Mycosis fungoides

From Compendium of Cancer Genome Aberrations
Jump to navigation Jump to search

Haematolymphoid Tumours (5th ed.)

editHAEM5 Conversion Notes
This page was converted to the new template on 2023-12-07. The original page can be found at HAEM4:Mycosis Fungoides.

(General Instructions – The main focus of these pages is the clinically significant genetic alterations in each disease type. Use HUGO-approved gene names and symbols (italicized when appropriate), HGVS-based nomenclature for variants, as well as generic names of drugs and testing platforms or assays if applicable. Please complete tables whenever possible and do not delete them (add N/A if not applicable in the table and delete the examples). Please do not delete or alter the section headings. The use of bullet points alongside short blocks of text rather than only large paragraphs is encouraged. Additional instructions below in italicized blue text should not be included in the final page content. Please also see Author_Instructions and FAQs as well as contact your Associate Editor or Technical Support)

Primary Author(s)*

Jane Scribner, MD and Daynna J. Wolff, PhD

Cancer Category / Type

Cancer Sub-Classification / Subtype

  • Mycosis Fungoides (MF)

Definition / Description of Disease

MF is a primary cutaneous T-cell lymphoma (CTCL) of epidermotropic small to medium-sized T lymphocytes presenting with skin patches that progress slowly to plaques and tumors. The disease is postulated to be caused by skin-homing mature T cells, the majority of which are CD4-positive. Sézary syndrome (SS) is often used to refer to the leukemic phase of mycosis fungoides. However, recently studies have suggested that there are major genomic and phenotypical differences between these two entities. [1]

Synonyms / Terminology

  • Cutaneous T-cell lymphoma
  • Alibert-Bazin syndrome

Epidemiology / Prevalence

  • Most common CTCL subtype (54% of CTCL)[2]
  • Incidence 4.1/1,000,000 person/years (increasing)
    • Highest in males, blacks, and those over 50 years old[3][4][5]
    • Can occur in children and adolescents[6][7]
  • Prevalence 6.4 million people

Clinical Features

Put your text here and fill in the table (Instruction: Can include references in the table)

Signs and Symptoms EXAMPLE Asymptomatic (incidental finding on complete blood counts)

EXAMPLE B-symptoms (weight loss, fever, night sweats)

EXAMPLE Fatigue

EXAMPLE Lymphadenopathy (uncommon)

Laboratory Findings EXAMPLE Cytopenias

EXAMPLE Lymphocytosis (low level)


editv4:Clinical Features
The content below was from the old template. Please incorporate above.
  • Pruritic, erythematous or poikilodermatous and atrophic skin lesions with a fine scale
  • Progresses over years to decades from patches to plaques and less commonly to nodules/tumors
  • Rarely, MF can present with erythroderma without a diagnosis of Sézary syndrome[8]
  • May have symptoms and/or diagnosis of other inflammatory skin diseases (e.g., eczema or psoriasis) for 3 - 4 years prior to the diagnosis of MF[9]

Sites of Involvement

  • Skin
    • Typically sun protected areas: trunk, buttock, upper thighs
    • Some MF variants: head and neck, axillae and groin, or acral sites
  • Advanced disease - lymph nodes, liver, spleen, lungs and blood

Morphologic Features

Pathologic features are variable and may be non-specific with overlap with benign reactive processes.

  • Early patch stage: superficial band-like or lichenoid infiltrate of lymphocytes and histiocytes; atypical small to medium-sized lymphocytes with cerebriform nuclei confined to basilar epidermis, may have halo and "tag" the basilar layer keratinocytes
  • Plaque stage: Epidermotropism (atypical lymphocytes in epidermis without associated spongiosis); intraepidermal collections of atypical cells (Pautrier microabscesses); papillary dermal fibrosis
  • Tumor stage: Dermal infiltrate more diffuse; may lose epidermotropism

Histologic large cell transformation, defined by >25% of large lymphoid cells in the dermal infiltrate, may occur at any stage, but most commonly in tumor stage mycosis fungoides. They may be positive or negative for CD30[8]

Immunophenotype

The immunologic milieu of mycosis fungoides is predominantly that of mature memory Th2 gene expression and associated cytokine production. [10]

The aberrant loss of normal T-cell antigen expression by immunohistochemistry staining is a useful ancillary test in the diagnosis of mycosis fungoides. [11]

Finding Marker
Positive (T-cell lineage markers) CD3, CD4, CD45RO
Variable expression CD2, CD5, CD7 (often lost, significant only if 90% loss)
Markers with aberrant expression CD8 (CD4:CD8 ratio of 10:1 suggestive of mycosis fungoides), CD30 (may indicate transformation)

The majority of lymphocytes in mycosis fungoides express the αβ TCR, but rare cases have been reported that express γδ TCR.

Mycosis fungoides T-cells are T resident memory cells, exhibiting CCR4+/CLA+/L-selectin-/CCR7- expression. [12]

Chromosomal Rearrangements (Gene Fusions)

Put your text here and fill in the table

Chromosomal Rearrangement Genes in Fusion (5’ or 3’ Segments) Pathogenic Derivative Prevalence Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE t(9;22)(q34;q11.2) EXAMPLE 3'ABL1 / 5'BCR EXAMPLE der(22) EXAMPLE 20% (COSMIC)

EXAMPLE 30% (add reference)

Yes No Yes EXAMPLE

The t(9;22) is diagnostic of CML in the appropriate morphology and clinical context (add reference). This fusion is responsive to targeted therapy such as Imatinib (Gleevec) (add reference).


editv4:Chromosomal Rearrangements (Gene Fusions)
The content below was from the old template. Please incorporate above.
  • No consistent gene fusions
  • CD28-CTLA4 gene fusion identified, resulting in activation of TCR signaling [13]


editv4:Clinical Significance (Diagnosis, Prognosis and Therapeutic Implications).
Please incorporate this section into the relevant tables found in:
  • Chromosomal Rearrangements (Gene Fusions)
  • Individual Region Genomic Gain/Loss/LOH
  • Characteristic Chromosomal Patterns
  • Gene Mutations (SNV/INDEL)

The staging system for mycosis fungoides, which also includes Sézary syndrome, was updated in 2007 by the International Society for Cutaneous Lymphomas (ISCL) and the European Organization of Research and Treatment of Cancer (EORTC) [14]. The revised TNMB staging of MF/SS determines management and treatment, and has been demonstrated to have prognostic significance. [4] The TNMB staging forms the basis for a risk-adapted approach to treatment of mycosis fungoides.

TNMB Stages
Skin
T1 Limited patches, papules, and/or plaques covering <10% of the skin surface.  May futher stratify to T1a (patch only) vs T1b (plaque +/- patch)
T1a Patches only < 10% of skin surface
T1b Plaques +/- patches < 10% of skin surface
T2 Patches, papules or plaques covering > 10% of skin surface.  May further stratify to T2a (patch only) vs T2b (plaque +/- patch)
T2a Patches only > 10% of skin surface
T2b Plaques +/- patches > 10% of skin surface
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 (firm, irregular, clustered, fixed, or 1.5 cm or larger), biopsy not required
N1 Clinically abnormal peripheral lymph nodes; histopathology Dutch grade 1 or NCI LN0-2
N1a Clone negative (defined by PCR or Souther blod analysis of T-cell receptor gene)
N1b Clone positive defined by PCR or Souther blod analysis of T-cell receptor gene)
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 NVI LN4 (clone positive or negative)
Nx Clinically abnormal peripheral lymph nodes; no histologic confirmation
Visceral
M0 No visceral organ invovlement
M1 Visceral invovlement (must have pathology confirmation, for spleen and liver may be diagnosed by imaging criteria; specify organ involved)
Blood
B0 Absence of significant blood invovlement: , 5% of peripheral blood lymphocytes are atypical (Sezary) cells
B0a Clone negative
B0b Clone positive
B1 Low blood tumor burdern: > 5% of peripheral blood lymphocytes are atypical (Sezary) cells but does not meet the criteria of B2
B1a Clone negative
B1b Clone positive
B2 High blood tumor burdern: > 1000/µL Sezary cells with positive clone

The above listed chromosome rearrangement and several gene mutations provide the rational for the use of targeted therapy in mycosis fungoides. These are summarized in the table below.

Review of molecular biomarkers as therapeutic targets for CTCL[15]
Molecular marker/mutation Targeted therapy
CD30 Brentuximab
CTLA4 Ipilimumab
CD28-CTLA4 fusion Ipilimumab
CCR4 Mogamulizumab
NFkB Bortezomib
TNFRSF1B Bortezomib
CD158k/KIR3DL2 IPH4102
JAK/STAT pathway Ruxolitinib (JAK 1/3), Tofacitinib (JAK 1/2)
PDCD1 Pembrolizumab

Individual Region Genomic Gain / Loss / LOH

Put your text here and fill in the table (Instructions: Includes aberrations not involving gene fusions. Can include references in the table. Can refer to CGC workgroup tables as linked on the homepage if applicable.)

Chr # Gain / Loss / Amp / LOH Minimal Region Genomic Coordinates [Genome Build] Minimal Region Cytoband Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE

7

EXAMPLE Loss EXAMPLE

chr7:1- 159,335,973 [hg38]

EXAMPLE

chr7

Yes Yes No EXAMPLE

Presence of monosomy 7 (or 7q deletion) is sufficient for a diagnosis of AML with MDS-related changes when there is ≥20% blasts and no prior therapy (add reference).  Monosomy 7/7q deletion is associated with a poor prognosis in AML (add reference).

EXAMPLE

8

EXAMPLE Gain EXAMPLE

chr8:1-145,138,636 [hg38]

EXAMPLE

chr8

No No No EXAMPLE

Common recurrent secondary finding for t(8;21) (add reference).

editv4:Genomic Gain/Loss/LOH
The content below was from the old template. Please incorporate above.

Mycosis fungoides does not have a defining disease specific molecular abnormality. However, the molecular profile of mycosis fungoides and other CTCLs continues to be investigated.

Large-scale genomic analysis using array comparative genomic hybridization and next generation sequencing including whole genome, transcriptome and exome sequencing, reveals significant genomic complexity in mycosis fungoides, [16] with an observation that there is an inverse correlation between genomic complexity and survival. [17]

Chromosome Number Gain/Loss/Amp/LOH Percent of cases Consequence
1p Loss 38% Minimal region of deletion at D1S228; other candidate genes in mycosis fungoides map to 1p12p26
17p Loss 21% Disease progression by inactivation of genes on 17p, including p53
4/4q Gain 18% Oncogenes on chromosome 4: hPTTG, TBC1D1, FGFR3, KIT, PDGFRA
10q/10 Loss 15% Minimal region of deletion at 10q26; cancer-related genes mapped to 10q: PTEN, LG11, DMBT1, FAS
19 Loss 15%
18 Gain 15%
17q/17 Gain 12%

Chromothripsis has been observed in up to 65% of CTCLs. Most events occur in chromosomes 10, 2, and 1. It is possible that these events may be the mechanism for simultaneous inactivation of multiple tumor suppressor genes see in mycosis fungoides. [18]


editUnassigned References
The following referenees were placed in the header. Please place them into the appropriate locations in the text.

[19]

Characteristic Chromosomal Patterns

Put your text here (EXAMPLE PATTERNS: hyperdiploid; gain of odd number chromosomes including typically chromosome 1, 3, 5, 7, 11, and 17; co-deletion of 1p and 19q; complex karyotypes without characteristic genetic findings; chromothripsis)

Chromosomal Pattern Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE

Co-deletion of 1p and 18q

Yes No No EXAMPLE:

See chromosomal rearrangements table as this pattern is due to an unbalanced derivative translocation associated with oligodendroglioma (add reference).

editv4:Characteristic Chromosomal Aberrations / Patterns
The content below was from the old template. Please incorporate above.

Complex chromosomal abnormalities have been identified in mycosis fungoides, usually in tumor stage. Specific translocations have not been identified.

Gene Mutations (SNV / INDEL)

Put your text here and fill in the table (Instructions: This table is not meant to be an exhaustive list; please include only genes/alterations that are recurrent and common as well either disease defining and/or clinically significant. Can include references in the table. For clinical significance, denote associations with FDA-approved therapy (not an extensive list of applicable drugs) and NCCN or other national guidelines if applicable; Can also refer to CGC workgroup tables as linked on the homepage if applicable as well as any high impact papers or reviews of gene mutations in this entity.)

Gene; Genetic Alteration Presumed Mechanism (Tumor Suppressor Gene [TSG] / Oncogene / Other) Prevalence (COSMIC / TCGA / Other) Concomitant Mutations Mutually Exclusive Mutations Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE: TP53; Variable LOF mutations

EXAMPLE:

EGFR; Exon 20 mutations

EXAMPLE: BRAF; Activating mutations

EXAMPLE: TSG EXAMPLE: 20% (COSMIC)

EXAMPLE: 30% (add Reference)

EXAMPLE: IDH1 R123H EXAMPLE: EGFR amplification EXAMPLE:  Excludes hairy cell leukemia (HCL) (add reference).


Note: A more extensive list of mutations can be found in cBioportal (https://www.cbioportal.org/), COSMIC (https://cancer.sanger.ac.uk/cosmic), ICGC (https://dcc.icgc.org/) and/or other databases. When applicable, gene-specific pages within the CCGA site directly link to pertinent external content.


editv4:Gene Mutations (SNV/INDEL)
The content below was from the old template. Please incorporate above.

Although no disease specific molecular abnormalities exist in mycosis fungoides, but some changes are seen more frequently than others.[15] Single copy number variations (SCNV) are important mutational drivers for CTCLs and mycosis fungoides and are seen with greater frequency than somatic single nucleotide variations (SSNV) when compared to other cancers with significantly higher SCNV/SSNV ratios.[16] [18] Alterations in tumor suppressor genes are frequently implication the pathogenesis of mycosis fungoides, and more than 90% of variants arise from copy number alterations. [16]

Gene Mutation Oncogene/Tumor Suppressor/Other Presumed Mechanism (LOF/GOF/Other; Driver/Passenger) Prevalence (COSMIC/TCGA/Other)
TNFRSF1B Thr377Ile Recurrent point mutation; constitutive activation of NFkB signaling pathway 18%[20]
HNRNPK Tumor Suppressor Inhibitor of JAK-STAT signaling[21]
SOCS1 Tumor Suppressor Inhibitor of JAK-STAT signaling [21]
ZEB1 Tumor Suppressor Zinc-finger transcription repressor 56-65%[18]
PDCD1 Expressing PDL1, deleted 36%[18]
TP53 Tumor Suppressor Deletion 92.5% [18]
TOX Encodes member of homeobox family, upregulated in MF[22]

Epigenomic Alterations

Epigenetic modifiers of DNA, including DNMT3A, are frequently mutated in mycosis fungoides. [23]

An epigenetic mediator, the writer KMT2C of the histone methyltransferases (KMT) family, is recurrently deleted in mycosis fungoides. [18]

Genes and Main Pathways Involved

Put your text here and fill in the table (Instructions: Can include references in the table.)

Gene; Genetic Alteration Pathway Pathophysiologic Outcome
EXAMPLE: BRAF and MAP2K1; Activating mutations EXAMPLE: MAPK signaling EXAMPLE: Increased cell growth and proliferation
EXAMPLE: CDKN2A; Inactivating mutations EXAMPLE: Cell cycle regulation EXAMPLE: Unregulated cell division
EXAMPLE:  KMT2C and ARID1A; Inactivating mutations EXAMPLE:  Histone modification, chromatin remodeling EXAMPLE:  Abnormal gene expression program
editv4:Genes and Main Pathways Involved
The content below was from the old template. Please incorporate above.

Genetic Diagnostic Testing Methods

A definitive diagnosis of mycosis fungoides may be made on the basis of clinical, histopathologic and immunohistochemical features alone.

Assessment of aberrant loss of T-cell antigen expression by immunohistochemical staining for CD2, CD3, CD5, and CD7 are useful ancillary studies.

Demonstration of T-cell receptor clonality often facilitates diagnosis and serves as an adjunct to the diagnosis.[24] Presence of identical clones from two different biopsy sites is quit specific for mycosis fungoides[25] and may be useful in early or histologically equivocal cases. However early mycosis fungoides may commonly be negative for clonal TCR rearrangements [26] and T-cell clonality itself is not diagnostic of a T cell lymphoma as many dermatitis may have dominant T cells clones[15].

There are various methods to assess TCR clonality, with different techniques and sensitivities. PCR based assays have sensitivities ranging from 50-90% in various studies[27] [28]. The use of Next generation sequencing (NGS)/high throughput sequencing has improved the sensitivity of detection of TCR clonality, and may be as high as 85%[29][30].

In 2020, the American Society of Clinical Pathology, the College of American pathologists, and the American Society of Hematology released a statement from an expert panel convened to develop evidence-based guidelines for appropriate evaluation process for adult patients with suspected lymphomas[31]. In their summary, it is stated that clinical care providers should not routinely use up-front PCR-based clonality studies of antigen receptor genes (i.e., T-cell receptor) in the initial investigation of lymphoma. However, there may be a confirmatory role of these tests in certain settings. Providers should rely on immunophenotyping by flow cytometry and/or IHC in addition to morphology for the evaluation of specimens for the diagnosis and sub-classification of lymphoma[31]. A recent study demonstrated that positive TCR gene rearrangement studies are not predictive of lymphoproliferative disorders in patients which otherwise negative phenotyping[32].

Summary of additional diagnostic and prognostic testing methods:

  • TCR gene rearrangement studies may be utilized for monitoring of residual disease[33]
  • Tumor clone frequency as determined by high throughput sequencing of TCRβ gene has been investigated as a marker for progression and overall survival in mycosis fungoides[34]
  • In cases with suspected large cell transformation, staining for CD30 positivity may provide additional therapeutic options[35]
  • Recent diagnostic panels of gene expression profiling of MF/SS have been developed. A 17 gene signature panel including IL2RA, CCR4, STAT5A and TOX has been described to identify patients at risk for progression of disease[36]
  • microRNA profiling may have a role in diagnosis and management of mycosis fungoides, with potential 95% sensitivity and specificity in differentiating early mycosis fungoides from benign lesions[37]

Familial Forms

  • There are rare reports of familial MF, suggesting that a host genetic factor may contribute to the development of the disease[38]

Additional Information

  • N/A

Links

  • N/A

References

(use the "Cite" icon at the top of the page) (Instructions: Add each reference into the text above by clicking on where you want to insert the reference, selecting the “Cite” icon at the top of the page, and using the “Automatic” tab option to search such as by PMID to select the reference to insert. The reference list in this section will be automatically generated and sorted. If a PMID is not available, such as for a book, please use the “Cite” icon, select “Manual” and then “Basic Form”, and include the entire reference.)

  1. Campbell, James J.; et al. (2010-08-05). "Sézary syndrome and mycosis fungoides arise from distinct T-cell subsets: a biologic rationale for their distinct clinical behaviors". Blood. 116 (5): 767–771. doi:10.1182/blood-2009-11-251926. ISSN 0006-4971. PMC 2918332. PMID 20484084.
  2. Bradford, Porcia T.; et al. (2009-05-21). "Cutaneous lymphoma incidence patterns in the United States: a population-based study of 3884 cases". Blood. 113 (21): 5064–5073. doi:10.1182/blood-2008-10-184168. ISSN 1528-0020. PMC 2686177. PMID 19279331.
  3. Criscione, Vincent D.; et al. (2007-07-01). "Incidence of Cutaneous T-Cell Lymphoma in the United States, 1973-2002". Archives of Dermatology. 143 (7). doi:10.1001/archderm.143.7.854. ISSN 0003-987X.
  4. 4.0 4.1 Agar, Nita Sally; et al. (2010-11-01). "Survival outcomes and prognostic factors in mycosis fungoides/Sézary syndrome: validation of the revised International Society for Cutaneous Lymphomas/European Organisation for Research and Treatment of Cancer staging proposal". Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 28 (31): 4730–4739. doi:10.1200/JCO.2009.27.7665. ISSN 1527-7755. PMID 20855822.
  5. Hinds, Ginette A.; et al. (2009-03). "Cutaneous T-cell lymphoma in skin of color". Journal of the American Academy of Dermatology. 60 (3): 359–375, quiz 376–378. doi:10.1016/j.jaad.2008.10.031. ISSN 1097-6787. PMID 19231637. Check date values in: |date= (help)
  6. Boccara, Olivia; et al. (2012-02). "Cutaneous hematologic disorders in children". Pediatric Blood & Cancer. 58 (2): 226–232. doi:10.1002/pbc.23103. ISSN 1545-5017. PMID 21445946. Check date values in: |date= (help)
  7. Fink-Puches, Regina; et al. (2004-09). "The spectrum of cutaneous lymphomas in patients less than 20 years of age". Pediatric Dermatology. 21 (5): 525–533. doi:10.1111/j.0736-8046.2004.21500.x. ISSN 0736-8046. PMID 15461755. Check date values in: |date= (help)
  8. 8.0 8.1 Elder DE, Massi D, Scolyer RA, Willemze R. WHO Classification of Skin Tumours. 4th edn. Lyon, France: International Agency for Research on Cancer; 2018
  9. Kim, Youn H.; et al. (2003-07-01). "Long-term Outcome of 525 Patients With Mycosis Fungoides and Sézary Syndrome: Clinical Prognostic Factors and Risk for Disease Progression". Archives of Dermatology. 139 (7). doi:10.1001/archderm.139.7.857. ISSN 0003-987X.
  10. Wilcox, Ryan A. (2016-01). "Cutaneous T-cell lymphoma: 2016 update on diagnosis, risk-stratification, and management". American Journal of Hematology. 91 (1): 151–165. doi:10.1002/ajh.24233. ISSN 1096-8652. PMC 4715621. PMID 26607183. Check date values in: |date= (help)
  11. Hristov, Alexandra C.; et al. (2019-07-31). "Mycosis fungoides and Sézary syndrome: 2019 update on diagnosis, risk‐stratification, and management". American Journal of Hematology. 94 (9): 1027–1041. doi:10.1002/ajh.25577. ISSN 0361-8609.
  12. Campbell, James J.; et al. (2010-08-05). "Sézary syndrome and mycosis fungoides arise from distinct T-cell subsets: a biologic rationale for their distinct clinical behaviors". Blood. 116 (5): 767–771. doi:10.1182/blood-2009-11-251926. ISSN 0006-4971. PMC 2918332. PMID 20484084.CS1 maint: PMC format (link)
  13. Ungewickell, Alexander; et al. (2015-09). "Genomic analysis of mycosis fungoides and Sézary syndrome identifies recurrent alterations in TNFR2". Nature Genetics. 47 (9): 1056–1060. doi:10.1038/ng.3370. ISSN 1061-4036. PMC 6091217. PMID 26258847. Check date values in: |date= (help)CS1 maint: PMC format (link)
  14. Olsen, Elise; et al. (2007-09-15). "Revisions to the staging and classification of mycosis fungoides and Sezary 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. 110 (6): 1713–1722. doi:10.1182/blood-2007-03-055749. ISSN 0006-4971. PMID 17540844.
  15. 15.0 15.1 15.2 Walia, Ritika; et al. (2020-01-22). "An Update on Molecular Biology of Cutaneous T Cell Lymphoma". Frontiers in Oncology. 9: 1558. doi:10.3389/fonc.2019.01558. ISSN 2234-943X. PMC 6987372. PMID 32039026 Check |pmid= value (help).CS1 maint: PMC format (link)
  16. 16.0 16.1 16.2 Elenitoba-Johnson, Kojo S.J.; et al. (2017-01). "A new molecular paradigm in mycosis fungoides and Sézary syndrome". Seminars in Diagnostic Pathology. 34 (1): 15–21. doi:10.1053/j.semdp.2016.11.002. Check date values in: |date= (help)
  17. Wilcox, Ryan A. (2011-11). "Cutaneous T-cell lymphoma: 2011 update on diagnosis, risk-stratification, and management". American Journal of Hematology. 86 (11): 928–948. doi:10.1002/ajh.22139. ISSN 1096-8652. PMID 21990092. Check date values in: |date= (help)
  18. 18.0 18.1 18.2 18.3 18.4 18.5 Choi, Jaehyuk; et al. (2015-09). "Genomic landscape of cutaneous T cell lymphoma". Nature Genetics. 47 (9): 1011–1019. doi:10.1038/ng.3356. ISSN 1546-1718. PMC 4552614. PMID 26192916. Check date values in: |date= (help)
  19. Mao, X.; et al. (2002-09). "Molecular cytogenetic analysis of cutaneous T-cell lymphomas: identification of common genetic alterations in Sézary syndrome and mycosis fungoides". The British Journal of Dermatology. 147 (3): 464–475. doi:10.1046/j.1365-2133.2002.04966.x. ISSN 0007-0963. PMID 12207585. Check date values in: |date= (help)
  20. Izban, K (2000-12). "Constitutive expression of NF-κB is a characteristic feature of mycosis fungoides: Implications for apoptosis resistance and pathogenesis". Human Pathology. 31 (12): 1482–1490. doi:10.1053/hupa.2000.20370. Check date values in: |date= (help)
  21. 21.0 21.1 Bastidas Torres, Armando N.; et al. (2018-12). "Genomic analysis reveals recurrent deletion of JAK-STAT signaling inhibitors HNRNPK and SOCS1 in mycosis fungoides". Genes, Chromosomes and Cancer. 57 (12): 653–664. doi:10.1002/gcc.22679. PMC 6282857. PMID 30144205. Check date values in: |date= (help)CS1 maint: PMC format (link)
  22. Huang, Yuanshen; et al. (2014-06-30). "Thymocyte selection-associated high mobility group box gene (TOX) is aberrantly over-expressed in mycosis fungoides and correlates with poor prognosis". Oncotarget. 5 (12): 4418–4425. doi:10.18632/oncotarget.2031. ISSN 1949-2553. PMC 4147334. PMID 24947046.CS1 maint: PMC format (link)
  23. Kiel, Mark J.; et al. (2015-09-29). "Genomic analyses reveal recurrent mutations in epigenetic modifiers and the JAK–STAT pathway in Sézary syndrome". Nature Communications. 6 (1): 8470. doi:10.1038/ncomms9470. ISSN 2041-1723. PMC 4598843. PMID 26415585.CS1 maint: PMC format (link)
  24. Kirsch, Ilan R.; et al. (2015-10-07). "TCR sequencing facilitates diagnosis and identifies mature T cells as the cell of origin in CTCL". Science Translational Medicine. 7 (308): 308ra158. doi:10.1126/scitranslmed.aaa9122. ISSN 1946-6242. PMC 4765389. PMID 26446955.
  25. Thurber, Stacy E.; et al. (2007-11). "T-cell clonality analysis in biopsy specimens from two different skin sites shows high specificity in the diagnosis of patients with suggested mycosis fungoides". Journal of the American Academy of Dermatology. 57 (5): 782–790. doi:10.1016/j.jaad.2007.06.004. ISSN 1097-6787. PMID 17646032. Check date values in: |date= (help)
  26. Glusac, Earl J. (2003-06). "Criterion by criterion, mycosis fungoides". The American Journal of Dermatopathology. 25 (3): 264–269. doi:10.1097/00000372-200306000-00014. ISSN 0193-1091. PMID 12775992. Check date values in: |date= (help)
  27. Hsiao, Pa-Fan; et al. (2007-01-01). "Histopathologic-molecular Correlation in Early Mycosis Fungoides Using T-cell Receptor γ Gene Rearrangement by Polymerase Chain Reaction with Laser Capture Microdissection". Journal of the Formosan Medical Association. 106 (4): 265–272. doi:10.1016/S0929-6646(09)60251-5. ISSN 0929-6646.
  28. Ponti, Renata; et al. (2008-04). "TCRγ-Chain Gene Rearrangement by PCR-Based GeneScan: Diagnostic Accuracy Improvement and Clonal Heterogeneity Analysis in Multiple Cutaneous T-Cell Lymphoma Samples". Journal of Investigative Dermatology. 128 (4): 1030–1038. doi:10.1038/sj.jid.5701109. ISSN 0022-202X. Check date values in: |date= (help)
  29. Sufficool, Kari E.; et al. (2015-08). "T-cell clonality assessment by next-generation sequencing improves detection sensitivity in mycosis fungoides". Journal of the American Academy of Dermatology. 73 (2): 228–236.e2. doi:10.1016/j.jaad.2015.04.030. ISSN 0190-9622. Check date values in: |date= (help)
  30. Rea, Bryan; et al. (2018-09). "Role of high-throughput sequencing in the diagnosis of cutaneous T-cell lymphoma". Journal of Clinical Pathology. 71 (9): 814–820. doi:10.1136/jclinpath-2018-205004. ISSN 0021-9746. Check date values in: |date= (help)
  31. 31.0 31.1 Kroft, Steven H.; et al. (2020-11-11). "Laboratory Workup of Lymphoma in Adults". Archives of Pathology & Laboratory Medicine. doi:10.5858/arpa.2020-0261-SA. ISSN 1543-2165.
  32. Mendoza, Hadrian; et al. (2021-02-01). "Evaluation of Positive B- and T-Cell Gene Rearrangement Studies in Patients With Negative Morphology, Flow Cytometry, and Immunohistochemistry". Archives of Pathology & Laboratory Medicine. 145 (2): 227–230. doi:10.5858/arpa.2019-0663-OA. ISSN 1543-2165.
  33. Weng, W.-K.; et al. (2013-12-04). "Minimal Residual Disease Monitoring with High-Throughput Sequencing of T Cell Receptors in Cutaneous T Cell Lymphoma". Science Translational Medicine. 5 (214): 214ra171–214ra171. doi:10.1126/scitranslmed.3007420. ISSN 1946-6234.
  34. de Masson, Adele; et al. (2018-05-09). "High-throughput sequencing of the T cell receptor β gene identifies aggressive early-stage mycosis fungoides". Science Translational Medicine. 10 (440): eaar5894. doi:10.1126/scitranslmed.aar5894. ISSN 1946-6234. PMC 6366329. PMID 29743350.CS1 maint: PMC format (link)
  35. Krathen, Michael; et al. (2012-11-16). "Brentuximab Vedotin Demonstrates Significant Clinical Activity in Relapsed or Refractory Mycosis Fungoides with Variable CD30 Expression". Blood. 120 (21): 797–797. doi:10.1182/blood.V120.21.797.797. ISSN 0006-4971.
  36. Litvinov, Ivan V.; et al. (2015-06-15). "The Use of Transcriptional Profiling to Improve Personalized Diagnosis and Management of Cutaneous T-cell Lymphoma (CTCL)". Clinical Cancer Research. 21 (12): 2820–2829. doi:10.1158/1078-0432.CCR-14-3322. ISSN 1078-0432. PMC 4470792. PMID 25779945.CS1 maint: PMC format (link)
  37. Ralfkiaer, Ulrik; et al. (2011-11-24). "Diagnostic microRNA profiling in cutaneous T-cell lymphoma (CTCL)". Blood. 118 (22): 5891–5900. doi:10.1182/blood-2011-06-358382. ISSN 0006-4971. PMC 3342856. PMID 21865341.CS1 maint: PMC format (link)
  38. Hodak, Emmilia; et al. (2005-03). "Familial mycosis fungoides: report of 6 kindreds and a study of the HLA system". Journal of the American Academy of Dermatology. 52 (3 Pt 1): 393–402. doi:10.1016/j.jaad.2003.12.052. ISSN 1097-6787. PMID 15761416. Check date values in: |date= (help)

Notes

*Primary authors will typically be those that initially create and complete the content of a page.  If a subsequent user modifies the content and feels the effort put forth is of high enough significance to warrant listing in the authorship section, please contact the CCGA coordinators (contact information provided on the homepage).  Additional global feedback or concerns are also welcome. *Citation of this Page: “Mycosis fungoides”. Compendium of Cancer Genome Aberrations (CCGA), Cancer Genomics Consortium (CGC), updated 12/13/2023, https://ccga.io/index.php/HAEM5:Mycosis_fungoides.