in via your Institution