The WHO Classification of Tumours aims to provide an evidence-based taxonomy for tumors of each organ system to standardize diagnostic practice worldwide and to promote translational research on cancer [1]. The 5th edition of the WHO Classification of Tumours of the Eye and Orbit (WHO Eye5) builds and improves upon the 4th edition (WHO Eye4) by furnishing a comprehensive summary of all tumors of the eye and ocular adnexa that an ophthalmic oncologist and ophthalmic pathologist may encounter [2, 3]. It employs a standardized, evidence-based approach to integrate the salient epidemiologic, ophthalmic, genetic, diagnostic, and prognostic data into a format that allows the reader to grasp the site-specific information on each tumor type and allows cross-referencing with other WHO 5th edition volumes [1, 2]. This review outlines the major general and specific changes in the WHO Eye5, which is currently available online as a beta version ahead of print [2].

The goal of the WHO Classification of Tumours is to provide an up-to-date synthesis of information on cancer, based on expert consensus review of evidence-based published data [1]. Two hundred authors and editors have participated in the production of the WHO Eye5 [2]. The revision was led by the WHO Classification of Tumours Editorial Board, composed of standing and expert members. The standing members have been nominated by pathology organizations and are the equivalent of the series editors of previous editions. The expert members have been selected on the basis of informed bibliometric analysis and advice from the standing members and are equivalent to the volume editors of previous editions. In recognition of the increasingly multidisciplinary approach to cancer diagnosis, prognostication, and management, the 5th edition authors, expert members, and editorial board included oncologists, radiologists, molecular biologists, geneticists, and pathology experts from the overlapping pathology disciplines (head and neck, dermatopathology, soft tissue pathology, and hematopathology) to address WHO Eye5-specific needs [2].

Taxonomy

The WHO Eye5 aims to provide the most comprehensive overview of the eye and orbit tumors an ophthalmic oncologist and pathologist may encounter, with cross-reference to other volumes as necessary. Although the WHO Eye5 continues to be organized by anatomical site (e.g., conjunctiva, eyelid, uvea), its major differences with the WHO Eye4 are incorporation of tumor lineage (e.g., epithelial, adnexal, melanocytic) into the classification system, and synthesis of ocular and periocular soft tissue and bone tumors, hematolymphoid tumors, metastases, and genetic tumor predisposition syndromes into separate chapters, following the structure of the other WHO 5th edition volumes [1, 2]. In line with the 5th edition format, the WHO Eye5 content logically progresses from non-neoplastic tumor-like lesions, to benign neoplasms and, finally, to malignant neoplasms. The standardized, systematic approach is adopted to illuminate each tumor, with addition of the ICD11-codes, imaging features when relevant, macroscopic appearance, cytology, diagnostic molecular pathology, essential and desirable diagnostic criteria, and staging (Table 1).

Table 1.

WHO Eye5: subsection headings and definitions*

Definition 
A concise statement/description of the nature of the tumor 
ICD-0 coding 
ICD-11 coding 
Related terminology 
Synonyms, older terms, and eponymous terms are listed as either “acceptable” or “not recommended” 
Subtypes (formerly known as “variant”) 
A tumor variant in which one or two parameters (e.g., clinical, location, histopathological, and/or molecular) make it desirable to recognize it as being distinct from other subtypes but still related to the parent type (tumor “entity”) 
Localization 
Clinical features (including imaging) 
Epidemiology 
Etiology 
Known risk factors and genetic factors 
Pathogenesis 
Explicates how the causes described in the “Etiology” subsection lead to tumor formation 
Macroscopic appearance 
Histopathology 
H&E appearance and variation in pathological appearance (morphologic patterns) 
Description of recognized histopathological subtypes 
Diagnostic immunohistochemistry (IHC) 
Grading (where applicable) 
Differential diagnosis 
Cytology (when clinically relevant) 
Diagnostic molecular pathology 
Analytic signature of the tumor that is diagnostically useful 
Essential and desirable diagnostic criteria 
Essential criteria – minimal criteria that are mandatory for diagnosis 
Desirable diagnostic criteria – recommended criteria that are not mandatory for diagnosis 
Staging 
Staging system relevant to tumor, such as American Joint Committee on Cancer (AJCC) and the Union for International Cancer Control (UICC) TNM classification, Lugano, etc. 
Prognosis and prediction 
Includes evidence-based information on prognostic and predictive biomarkers, when available 
Definition 
A concise statement/description of the nature of the tumor 
ICD-0 coding 
ICD-11 coding 
Related terminology 
Synonyms, older terms, and eponymous terms are listed as either “acceptable” or “not recommended” 
Subtypes (formerly known as “variant”) 
A tumor variant in which one or two parameters (e.g., clinical, location, histopathological, and/or molecular) make it desirable to recognize it as being distinct from other subtypes but still related to the parent type (tumor “entity”) 
Localization 
Clinical features (including imaging) 
Epidemiology 
Etiology 
Known risk factors and genetic factors 
Pathogenesis 
Explicates how the causes described in the “Etiology” subsection lead to tumor formation 
Macroscopic appearance 
Histopathology 
H&E appearance and variation in pathological appearance (morphologic patterns) 
Description of recognized histopathological subtypes 
Diagnostic immunohistochemistry (IHC) 
Grading (where applicable) 
Differential diagnosis 
Cytology (when clinically relevant) 
Diagnostic molecular pathology 
Analytic signature of the tumor that is diagnostically useful 
Essential and desirable diagnostic criteria 
Essential criteria – minimal criteria that are mandatory for diagnosis 
Desirable diagnostic criteria – recommended criteria that are not mandatory for diagnosis 
Staging 
Staging system relevant to tumor, such as American Joint Committee on Cancer (AJCC) and the Union for International Cancer Control (UICC) TNM classification, Lugano, etc. 
Prognosis and prediction 
Includes evidence-based information on prognostic and predictive biomarkers, when available 

Nomenclature

The WHO Eye5 aims to use the most recent evidence-based nomenclature for classification of tumors. One major change was adoption, where possible, of the terminology proposed in 2018 by the International Society for the Study of Vascular Anomalies (ISSVA) to classify vascular tumors and malformations [4]. The definitions of tumor “type,” “subtype,” and “morphologic pattern” are implemented to classify the tumors (Table 2) [1, 2]. The preference for “subtype” over “variant” stems from the need to differentiate this concept from the homonymous genetic term “variant” to avoid potential confusion. Where appropriate, the WHO 5th edition volumes use the Human Genome Organization (HUGO) Gene Nomenclature Committee (HGNC) system for gene symbols and names (https://www.genenames.org/) [5] and the Human Genome Variation Society (HGVS) recommendations for sequence variants (http://varnomen.hgvs.org) [6]. Another notable change in the 5th edition volumes is conversion of the mitotic count from the traditional denominator, number of high-power fields, to a defined area expressed in mm2[7].

Table 2.

WHO Eye5: definitions of tumor type, subtype, and pattern*

Type (formerly known as “entity”) 
Separate entity in which multiple parameters (e.g., clinical, location, histopathological, and/or molecular) differ from those of other types 
Subtype (formerly known as “variant”) 
A variant of a tumor type in which one or two parameters (e.g., clinical, location, histopathological, and/or molecular) make it desirable to recognize it as being distinct from other subtypes but still related to the parent type 
Pattern 
Morphologic or immunohistochemical pattern in a tumor, without distinct clinical, molecular, or prognostic parameters that make it desirable to be recognized as a subtype 
Type (formerly known as “entity”) 
Separate entity in which multiple parameters (e.g., clinical, location, histopathological, and/or molecular) differ from those of other types 
Subtype (formerly known as “variant”) 
A variant of a tumor type in which one or two parameters (e.g., clinical, location, histopathological, and/or molecular) make it desirable to recognize it as being distinct from other subtypes but still related to the parent type 
Pattern 
Morphologic or immunohistochemical pattern in a tumor, without distinct clinical, molecular, or prognostic parameters that make it desirable to be recognized as a subtype 

Online version

The WHO Classification of Tumours Online website, which currently features the WHO Eye4, was launched in 2019 [1]. The WHO Eye5 will replace this on the website. In addition to the text, tables, and figures, the online edition will provide a whole slide image library, which will allow a more detailed histopathologic representation of various tumors. The online format also allows easy cross-referencing of tumor entities across body sites and organ systems for non-site-specific details.

Tumors of the Conjunctiva and Caruncle (Table 3)

This chapter provides an update on pathogenesis and molecular genetics of melanocytic conjunctival lesions (Table 4) [2, 8‒27]. Spitz nevus is removed from this chapter because of the lack of molecularly well-documented cases of this tumor in the conjunctiva. Conversely, the WNT-activated deep penetrating/plexiform melanocytoma (nevus) (DPN) – the recently agreed terminology for DPN – is added as a new well-characterized entity in the conjunctiva. This follows the 5th edition WHO Classification of the Tumours of the Skin that acknowledges that melanocytomas are genetically “intermediate” between nevus and melanoma because they carry pathogenic mutations additional to the initiating mutation that is the sole alteration in a nevus. Unlike in the skin, a WNT-activated DPN in the conjunctiva is usually a part of a combined nevus (with a common nevocellular or an inflamed juvenile nevus; shown in Fig. 1) [2, 14‒17]. Another notable change is the evolving WHO classification of the conjunctival melanocytic intraepithelial lesions, based on the recent international study and on the WHO consensus editorial meeting between dermatopathologists and ophthalmic pathologists (Table 5) [2, 28, 29]. Non-neoplastic pterygium and pinguecula were added as “tumor-like lesions” to delineate them from conjunctival squamous intraepithelial neoplasia. The term adenosquamous carcinoma, introduced in the Eye4 as the preferred term for mucoepidermoid carcinoma, is now recommended to be applied only to neoplasms with bi-phenotypic differentiation comprising squamous cell carcinoma and adenocarcinoma with mucin-containing cells (shown in Fig. 2) [2, 30]. Tumors that do not form true neoplastic glands/ducts or confluent sheets of cells with intracytoplasmic mucin and, instead, show scattered or small clusters of cells with such mucin in a background of malignant squamous cells are designated as “squamous cell carcinoma with mucinous differentiation” (shown in Fig. 3) [2, 30]. Sebaceous carcinoma, covered in Tumors of the Eyelid chapter, has been removed. Soft tissue tumors, hematolymphoid neoplasms, and secondary tumors of the conjunctiva are discussed in separate chapters.

Table 3.

WHO Eye5: tumors of the conjunctiva and caruncle [2]

Tumor-like lesions and choristomas of the conjunctiva 
Cysts of the conjunctiva and caruncle 
Epithelial conjunctival inclusion cyst 
Reactive, epithelial, and degenerative conjunctival lesions 
Reactive epithelial hyperplasia 
Papillary and follicular conjunctivitis 
Pterygium and pinguecula 
Choristomas 
Epibulbar choristoma (dermoid; dermolipoma; complex) 
Epibulbar osseous choristoma 
Epithelial tumors of the conjunctiva 
Benign epithelial tumors 
Conjunctival squamous papilloma 
Conjunctival oncocytoma 
Hereditary benign intraepithelial dyskeratosis 
Premalignant and malignant epithelial tumors of the conjunctiva 
Conjunctival squamous intraepithelial neoplasia 
Conjunctival squamous cell carcinoma 
Adenosquamous carcinoma 
Melanocytic conjunctival tumors 
Benign melanocytic conjunctival tumors 
Benign epithelial melanosis of the conjunctiva 
Junctional, compound, and subepithelial naevi 
Inflamed juvenile conjunctival nevus 
Blue nevus of the conjunctiva 
WNT-activated DPN 
Combined nevus of the conjunctiva 
Premalignant and malignant melanocytic conjunctival tumors 
Conjunctival melanocytic intraepithelial lesions 
Conjunctival melanoma 
Tumor-like lesions and choristomas of the conjunctiva 
Cysts of the conjunctiva and caruncle 
Epithelial conjunctival inclusion cyst 
Reactive, epithelial, and degenerative conjunctival lesions 
Reactive epithelial hyperplasia 
Papillary and follicular conjunctivitis 
Pterygium and pinguecula 
Choristomas 
Epibulbar choristoma (dermoid; dermolipoma; complex) 
Epibulbar osseous choristoma 
Epithelial tumors of the conjunctiva 
Benign epithelial tumors 
Conjunctival squamous papilloma 
Conjunctival oncocytoma 
Hereditary benign intraepithelial dyskeratosis 
Premalignant and malignant epithelial tumors of the conjunctiva 
Conjunctival squamous intraepithelial neoplasia 
Conjunctival squamous cell carcinoma 
Adenosquamous carcinoma 
Melanocytic conjunctival tumors 
Benign melanocytic conjunctival tumors 
Benign epithelial melanosis of the conjunctiva 
Junctional, compound, and subepithelial naevi 
Inflamed juvenile conjunctival nevus 
Blue nevus of the conjunctiva 
WNT-activated DPN 
Combined nevus of the conjunctiva 
Premalignant and malignant melanocytic conjunctival tumors 
Conjunctival melanocytic intraepithelial lesions 
Conjunctival melanoma 
Table 4.

WHO Eye5: melanocytic tumors of the conjunctiva [2]

Conjunctival melanocytic lesionMolecular genetic alterationsReferences
Junctional, compound, and subepithelial nevi; inflamed juvenile nevus BRAF c.1799T>A p.V600E, NRAS mutations [7–11] 
Blue nevus GNAQ mutations [10, 12] 
WNT-activated DPN BRAF c.1799T>A p.V600E and CTNNB1 mutations [13–16] 
Melanoma High-frequency mutations: NF1 (50–33%), BRAF (46–29%), NRAS (26–11%), ATRX (25%), TERT promoter (up to 54%), CTNNB1 (17%)Other mutations: ACSS3, ET, TP53, CKIT, TET2, CDKN2A, MAPK2, RAC1, MET, SF3B1, GNAQ, GNA1Multiple chromosomal alterations: 6p gain, 11q gain, 6q loss, 10q loss [17–26] 
Conjunctival melanocytic lesionMolecular genetic alterationsReferences
Junctional, compound, and subepithelial nevi; inflamed juvenile nevus BRAF c.1799T>A p.V600E, NRAS mutations [7–11] 
Blue nevus GNAQ mutations [10, 12] 
WNT-activated DPN BRAF c.1799T>A p.V600E and CTNNB1 mutations [13–16] 
Melanoma High-frequency mutations: NF1 (50–33%), BRAF (46–29%), NRAS (26–11%), ATRX (25%), TERT promoter (up to 54%), CTNNB1 (17%)Other mutations: ACSS3, ET, TP53, CKIT, TET2, CDKN2A, MAPK2, RAC1, MET, SF3B1, GNAQ, GNA1Multiple chromosomal alterations: 6p gain, 11q gain, 6q loss, 10q loss [17–26] 
Fig. 1.

Combined nevocellular and WNT-activated deep penetrating/plexiform melanocytoma (nevus) (DPN) of the conjunctiva. a Circumscribed variably pigmented nodule in plica semilunaris, associated with a feeder vessel. b DPN component of the lesion is composed of lightly pigmented spindle melanocytes with abundant cytoplasm (asterisk), associated with scattered darkly pigmented melanophages (arrowhead). Mostly amelanotic nevocellular nevus (arrow) is present in the periphery. The DPN component of the lesion is positive for HMB45 (red chromogen, c), nuclear cyclin D1 (brown chromogen, d), and cytoplasmic and nuclear beta-catenin (brown chromogen, e). The nevocellular nevoid component of the lesion is negative for these markers (arrows, c–e). No proliferative activity is highlighted with Ki-67 (f) [hematoxylin-eosin (b), HMB45 (c), cyclin D1 (d), beta-catenin (e), Ki-67 (f); all figures. ×200].

Fig. 1.

Combined nevocellular and WNT-activated deep penetrating/plexiform melanocytoma (nevus) (DPN) of the conjunctiva. a Circumscribed variably pigmented nodule in plica semilunaris, associated with a feeder vessel. b DPN component of the lesion is composed of lightly pigmented spindle melanocytes with abundant cytoplasm (asterisk), associated with scattered darkly pigmented melanophages (arrowhead). Mostly amelanotic nevocellular nevus (arrow) is present in the periphery. The DPN component of the lesion is positive for HMB45 (red chromogen, c), nuclear cyclin D1 (brown chromogen, d), and cytoplasmic and nuclear beta-catenin (brown chromogen, e). The nevocellular nevoid component of the lesion is negative for these markers (arrows, c–e). No proliferative activity is highlighted with Ki-67 (f) [hematoxylin-eosin (b), HMB45 (c), cyclin D1 (d), beta-catenin (e), Ki-67 (f); all figures. ×200].

Close modal
Table 5.

2022 World Health Organization Classification of Conjunctival Melanocytic Intraepithelial Lesions [2]

WHOAcceptable alternative terminologyIncreased cellularityHistologic featuresRisk of association with or progression to invasive melanoma
Not applicable Benign melanosisc-MIN (grades (0–1)PAM without atypia No/minimal Conjunctival hypermelanosis (increased pigment in epithelial cells without melanocytic hyperplasia or atypia). Slight or focal melanocytic hyperplasia without atypia (parabasal melanocytes with condensed round nuclei, smaller than basal epithelial cell, inconspicuous nucleoli, and inconspicuous cytoplasm) may be seen None 
Low-grade CMIL PAM with mild atypiac-MIN (grades 2–4) Yes Predominantly basilar melanocytic proliferation with low-grade atypia (dendritic or small to moderate size polyhedral, usually non-epithelioid melanocytes with round to irregular nuclear contours, often nuclear hyperchromasia, inconspicuous nucleoli, and inconspicuous or scant cytoplasm) Lower 
High-grade CMIL PAM with moderate to severe atypiac-MIN (grade 5–10) Yes More confluent basilar and significant non-basilar proliferation of melanocytes with high-grade atypia (moderate to severe), evidence of intraepithelial nested and/or pagetoid growth, and epithelioid cell cytomorphology Higher 
Melanoma in situ Yes The term melanoma in situ may be used for (1) the most atypical high-grade CMILs involving close to full thickness of the epithelium, (2) histologically obvious melanomas without documented evidence of subepithelial invasion Highest 
WHOAcceptable alternative terminologyIncreased cellularityHistologic featuresRisk of association with or progression to invasive melanoma
Not applicable Benign melanosisc-MIN (grades (0–1)PAM without atypia No/minimal Conjunctival hypermelanosis (increased pigment in epithelial cells without melanocytic hyperplasia or atypia). Slight or focal melanocytic hyperplasia without atypia (parabasal melanocytes with condensed round nuclei, smaller than basal epithelial cell, inconspicuous nucleoli, and inconspicuous cytoplasm) may be seen None 
Low-grade CMIL PAM with mild atypiac-MIN (grades 2–4) Yes Predominantly basilar melanocytic proliferation with low-grade atypia (dendritic or small to moderate size polyhedral, usually non-epithelioid melanocytes with round to irregular nuclear contours, often nuclear hyperchromasia, inconspicuous nucleoli, and inconspicuous or scant cytoplasm) Lower 
High-grade CMIL PAM with moderate to severe atypiac-MIN (grade 5–10) Yes More confluent basilar and significant non-basilar proliferation of melanocytes with high-grade atypia (moderate to severe), evidence of intraepithelial nested and/or pagetoid growth, and epithelioid cell cytomorphology Higher 
Melanoma in situ Yes The term melanoma in situ may be used for (1) the most atypical high-grade CMILs involving close to full thickness of the epithelium, (2) histologically obvious melanomas without documented evidence of subepithelial invasion Highest 

CMIL, conjunctival melanocytic intraepithelial lesion.

Fig. 2.

Adenosquamous carcinoma of the conjunctiva. a Leukoplakic mass in the inferior fornix. b The invasive adenocarcinoma component is composed of epithelial cells with focal intracytoplasmic mucin (arrow) arranged in glands, in a background of desmoplastic stroma. Intracytoplasmic and luminal mucin is highlighted with Alcian blue stain (inset). c Sheets of atypical goblet cells (arrow). d Squamous cell carcinoma component is composed of irregular islands and bands of squamous cells [hematoxylin-eosin (b-d); ×100 (b), ×200 (c, d)].

Fig. 2.

Adenosquamous carcinoma of the conjunctiva. a Leukoplakic mass in the inferior fornix. b The invasive adenocarcinoma component is composed of epithelial cells with focal intracytoplasmic mucin (arrow) arranged in glands, in a background of desmoplastic stroma. Intracytoplasmic and luminal mucin is highlighted with Alcian blue stain (inset). c Sheets of atypical goblet cells (arrow). d Squamous cell carcinoma component is composed of irregular islands and bands of squamous cells [hematoxylin-eosin (b-d); ×100 (b), ×200 (c, d)].

Close modal
Fig. 3.

Squamous cell carcinoma with mucinous differentiation. a Irregular islands and bands of invasive nonkeratinizing squamous cell carcinoma with focal intracytoplasmic mucin (b), highlighted with Alcian blue stain (c) [hematoxylin-eosin. ×50 (b), ×400 (b, c)].

Fig. 3.

Squamous cell carcinoma with mucinous differentiation. a Irregular islands and bands of invasive nonkeratinizing squamous cell carcinoma with focal intracytoplasmic mucin (b), highlighted with Alcian blue stain (c) [hematoxylin-eosin. ×50 (b), ×400 (b, c)].

Close modal

Tumors of the Eyelid (Table 6)

This chapter covers common benign and malignant eyelid tumors with attention to their site-specific features. Rare adnexal tumors with predilection for the eyelid, such as sebaceous neoplasms and endocrine mucin-producing sweat gland carcinoma (shown in Fig. 4), and lesions that are unique to the eyelid, such as intratarsal keratinous cyst (shown in Fig. 5a–c) and phakomatous choristoma (shown in Fig. 5d–f), are also illuminated in this chapter [31‒36]. The material is coordinated with the 5th edition WHO Classification of the Tumours of the Skin, particularly with regard to cutaneous melanocytic tumors.

Fig. 4.

Endocrine mucin-producing sweat gland carcinoma of the eyelid. a Reddish dermal-based nodule in the lower eyelid along the eyelash line. b Expansile nests of cells in cribriform arrangement. c Cuboidal neoplastic cells have ovoid, mildly pleomorphic nuclei, inconspicuous nucleoli, and moderate amounts of eosinophilic cytoplasm. d Alcian blue stain highlights intracytoplasmic and extracellular mucin. e Strong diffuse nuclear expression of estrogen receptors and insulinoma-associated protein 1 (INSM1) (f) [hematoxylin-eosin (b, c), Alcian blue (d), ER (e), INSM1 (f), ×50 (b), ×200 (c-f)].

Fig. 4.

Endocrine mucin-producing sweat gland carcinoma of the eyelid. a Reddish dermal-based nodule in the lower eyelid along the eyelash line. b Expansile nests of cells in cribriform arrangement. c Cuboidal neoplastic cells have ovoid, mildly pleomorphic nuclei, inconspicuous nucleoli, and moderate amounts of eosinophilic cytoplasm. d Alcian blue stain highlights intracytoplasmic and extracellular mucin. e Strong diffuse nuclear expression of estrogen receptors and insulinoma-associated protein 1 (INSM1) (f) [hematoxylin-eosin (b, c), Alcian blue (d), ER (e), INSM1 (f), ×50 (b), ×200 (c-f)].

Close modal
Fig. 5.

Tarsal cyst and phakomatous choristoma. a–c Tarsal cyst. a Tan-gray subepithelial tarsal-based nodules. b The cyst is lined by stratified squamous epithelium with eosinophilic corrugated delicately keratinized cuticle (arrow) and filled with keratin (asterisk). Meibomian gland lobules with associated ducts, lined by the epithelium morphologically similar to the cyst (arrowhead), are present in the adjacent tarsus. c Higher magnification of the cyst lining and delicate lamellated luminal keratin. d–f Phakomatous choristoma. d Cataractous-appearing tissue composed of bladder cells (arrowheads) and cuboidal lens epithelium associated with a prominent periodic acid-Schiff (PAS)-positive basement membrane, reminiscent of lens capsule (arrow). The lens epithelial cells express nuclear PAX8 (e) and cytoplasmic S100 (f) [hematoxylin-eosin (b, c), PAS (d), PAX8 (e), S100 (f), ×100 (b), ×1,000 (c), ×400 (c-f)].

Fig. 5.

Tarsal cyst and phakomatous choristoma. a–c Tarsal cyst. a Tan-gray subepithelial tarsal-based nodules. b The cyst is lined by stratified squamous epithelium with eosinophilic corrugated delicately keratinized cuticle (arrow) and filled with keratin (asterisk). Meibomian gland lobules with associated ducts, lined by the epithelium morphologically similar to the cyst (arrowhead), are present in the adjacent tarsus. c Higher magnification of the cyst lining and delicate lamellated luminal keratin. d–f Phakomatous choristoma. d Cataractous-appearing tissue composed of bladder cells (arrowheads) and cuboidal lens epithelium associated with a prominent periodic acid-Schiff (PAS)-positive basement membrane, reminiscent of lens capsule (arrow). The lens epithelial cells express nuclear PAX8 (e) and cytoplasmic S100 (f) [hematoxylin-eosin (b, c), PAS (d), PAX8 (e), S100 (f), ×100 (b), ×1,000 (c), ×400 (c-f)].

Close modal
Table 6.

WHO Eye5: tumors of the eyelid [2]

Tumor-like lesions, hamartomas, and choristomas of the eyelid 
Eyelid cysts 
Epidermal inclusion cyst 
Intratarsal cyst 
Choristomas of the eyelid 
Phakomatous choristoma 
Epithelial tumors of the eyelid 
Benign and premalignant epithelial tumors of the eyelid 
Molluscum contagiosum 
Verruca vulgaris 
Squamous papilloma 
Seborrheic keratosis 
Inverted follicular keratosis 
Actinic keratosis 
Malignant epithelial tumors of the eyelid 
Basal cell carcinoma 
Squamous cell carcinoma 
Merkel cell carcinoma 
Adnexal tumors of the eyelid 
Benign adnexal tumors of the eyelid 
Ductal hidrocystoma 
Sebaceous hyperplasia, adenoma, and sebaceoma 
Syringoma 
Mixed tumor 
Tubular adenoma 
Syringocystadenoma papilliferum 
Pilomatrixoma 
Trichilemmoma 
Malignant adnexal tumors of the eyelid 
Sebaceous carcinoma 
Adenoid cystic carcinoma 
Signet-ring cell/histiocytoid carcinoma 
Mucinous carcinoma 
Endocrine mucin-producing sweat gland carcinoma 
Microcystic adnexal carcinoma 
Apocrine adenocarcinoma 
Melanocytic eyelid tumors 
Benign and premalignant melanocytic eyelid tumors 
Nevi of the eyelid 
Spitz naevus 
In situ cutaneous melanoma 
Malignant melanocytic eyelid tumors 
Invasive cutaneous melanoma 
Tumor-like lesions, hamartomas, and choristomas of the eyelid 
Eyelid cysts 
Epidermal inclusion cyst 
Intratarsal cyst 
Choristomas of the eyelid 
Phakomatous choristoma 
Epithelial tumors of the eyelid 
Benign and premalignant epithelial tumors of the eyelid 
Molluscum contagiosum 
Verruca vulgaris 
Squamous papilloma 
Seborrheic keratosis 
Inverted follicular keratosis 
Actinic keratosis 
Malignant epithelial tumors of the eyelid 
Basal cell carcinoma 
Squamous cell carcinoma 
Merkel cell carcinoma 
Adnexal tumors of the eyelid 
Benign adnexal tumors of the eyelid 
Ductal hidrocystoma 
Sebaceous hyperplasia, adenoma, and sebaceoma 
Syringoma 
Mixed tumor 
Tubular adenoma 
Syringocystadenoma papilliferum 
Pilomatrixoma 
Trichilemmoma 
Malignant adnexal tumors of the eyelid 
Sebaceous carcinoma 
Adenoid cystic carcinoma 
Signet-ring cell/histiocytoid carcinoma 
Mucinous carcinoma 
Endocrine mucin-producing sweat gland carcinoma 
Microcystic adnexal carcinoma 
Apocrine adenocarcinoma 
Melanocytic eyelid tumors 
Benign and premalignant melanocytic eyelid tumors 
Nevi of the eyelid 
Spitz naevus 
In situ cutaneous melanoma 
Malignant melanocytic eyelid tumors 
Invasive cutaneous melanoma 

Tumors of the Uveal Tract (Table 7)

This chapter is limited to the non-neoplastic iris lesions and uveal melanocytic tumors and tumor-like lesions. Entities that can involve any portion of the uveal tract are described according to their dominant site. The most notable update is addition of diagnostic cytopathology for benign and malignant melanocytic tumors of the iris, ciliary body, and choroid (Table 8) [2, 37‒42]. This chapter provides an update on pathogenesis of uveal melanocytoma, which harbors solitary GNAQ mutations without additional oncogenic alterations and thus distinct from cutaneous and conjunctival melanocytoma [43], and that of bilateral diffuse uveal melanocytic proliferation, which may involve autoantibodies to hepatocyte growth factor [44, 45]. Soft tissue tumors of the uveal tract, including hemangioma, peripheral nerve sheath tumors, and smooth muscle tumors are covered in chapters dedicated to these entities.

Table 7.

WHO Eye5: tumors of the uveal tract [2]

Iris tumors 
Benign lesions and hamartomas of the iris 
Ectopic tissue 
Implantation cyst 
Pigmented epithelial cyst 
Lisch nodule 
Melanocytic tumors of the iris 
Ocular melanocytosis 
Melanocytic nevus 
Iris melanoma 
Ciliary body and choroid tumors 
Benign melanocytic lesions of the choroid and ciliary body 
Choroidal and ciliary body nevi 
Uveal melanocytoma 
BDUMP 
Malignant melanocytic tumors of the choroid and ciliary body 
Choroidal and ciliary body melanomas 
Iris tumors 
Benign lesions and hamartomas of the iris 
Ectopic tissue 
Implantation cyst 
Pigmented epithelial cyst 
Lisch nodule 
Melanocytic tumors of the iris 
Ocular melanocytosis 
Melanocytic nevus 
Iris melanoma 
Ciliary body and choroid tumors 
Benign melanocytic lesions of the choroid and ciliary body 
Choroidal and ciliary body nevi 
Uveal melanocytoma 
BDUMP 
Malignant melanocytic tumors of the choroid and ciliary body 
Choroidal and ciliary body melanomas 

BDUMP, bilateral diffuse uveal melanocytic proliferation.

Table 8.

WHO Eye5: diagnostic cytology of melanocytic uveal tumors [2]

Uveal melanocytic tumorDiagnostic cytologyReferences
Conventional nevus Paucicellular sample with predominantly small spindle to round cells with or without pigmentation and with small or absent nucleoli. The cells are immunoreactive for melanocytic markers and show rare or no cells in cycle with Ki-67 immunostaining. Molecular genetic profiling may allow differentiation from melanoma in cytologically borderline lesions [2] 
Melanocytoma Large, ovoid cells with small centrally located nuclei and heavily pigmented cytoplasm. Melanin can obscure nuclear detail. The cells are immunoreactive for melanocytic markers and show rare or no cells in cycle with Ki-67 immunostaining. Molecular genetic profiling may allow differentiation from melanoma in cytologically borderline lesions [37] 
Melanoma Individually dispersed, or small cohesive groups of atypical cells demonstrating spindle-shaped or rounded pleomorphic nuclei with nucleoli, coarsely hyperchromatic chromatin, and eosinophilic cytoplasm that is spindle-shaped in spindle melanoma cells and ovoid in epithelioid melanoma cells. Variable numbers of cytoplasmic finely granular melanosomes are present. Binucleation and mitotic figures can be seen. Molecular genetic profiling of cytology material plays an important role in prognostication [38–42] 
Uveal melanocytic tumorDiagnostic cytologyReferences
Conventional nevus Paucicellular sample with predominantly small spindle to round cells with or without pigmentation and with small or absent nucleoli. The cells are immunoreactive for melanocytic markers and show rare or no cells in cycle with Ki-67 immunostaining. Molecular genetic profiling may allow differentiation from melanoma in cytologically borderline lesions [2] 
Melanocytoma Large, ovoid cells with small centrally located nuclei and heavily pigmented cytoplasm. Melanin can obscure nuclear detail. The cells are immunoreactive for melanocytic markers and show rare or no cells in cycle with Ki-67 immunostaining. Molecular genetic profiling may allow differentiation from melanoma in cytologically borderline lesions [37] 
Melanoma Individually dispersed, or small cohesive groups of atypical cells demonstrating spindle-shaped or rounded pleomorphic nuclei with nucleoli, coarsely hyperchromatic chromatin, and eosinophilic cytoplasm that is spindle-shaped in spindle melanoma cells and ovoid in epithelioid melanoma cells. Variable numbers of cytoplasmic finely granular melanosomes are present. Binucleation and mitotic figures can be seen. Molecular genetic profiling of cytology material plays an important role in prognostication [38–42] 

Tumors of the Retina and Neuroepithelium (Table 9)

This chapter is devoted to the tumors and tumor-like lesions of the retina, retinal pigment epithelium, and the iris and ciliary body epithelia. Retinocytoma is classified as a premalignant tumor, considering the genetic data that these tumors carry biallelic RB1 mutations and clinical evidence that they can dedifferentiate into retinoblastoma [45, 46]. The chapter includes recent consensus protocols on screening of children at risk for retinoblastoma [47, 48] and data on MYCN-amplified, RB1 wild-type retinoblastoma (shown in Fig. 6) [49]. Novel molecular genetic methods, like liquid biopsies of tumor-derived cell-free DNA from aqueous fluid for prognostication and from plasma for prenatal testing and detection of nonocular primary malignancies in children with retinoblastoma, are discussed [50‒52]. Coverage of diagnostic cytopathology for tumors of the retina and neuroepithelium is a notable addition (Table 10) [2, 53‒60]. The spectrum of nodular and massive gliosis is described, nodular gliosis being the preferred term for vasoproliferative tumor [2]. True vascular lesions of the retina are covered in the chapters dedicated to vascular malformations and vascular neoplasms.

Table 9.

WHO Eye5: tumors of the retina and neuroepithelium [2]

Tumors of the neurosensory retina 
Benign retinal tumors 
Astrocytic tumors 
Nodular and massive retinal gliosis 
Premalignant and malignant retinal tumors 
Retinocytoma 
Retinoblastoma 
Tumors of the retinal pigment epithelium (RPE) 
Reactive and hamartomatous lesions of the RPE 
Hamartoma of the RPE 
Congenital hypertrophy of the RPE 
Reactive hyperplasia of the RPE 
Benign and malignant tumors of the RPE 
Adenoma and adenocarcinoma of the RPE 
Tumors of the iris and ciliary body neuroepithelium 
Reactive and cystic lesions of the iris and ciliary body pigment epithelium 
Reactive epithelial hyperplasia of the ciliary epithelium 
Nodular hyperplasia of the ciliary epithelium 
Glioneuronal hamartoma of the ciliary body 
Benign and malignant tumors of the iris and ciliary body pigment epithelium 
Adenoma and adenocarcinoma of the ciliary body 
Medulloepithelioma of the ciliary body 
Tumors of the neurosensory retina 
Benign retinal tumors 
Astrocytic tumors 
Nodular and massive retinal gliosis 
Premalignant and malignant retinal tumors 
Retinocytoma 
Retinoblastoma 
Tumors of the retinal pigment epithelium (RPE) 
Reactive and hamartomatous lesions of the RPE 
Hamartoma of the RPE 
Congenital hypertrophy of the RPE 
Reactive hyperplasia of the RPE 
Benign and malignant tumors of the RPE 
Adenoma and adenocarcinoma of the RPE 
Tumors of the iris and ciliary body neuroepithelium 
Reactive and cystic lesions of the iris and ciliary body pigment epithelium 
Reactive epithelial hyperplasia of the ciliary epithelium 
Nodular hyperplasia of the ciliary epithelium 
Glioneuronal hamartoma of the ciliary body 
Benign and malignant tumors of the iris and ciliary body pigment epithelium 
Adenoma and adenocarcinoma of the ciliary body 
Medulloepithelioma of the ciliary body 
Fig. 6.

MYCN-amplified retinoblastoma. a Gross photograph of the eye enucleated for failure to respond to local therapy demonstrates calcific post-treatment regression scar (arrow) and diffuse involvement of the detached retina by viable-appearing tumor (black arrowheads). Focal choroidal invasion is present (white arrowhead). Hemorrhage and necrosis are present in subretinal space (asterisk). b Histopathology demonstrates discohesive undifferentiated retinoblastoma cells, which focally invade choroid (arrow). c Discohesive retinoblastoma cells with prominent central nucleoli and scant cytoplasm (arrow), with brisk apoptotic bodies. d Retained retinoblastoma 1 (RB1) protein expression in the tumor [hematoxylin-eosin (b, c), RB1 (d), ×100 (b), ×400 (C), ×200 (d)]. Images courtesy of Ralph C. Eagle, Jr. MD.

Fig. 6.

MYCN-amplified retinoblastoma. a Gross photograph of the eye enucleated for failure to respond to local therapy demonstrates calcific post-treatment regression scar (arrow) and diffuse involvement of the detached retina by viable-appearing tumor (black arrowheads). Focal choroidal invasion is present (white arrowhead). Hemorrhage and necrosis are present in subretinal space (asterisk). b Histopathology demonstrates discohesive undifferentiated retinoblastoma cells, which focally invade choroid (arrow). c Discohesive retinoblastoma cells with prominent central nucleoli and scant cytoplasm (arrow), with brisk apoptotic bodies. d Retained retinoblastoma 1 (RB1) protein expression in the tumor [hematoxylin-eosin (b, c), RB1 (d), ×100 (b), ×400 (C), ×200 (d)]. Images courtesy of Ralph C. Eagle, Jr. MD.

Close modal
Table 10.

WHO Eye5: diagnostic cytology of tumors of the retina and neuroepithelium [2]

Retinal and neuroepithelial tumorDiagnostic cytologyReferences
Retinoblastoma Generally, biopsy is avoided due to the risk of extraocular recurrence and metastasis. Cytological samples often have pitfalls (sampling of retinal neurons, scant material) that may be misleading. Diagnostic features of malignancy such as necrosis, mitoses, and nuclear atypia should be present when diagnosing retinoblastoma in a cytology. Consultation with an experienced ophthalmic cytopathologist is recommended if a biopsy is considered [2] 
Medulloepithelioma The polymorphous nature of medulloepithelioma can reduce the sensitivity and specificity of aspiration cytology. In many reports, diagnosis is limited to a malignant round cell tumor, although more specific rosettes and focal epithelial-like fragments with cellular polarity and multilayering occasionally are observed. Immunohistochemical staining for medulloepitheliomas expresses LIN28A and PAX8 by immunohistochemistry (IHC), which may help distinguish these tumors from retinoblastoma [53–57] 
Adenoma and adenocarcinoma of the ciliary body epithelium (CBE) CBE adenoma shows polygonal epithelial cells with bland round to oval nuclei and small nucleoli, with or without melanin granules. Adenocarcinoma demonstrates cohesive clusters of atypical cuboidal to large epithelioid epithelial cells with hyperchromatic, enlarged nuclei, and prominent nucleoli. The cells in adenoma and adenocarcinoma can be pigmented or non-pigmented. The presence of basement membrane material highlighted with periodic acid-Schiff (PAS) stain helps differentiate these tumors from melanoma, other stromal tumors, and metastases. PAX8 IHC is useful in distinguishing CBE adenoma and adenocarcinoma from melanocytic lesions. Adenomas and adenocarcinomas of pigmented CBE harbor BRAF c.1799T>A p.V600E mutations and lack GNAQ or GNA11 mutations (histopathology data), a profile that may favor adenoma/adenocarcinoma over melanocytic uveal neoplasm [57–59] 
Adenoma and adenocarcinoma of retinal pigment epithelium (RPE) Adenocarcinoma of RPE and epithelioid melanoma cells can appear similar, and there is significant immunohistochemical overlap between RPE tumors and uveal melanoma. Documentation of PAS-positive basement material can be helpful in supporting the diagnosis of retinal pigment epithelial neoplasm. Correlation of cytologic findings with clinical and imaging findings is essential [57], [60] 
Retinal and neuroepithelial tumorDiagnostic cytologyReferences
Retinoblastoma Generally, biopsy is avoided due to the risk of extraocular recurrence and metastasis. Cytological samples often have pitfalls (sampling of retinal neurons, scant material) that may be misleading. Diagnostic features of malignancy such as necrosis, mitoses, and nuclear atypia should be present when diagnosing retinoblastoma in a cytology. Consultation with an experienced ophthalmic cytopathologist is recommended if a biopsy is considered [2] 
Medulloepithelioma The polymorphous nature of medulloepithelioma can reduce the sensitivity and specificity of aspiration cytology. In many reports, diagnosis is limited to a malignant round cell tumor, although more specific rosettes and focal epithelial-like fragments with cellular polarity and multilayering occasionally are observed. Immunohistochemical staining for medulloepitheliomas expresses LIN28A and PAX8 by immunohistochemistry (IHC), which may help distinguish these tumors from retinoblastoma [53–57] 
Adenoma and adenocarcinoma of the ciliary body epithelium (CBE) CBE adenoma shows polygonal epithelial cells with bland round to oval nuclei and small nucleoli, with or without melanin granules. Adenocarcinoma demonstrates cohesive clusters of atypical cuboidal to large epithelioid epithelial cells with hyperchromatic, enlarged nuclei, and prominent nucleoli. The cells in adenoma and adenocarcinoma can be pigmented or non-pigmented. The presence of basement membrane material highlighted with periodic acid-Schiff (PAS) stain helps differentiate these tumors from melanoma, other stromal tumors, and metastases. PAX8 IHC is useful in distinguishing CBE adenoma and adenocarcinoma from melanocytic lesions. Adenomas and adenocarcinomas of pigmented CBE harbor BRAF c.1799T>A p.V600E mutations and lack GNAQ or GNA11 mutations (histopathology data), a profile that may favor adenoma/adenocarcinoma over melanocytic uveal neoplasm [57–59] 
Adenoma and adenocarcinoma of retinal pigment epithelium (RPE) Adenocarcinoma of RPE and epithelioid melanoma cells can appear similar, and there is significant immunohistochemical overlap between RPE tumors and uveal melanoma. Documentation of PAS-positive basement material can be helpful in supporting the diagnosis of retinal pigment epithelial neoplasm. Correlation of cytologic findings with clinical and imaging findings is essential [57], [60] 

Tumors of the Optic Disk and Optic Nerve (Table 11)

This chapter focuses on primary lesions of the optic disk and pre-chiasmal optic nerve. Optic nerve choristoma, a rare, benign, developmental/malformative lesion composed of smooth muscle and adipose tissue within and around the optic nerve, is added to this chapter [2, 61, 62] that is coordinated with the 5th edition of WHO Classification of the Tumours of the Central Nervous System [63].

Table 11.

WHO Eye5: tumors of the optic disk and optic nerve [2]

Malformations of the optic disk and optic nerve
Optic nerve choristoma 
Primary neural tumors of the optic disk and optic nerve 
Gliomas of the optic nerve 
Pigment epithelial tumors of the optic disk and optic nerve 
Medulloepithelioma of the optic disk and optic nerve 
Meningeal tumors of the optic nerve 
Meningioma of the optic nerve 
Melanocytic tumors of the optic disk and optic nerve 
Melanocytoma of the optic disk and optic nerve 
Malformations of the optic disk and optic nerve
Optic nerve choristoma 
Primary neural tumors of the optic disk and optic nerve 
Gliomas of the optic nerve 
Pigment epithelial tumors of the optic disk and optic nerve 
Medulloepithelioma of the optic disk and optic nerve 
Meningeal tumors of the optic nerve 
Meningioma of the optic nerve 
Melanocytic tumors of the optic disk and optic nerve 
Melanocytoma of the optic disk and optic nerve 

Tumors of the Lacrimal Gland and Lacrimal Drainage System (Table 12)

Two separate chapters provide an update regarding molecular biology and diagnosis of benign and malignant epithelial lacrimal gland (Table 13) and lacrimal drainage system tumors and include new sections on diagnostic cytopathology [64‒106]. Obsolete entities, such as oncocytic carcinoma, have been removed. Tumors that are rarely encountered in the lacrimal gland or are controversial in this region, such as Warthin tumor, are also removed.

Table 12.

WHO Eye5: tumors of the lacrimal gland and lacrimal drainage system [2]

Tumors of the lacrimal gland 
Epithelial tumors of the lacrimal gland 
Cysts of the lacrimal gland 
Dacryops cyst 
Benign and premalignant epithelial tumors of the lacrimal gland 
Pleomorphic adenoma (PA) of the lacrimal gland 
Lacrimal gland oncocytoma 
Myoepithelioma 
Intraductal carcinoma 
Malignant epithelial tumors of the lacrimal gland 
Carcinoma ex PA 
Adenoid cystic carcinoma 
Primary ductal adenocarcinoma of the lacrimal gland 
Secretory carcinoma 
Mucoepidermoid carcinoma (MEC) 
Epithelial-myoepithelial carcinoma (EMC) 
Acinic cell carcinoma 
Adenocarcinoma, NOS 
Tumors of the lacrimal drainage system 
Epithelial tumors of the lacrimal drainage system 
Benign and premalignant epithelial tumors of the lacrimal drainage system Papillomas (squamous and inverted) 
Malignant epithelial tumors of the lacrimal drainage system 
Squamous cell carcinoma 
Lymphoepithelial carcinoma 
MEC 
Other salivary gland-type carcinomas of the lacrimal drainage system 
Adenocarcinoma NOS of the lacrimal drainage system 
Melanocytic tumors of the lacrimal drainage system 
Melanoma involving the lacrimal drainage system 
Tumors of the lacrimal gland 
Epithelial tumors of the lacrimal gland 
Cysts of the lacrimal gland 
Dacryops cyst 
Benign and premalignant epithelial tumors of the lacrimal gland 
Pleomorphic adenoma (PA) of the lacrimal gland 
Lacrimal gland oncocytoma 
Myoepithelioma 
Intraductal carcinoma 
Malignant epithelial tumors of the lacrimal gland 
Carcinoma ex PA 
Adenoid cystic carcinoma 
Primary ductal adenocarcinoma of the lacrimal gland 
Secretory carcinoma 
Mucoepidermoid carcinoma (MEC) 
Epithelial-myoepithelial carcinoma (EMC) 
Acinic cell carcinoma 
Adenocarcinoma, NOS 
Tumors of the lacrimal drainage system 
Epithelial tumors of the lacrimal drainage system 
Benign and premalignant epithelial tumors of the lacrimal drainage system Papillomas (squamous and inverted) 
Malignant epithelial tumors of the lacrimal drainage system 
Squamous cell carcinoma 
Lymphoepithelial carcinoma 
MEC 
Other salivary gland-type carcinomas of the lacrimal drainage system 
Adenocarcinoma NOS of the lacrimal drainage system 
Melanocytic tumors of the lacrimal drainage system 
Melanoma involving the lacrimal drainage system 
Table 13.

WHO Eye5: epithelial tumors of the lacrimal gland [2]

Epithelial tumor of the lacrimal gland/DefinitionMolecular genetic alterations/Diagnostic molecular pathologyImmunohistochemistry (IHC)Essential and desirable diagnostic criteriaRef
Pleomorphic adenoma (PA)Benign lacrimal gland neoplasm consisting of a mixture of epithelial, myoepithelial, and mesenchymal-appearing components Recurrent translocations ofPLAG1 on 8q12PLAG1 translocation t(5; 8) (p13; q12) is highly specific for PAHMGA2 translocation on 12q14 less frequentDiagnostic molecular pathologyUsually not required Epithelial cells: CK7+, CAM5.2+Myoepithelial cells: p40+, p63+, SMA+, calponin+Ki67 lowPLAG1 (nuclear)+GFAP +/- Essential: admixture of bilayered ducts, myoepithelial cells, and chondromyxoid/fibrous stroma in the absence of invasion and malignant cytomorphological featuresDesirable:PLAG1 or HMGA2 alteration (by IHC or molecular methods) in select cases [64–69, 73] 
Carcinoma ex PAMalignant lacrimal gland neoplasm that shows presence of an epithelial and/or myoepithelial malignancy arising in association with primary or recurrent PA PLAG1 and HMGA2 rearrangements identical to PASequential loss of heterozygosity of 8q, 12q, 17pAlterations in 12q genes (HMGIC, HMGA2, MDM2)Copy number alterations in 9p and 22q, resulting in activation of NFIB and PDGFBActivation of IL6/JAK/STAT3Diagnostic molecular pathologyPLAG1 or HMGA2 rearrangements serve as markers of pre-existing PA, differentiating from de novo lacrimal gland carcinomas Carcinoma areas: p53+, c-MYC+, Ki-67 highPA component: PLAG1+ and HMGA2+Androgen receptor (AR)+ in carcinoma component may indicate lacrimal duct carcinoma differentiation Essential: histological areas compatible with PA or history of previous PA in the same site; presence of an epithelial and/or myoepithelial malignant component [70–74] 
Adenoid cystic carcinomaMalignant lacrimal gland neoplasm composed of epithelial and myoepithelial cells (EMCs) arranged in tubular, cribriform, and solid patterns associated with a basophilic matrix and reduplicated basement membrane material, frequently accompanied by MYB::NFIB rearrangements t(6; 9) (q22-23; p23-24) (up to 50%) results in MYB::NFIB fusion and activation of the MYB oncogene and associated target genes such as KIT and BCL2 which are involved in cell growth, apoptosis, transcription, and cell cycle regulationLoss of 6q, 12q, and 17pGains of 19q, 8q, and 11qAberrant NOTCH signalingKRAS, NRAS, Met mutationsDiagnostic molecular pathologyMYB rearrangement in select cases MYB+ (although less sensitive, may be used as a surrogate for MYB rearrangement in select cases)KIT+p53 overexpressionMean Ki67 index 30% Essential: a malignant biphasic tumor composed of proliferating myoepithelial cells and luminal ductal cellsDesirable: perineural invasion. Positive MYB, KIT IHC, or MYB rearrangement in challenging cases [66, 75–79, 96] 
Primary ductal adenocarcinoma of the lacrimal glandHigh-grade adenocarcinoma and a counterpart of salivary duct carcinoma, resembling invasive ductal carcinoma of the breast Unknown, although at least a subset of cases show HER2 overexpression by IHC and/or HER2(ERBB2) gene amplification similar to salivary duct carcinomaDiagnostic molecular pathologyHER2 amplification in select cases AE1/AE3+, CK7+, CK19+, EMA+, AR+, GCDFP-15+, HER-2/neu+, p53+, cyclin D1+Usually SMA-, p63-, calponin-, ER-, PR-, PSA-Ki-67 up to 70% Essential: an invasive adenocarcinoma with ductal differentiation, arising de novo (not arising from a benign tumor such as PA) in the lacrimal glandDesirable: tumor comedonecrosis [80–84, 96] 
Secretory carcinomaRare monophasic lacrimal gland carcinoma characterized by a rearrangement of the ETV6 gene resulting in ETV6::NTRK3 fusion in most cases Lacrimal gland: ETV6 rearrangement resulting in ETV6::NTRK3 fusionSalivary glands: alternative ETV6 fusion partners include RET, MET, MAML3Diagnostic molecular pathologyETV6 gene rearrangement can be demonstrated by breaking apart FISH. Detection of NTRK3 fusion by RT-PCR or RNA sequencing may be predictive of response to NTRK inhibitors for unresectable or metastatic cases S100+, SOX10+, mammaglobin+, CK18+, Cam5.2+, GCDFP-15+, NTRK+ (or pan-TRK+)CK5-, p63-, DOG-1, AR-Ki-67 < 5% Essential: monophasic tumor with vacuolated secretory material, lacking cytoplasmic granules; IHC positivity for S100 protein, mammaglobin, and NTRK (or Pan-TRK) and negativity for p40/p63, and CK5Desirable:ETV6 and/or NTRK3 gene rearrangement [85–96] 
Mucoepidermoid carcinoma (MEC)Malignant lacrimal gland neoplasm characterized by squamous (epidermoid) cells, mucous cells, columnar cells, and intermediate cells CTRC1::MAML2 gene fusion is present in majority lacrimal gland MEC and is considered a key oncogenic driver leading to EGFR upregulationRAS/PIK3 mutations in poorly differentiated tumors involved in EGFR upregulationDiagnostic molecular pathologyMAML2 rearrangement/CTRC1::MAML2 fusion in select cases P63+, p40+S100-, SOX10- Essential: squamous cells, intermediate cells, and mucous cells in varying proportionsLower grade tumors: cystic, mucous cell rich, and well circumscribedHigher grade tumors: solid, higher proportion of squamous and intermediate cellsDesirable:MAML2 rearrangement/CRTC1::MAML2 fusion [96–98] 
EMCLow-grade malignant biphasic tumor of the lacrimal gland characterized by tubular structures, usually composed of tightly coupled inner ductal and prominent outer myoepithelial cells HRAS mutations, most commonly at the codon 61Diagnostic molecular pathologyAssessment of HRAS mutations is useful for diagnosing EMC and excluding its mimics; MYB fusion by molecular methods/IHC might help in differentiation from adenoid cystic carcinoma and hybrid tumors Inner cells: panCK+, CK7+Outer myoepithelial cells: p63+, SMA+, calponin+, S100+Diffuse membranous and cytoplasmic RAS Q61R expression, almost always restricted to myoepithelial cells in 65% of EMCs Essential: usually multinodular invasive growth; at least partly with a dual arrangement of inner ductal cells and outer prominent, and usually clear, myoepithelial cells [99–103] 
Acinic cell carcinomaRare malignant lacrimal gland neoplasm displaying predominantly serous acinar differentiation Salivary gland tumors: recurrent rearrangement [t(4; 9) (q13; q31)] leads to NR4A3 upregulationDiagnostic molecular pathologyNR4A subfamily rearrangements by molecular or IHC methods in challenging cases NR4A3+ or NR4A2+DOG1+, SOX10+p63-, S100-Mammaglobin-, pan-TRK- Essential: tumor cells with acinar differentiation; zymogen granules in focal areasDesirable: nuclear staining for NR4A3/NOR-1 or NR4A2/Nuur1Molecular demonstration of NR4A3 rearrangement [104–106] 
Epithelial tumor of the lacrimal gland/DefinitionMolecular genetic alterations/Diagnostic molecular pathologyImmunohistochemistry (IHC)Essential and desirable diagnostic criteriaRef
Pleomorphic adenoma (PA)Benign lacrimal gland neoplasm consisting of a mixture of epithelial, myoepithelial, and mesenchymal-appearing components Recurrent translocations ofPLAG1 on 8q12PLAG1 translocation t(5; 8) (p13; q12) is highly specific for PAHMGA2 translocation on 12q14 less frequentDiagnostic molecular pathologyUsually not required Epithelial cells: CK7+, CAM5.2+Myoepithelial cells: p40+, p63+, SMA+, calponin+Ki67 lowPLAG1 (nuclear)+GFAP +/- Essential: admixture of bilayered ducts, myoepithelial cells, and chondromyxoid/fibrous stroma in the absence of invasion and malignant cytomorphological featuresDesirable:PLAG1 or HMGA2 alteration (by IHC or molecular methods) in select cases [64–69, 73] 
Carcinoma ex PAMalignant lacrimal gland neoplasm that shows presence of an epithelial and/or myoepithelial malignancy arising in association with primary or recurrent PA PLAG1 and HMGA2 rearrangements identical to PASequential loss of heterozygosity of 8q, 12q, 17pAlterations in 12q genes (HMGIC, HMGA2, MDM2)Copy number alterations in 9p and 22q, resulting in activation of NFIB and PDGFBActivation of IL6/JAK/STAT3Diagnostic molecular pathologyPLAG1 or HMGA2 rearrangements serve as markers of pre-existing PA, differentiating from de novo lacrimal gland carcinomas Carcinoma areas: p53+, c-MYC+, Ki-67 highPA component: PLAG1+ and HMGA2+Androgen receptor (AR)+ in carcinoma component may indicate lacrimal duct carcinoma differentiation Essential: histological areas compatible with PA or history of previous PA in the same site; presence of an epithelial and/or myoepithelial malignant component [70–74] 
Adenoid cystic carcinomaMalignant lacrimal gland neoplasm composed of epithelial and myoepithelial cells (EMCs) arranged in tubular, cribriform, and solid patterns associated with a basophilic matrix and reduplicated basement membrane material, frequently accompanied by MYB::NFIB rearrangements t(6; 9) (q22-23; p23-24) (up to 50%) results in MYB::NFIB fusion and activation of the MYB oncogene and associated target genes such as KIT and BCL2 which are involved in cell growth, apoptosis, transcription, and cell cycle regulationLoss of 6q, 12q, and 17pGains of 19q, 8q, and 11qAberrant NOTCH signalingKRAS, NRAS, Met mutationsDiagnostic molecular pathologyMYB rearrangement in select cases MYB+ (although less sensitive, may be used as a surrogate for MYB rearrangement in select cases)KIT+p53 overexpressionMean Ki67 index 30% Essential: a malignant biphasic tumor composed of proliferating myoepithelial cells and luminal ductal cellsDesirable: perineural invasion. Positive MYB, KIT IHC, or MYB rearrangement in challenging cases [66, 75–79, 96] 
Primary ductal adenocarcinoma of the lacrimal glandHigh-grade adenocarcinoma and a counterpart of salivary duct carcinoma, resembling invasive ductal carcinoma of the breast Unknown, although at least a subset of cases show HER2 overexpression by IHC and/or HER2(ERBB2) gene amplification similar to salivary duct carcinomaDiagnostic molecular pathologyHER2 amplification in select cases AE1/AE3+, CK7+, CK19+, EMA+, AR+, GCDFP-15+, HER-2/neu+, p53+, cyclin D1+Usually SMA-, p63-, calponin-, ER-, PR-, PSA-Ki-67 up to 70% Essential: an invasive adenocarcinoma with ductal differentiation, arising de novo (not arising from a benign tumor such as PA) in the lacrimal glandDesirable: tumor comedonecrosis [80–84, 96] 
Secretory carcinomaRare monophasic lacrimal gland carcinoma characterized by a rearrangement of the ETV6 gene resulting in ETV6::NTRK3 fusion in most cases Lacrimal gland: ETV6 rearrangement resulting in ETV6::NTRK3 fusionSalivary glands: alternative ETV6 fusion partners include RET, MET, MAML3Diagnostic molecular pathologyETV6 gene rearrangement can be demonstrated by breaking apart FISH. Detection of NTRK3 fusion by RT-PCR or RNA sequencing may be predictive of response to NTRK inhibitors for unresectable or metastatic cases S100+, SOX10+, mammaglobin+, CK18+, Cam5.2+, GCDFP-15+, NTRK+ (or pan-TRK+)CK5-, p63-, DOG-1, AR-Ki-67 < 5% Essential: monophasic tumor with vacuolated secretory material, lacking cytoplasmic granules; IHC positivity for S100 protein, mammaglobin, and NTRK (or Pan-TRK) and negativity for p40/p63, and CK5Desirable:ETV6 and/or NTRK3 gene rearrangement [85–96] 
Mucoepidermoid carcinoma (MEC)Malignant lacrimal gland neoplasm characterized by squamous (epidermoid) cells, mucous cells, columnar cells, and intermediate cells CTRC1::MAML2 gene fusion is present in majority lacrimal gland MEC and is considered a key oncogenic driver leading to EGFR upregulationRAS/PIK3 mutations in poorly differentiated tumors involved in EGFR upregulationDiagnostic molecular pathologyMAML2 rearrangement/CTRC1::MAML2 fusion in select cases P63+, p40+S100-, SOX10- Essential: squamous cells, intermediate cells, and mucous cells in varying proportionsLower grade tumors: cystic, mucous cell rich, and well circumscribedHigher grade tumors: solid, higher proportion of squamous and intermediate cellsDesirable:MAML2 rearrangement/CRTC1::MAML2 fusion [96–98] 
EMCLow-grade malignant biphasic tumor of the lacrimal gland characterized by tubular structures, usually composed of tightly coupled inner ductal and prominent outer myoepithelial cells HRAS mutations, most commonly at the codon 61Diagnostic molecular pathologyAssessment of HRAS mutations is useful for diagnosing EMC and excluding its mimics; MYB fusion by molecular methods/IHC might help in differentiation from adenoid cystic carcinoma and hybrid tumors Inner cells: panCK+, CK7+Outer myoepithelial cells: p63+, SMA+, calponin+, S100+Diffuse membranous and cytoplasmic RAS Q61R expression, almost always restricted to myoepithelial cells in 65% of EMCs Essential: usually multinodular invasive growth; at least partly with a dual arrangement of inner ductal cells and outer prominent, and usually clear, myoepithelial cells [99–103] 
Acinic cell carcinomaRare malignant lacrimal gland neoplasm displaying predominantly serous acinar differentiation Salivary gland tumors: recurrent rearrangement [t(4; 9) (q13; q31)] leads to NR4A3 upregulationDiagnostic molecular pathologyNR4A subfamily rearrangements by molecular or IHC methods in challenging cases NR4A3+ or NR4A2+DOG1+, SOX10+p63-, S100-Mammaglobin-, pan-TRK- Essential: tumor cells with acinar differentiation; zymogen granules in focal areasDesirable: nuclear staining for NR4A3/NOR-1 or NR4A2/Nuur1Molecular demonstration of NR4A3 rearrangement [104–106] 

Tumors of the Orbit (Table 14)

This new chapter, which focuses primarily on orbital teratomas, choristomas, and hamartomas, adopts, where possible, ISSVA terminology to classify vascular malformations of the eye and orbit [4]. Cavernous vascular malformation (cavernous hemangioma), combined lymphatic-venous malformation (lymphangioma), and orbital venous malformation (orbital varix) are discussed, with an update on their molecular genetics and pathogenesis [2]. Vascular neoplasms are covered in the chapter dedicated to soft tissue tumors.

Table 14.

WHO Eye5: tumors of the orbit [2]

Teratomas, hamartomas, and choristomas 
Choristomas and cysts 
Orbital dermoid cysts and other orbital cysts 
Vascular malformations 
Cavernous vascular malformation 
Combined lymphatic-venous malformation of the orbit 
Orbital venous malformation 
Arteriovenous malformation 
Hamartomas 
Orbital glial heterotopia 
Mesenchymal hamartoma of the orbit 
Germ cell tumors 
Teratoma 
Yolk sac tumor 
Melanocytic tumors of the orbit 
Primary orbital melanoma 
Teratomas, hamartomas, and choristomas 
Choristomas and cysts 
Orbital dermoid cysts and other orbital cysts 
Vascular malformations 
Cavernous vascular malformation 
Combined lymphatic-venous malformation of the orbit 
Orbital venous malformation 
Arteriovenous malformation 
Hamartomas 
Orbital glial heterotopia 
Mesenchymal hamartoma of the orbit 
Germ cell tumors 
Teratoma 
Yolk sac tumor 
Melanocytic tumors of the orbit 
Primary orbital melanoma 

Hematolymphoid Tumors (Table 15)

In this chapter, reactive lesions of the eye and orbit, and intraocular and ocular adnexal hematolymphoid proliferations are presented separately. The chapter provides an update on the molecular genetics, diagnostic cytopathology, diagnostic molecular pathology, and essential and desirable diagnostic criteria of intraocular lymphomas (shown in Fig. 7, Fig. 8 and Fig. 9; Table 16) [107‒129]. Ocular adnexal lymphomas, myeloid neoplasms, and histiocytic and dendritic cell neoplasms are comprehensively discussed, emphasizing site-specific characteristics and the most up-to-date immunophenotypic, molecular genetic, and prognostic features [2, 130, 131]. The material is coordinated with the 5th edition of WHO Classification of the Hematolymphoid Tumours [132].

Table 15.

WHO Eye5: hematolymphoid tumors [2]

Reactive lymphoid lesions 
Reactive lymphoid lesions 
Reactive lymphoid hyperplasia 
IgG4-related disease 
Intraocular hematolymphoid proliferations 
Primary choroidal lymphoma (PCL) 
Primary vitreoretinal large cell B-cell lymphoma (PVR-LCBL) 
Ocular adnexal lymphomas 
B-cell lymphomas 
Extranodal marginal zone lymphoma 
Follicular lymphoma 
Mantle cell lymphoma 
Chronic lymphocytic leukemia/small lymphocytic lymphoma 
Diffuse large B-cell lymphoma 
Plasmacytoma 
T/NK cell lymphomas 
CD30-positive lymphoproliferative disorders 
T-cell lymphomas 
Extranodal lymphoma of mature NK or T-cell lineage 
Myeloid tumors 
Extramedullary myeloid sarcoma 
Histiocytic and dendritic cell tumors 
Juvenile xanthogranuloma 
Rosai-Dorfman disease 
Erdheim-Chester disease 
Langerhans cell tumors 
Xanthelasma 
Adult orbital xanthogranulomatous disease 
Reactive lymphoid lesions 
Reactive lymphoid lesions 
Reactive lymphoid hyperplasia 
IgG4-related disease 
Intraocular hematolymphoid proliferations 
Primary choroidal lymphoma (PCL) 
Primary vitreoretinal large cell B-cell lymphoma (PVR-LCBL) 
Ocular adnexal lymphomas 
B-cell lymphomas 
Extranodal marginal zone lymphoma 
Follicular lymphoma 
Mantle cell lymphoma 
Chronic lymphocytic leukemia/small lymphocytic lymphoma 
Diffuse large B-cell lymphoma 
Plasmacytoma 
T/NK cell lymphomas 
CD30-positive lymphoproliferative disorders 
T-cell lymphomas 
Extranodal lymphoma of mature NK or T-cell lineage 
Myeloid tumors 
Extramedullary myeloid sarcoma 
Histiocytic and dendritic cell tumors 
Juvenile xanthogranuloma 
Rosai-Dorfman disease 
Erdheim-Chester disease 
Langerhans cell tumors 
Xanthelasma 
Adult orbital xanthogranulomatous disease 
Fig. 7.

Primary choroidal lymphoma (PCL), clinical and imaging findings. a Creamy choroidal infiltrates. b Optical coherence tomography demonstrates thickened choroid with an undulating hyperreflective anterior border (arrow) and obscuration of underlying choroidal vasculature from an infiltrative process.

Fig. 7.

Primary choroidal lymphoma (PCL), clinical and imaging findings. a Creamy choroidal infiltrates. b Optical coherence tomography demonstrates thickened choroid with an undulating hyperreflective anterior border (arrow) and obscuration of underlying choroidal vasculature from an infiltrative process.

Close modal
Fig. 8.

Primary choroidal lymphoma (PCL), cytopathologic findings, fine needle aspiration biopsy. a Monomorphic small blue cells in a background of hemorrhage. Neoplastic cells express CD20 (b), bcl2 (c), and interferon regulatory factor 4 (IRF4/MUM1) (d). Neoplastic cells are negative for CD5, which highlights T cells (e) and CD10 (f). Overexpression of kappa light chain (g) over lambda (h). The Ki-67 labels rare nuclei (i) [hematoxylin-eosin (a), CD20 (b), bcl2 (c), MUM1 (d), CD5 (e), CD10 (f), kappa (g), lambda (h), ki-67 (i); all figures. ×400].

Fig. 8.

Primary choroidal lymphoma (PCL), cytopathologic findings, fine needle aspiration biopsy. a Monomorphic small blue cells in a background of hemorrhage. Neoplastic cells express CD20 (b), bcl2 (c), and interferon regulatory factor 4 (IRF4/MUM1) (d). Neoplastic cells are negative for CD5, which highlights T cells (e) and CD10 (f). Overexpression of kappa light chain (g) over lambda (h). The Ki-67 labels rare nuclei (i) [hematoxylin-eosin (a), CD20 (b), bcl2 (c), MUM1 (d), CD5 (e), CD10 (f), kappa (g), lambda (h), ki-67 (i); all figures. ×400].

Close modal
Fig. 9.

Primary vitreoretinal large B-cell lymphoma (PVR-LCBL), clinical and cytopathologic findings (subretinal fine needle aspiration biopsy and vitrectomy). a Punctate and nodular subretinal pigment epithelial (RPE) deposits (arrow), geographic placoid gray subretinal deposits focally associated with hemorrhage (black arrowhead), and areas of RPE atrophy. b Optical coherence tomography highlights subretinal (arrowhead) and sub-RPE (arrow) deposits. c Large cells with irregular nuclear contours, prominent nucleoli, and scant to moderate amounts of cytoplasm. d Foci of granulomatous inflammation (paraneoplastic granulomatous vitritis). e The large atypical cells and the apoptotic debris are positive for CD20 (e) and co-express bcl6 (f) and interferon regulatory factor 4 (IRF4/MUM1) (g). CD10 is negative (h). Ki-67 proliferative index is brisk (i). Targeted MYD88 mutation studies detected MYD88 p.L265P mutation [hematoxylin-eosin (c, d), CD20 (e), bcl6 (f), MUM1 (g), CD10 (h), Ki-67 (i), ×630 (c), ×400 (d–i)].

Fig. 9.

Primary vitreoretinal large B-cell lymphoma (PVR-LCBL), clinical and cytopathologic findings (subretinal fine needle aspiration biopsy and vitrectomy). a Punctate and nodular subretinal pigment epithelial (RPE) deposits (arrow), geographic placoid gray subretinal deposits focally associated with hemorrhage (black arrowhead), and areas of RPE atrophy. b Optical coherence tomography highlights subretinal (arrowhead) and sub-RPE (arrow) deposits. c Large cells with irregular nuclear contours, prominent nucleoli, and scant to moderate amounts of cytoplasm. d Foci of granulomatous inflammation (paraneoplastic granulomatous vitritis). e The large atypical cells and the apoptotic debris are positive for CD20 (e) and co-express bcl6 (f) and interferon regulatory factor 4 (IRF4/MUM1) (g). CD10 is negative (h). Ki-67 proliferative index is brisk (i). Targeted MYD88 mutation studies detected MYD88 p.L265P mutation [hematoxylin-eosin (c, d), CD20 (e), bcl6 (f), MUM1 (g), CD10 (h), Ki-67 (i), ×630 (c), ×400 (d–i)].

Close modal
Table 16.

WHO Eye5: intraocular hematolymphoid proliferations [2]

Primary choroidal lymphoma (PCL)Primary vitreoretinal large cell B-cell lymphoma (PVR-LCBL)Ref
Definition PCL is an extranodal marginal zone B-cell lymphoma (EMZL) of the uveal tract without evidence of systemic lymphoma PVR-LCBL is the most common high-grade lymphoma that arises in the eye and is frequently accompanied by involvement of the central nervous system (CNS) [2] 
Pathogenesis Morphological, immunohistological, and genetic alterations are similar to those of EMZL at other locationsClonal expansion of post-germinal center (GC) B cellst(11; 18) (q21; q21)MYD88 mutationsDysregulation of the NF-?B, BCR, and PI3K signaling pathways PVR-LCBL cells demonstrate similarity to LCBL of immunoprivileged sites (IP-LCBL)Genetic alterations that enable immune escape and downregulation of specific immune reactionsFeatures corresponding to mature GC-exit B cells, which have undergone a prolonged GC reaction with evidence of ongoing somatic hypermutation in their rearranged immunoglobulin (IG) genesRearrangement in the IGHV4-34 gene (55%)Concordant MYD88 (L265P) and CD79B mutations (likely early events)The genomic signature resembles the C5/MCD/MYD88 signature found in diffuse large B-cell lymphoma NOSOther mutations: PIM1 (71%), IGLL5 (52%), TBL1XR1 (48%), and ETV6 (45%)Translocations in IG and BCL6 genesFrequent genetic imbalances, particularly 9p21/CDKN2A deletions (75%) [107–122, 126, 127, 129] 
Histopathology (enucleation, tissue biopsy) Diffuse infiltration of choroid by small-to-medium lymphocytes with mildly irregular nuclear contours, small-to-inconspicuous nucleoli, typically with expansion of small centrocyte-like lymphocytes, and varying numbers of plasmacytoid and monocytoid B cellsLymphoid follicles with GCs variably colonized by neoplastic cellsThe infiltrate may extend to ciliary body, iris and/or sub-conjunctival tissue, or even through Bruch’s membrane Large lymphocytes with irregular nuclear contours and prominent nucleoli, with brisk apoptosis/necrosis in the vitreous, neural retina, and/or beneath the RPEThe highly characteristic sub-RPE infiltrates (of variable thickness) consist of neoplastic B cells accumulating anterior to the Bruch membraneThere is perivascular accumulation of atypical lymphocytes, often associated with occlusion of retinal vessels [107, 108, 123–125, 127] 
Diagnostic cytopathology Monomorphous population of small atypical B cells Large atypical B cells in a background of apoptosis, necrosis, small T cells, and macrophages [125, 127] 
Immunohistochemistry (IHC) and flow cytometry Neoplastic cells are CD20+, CD79a+, PAX5+, BCL2+, IgM+, CD43+/-, MUM1+, CD5-, CD23-, CCND1-, CD10-, frequent light chain restriction, with a low Ki67 index Neoplastic cells are CD20+, CD79a+, PAX5+, BCL2+, BCL6+, MUM1/IRF4+, IgM+, CD10- and have a high Ki67 index [107][121] 
Diagnostic molecular pathology Clonality analysis (IGH and/or IGK PCR) demonstrates clonal B-cell population Clonality analysis of IGH genes demonstrates clonal B-cell populationIn rare cases where a PVR T-cell lymphoma is suspected, clonality analysis of the T-cell receptor (TCR) geneMutational analysis: MYD88 and CD79B hotspot mutations (particularly useful in low cellularity cases) [108, 111, 116–119, 123, 125, 128] 
Essential diagnostic criteria Diffuse infiltrate of small- to medium-sized lymphoid cells in the uveal tissueExpression of B lineage markersExclusion of systemic lymphoma with secondary ocular involvement Large B-cell lymphoma primarily confined to the vitreous or retina at presentation, with or without concurrent CNS lymphomaExclusion of secondary involvement by a systemic DBCL (after clinicopathological correlation) [2] 
Desirable diagnostic criteria Demonstration of light chain restriction or clonal IG gene rearrangement Post-GC B-cell phenotype (CD20+; CD79a+; MUM1+; BCL6+; CD10-)Demonstration of a high Ki67 growth fractionAbsence of Epstein-Barr virusDemonstration of clonal B-cell population or a MYD88 and/or CD79B hotspot mutations in cases in which histology is not definitive (e.g., corticosteroid-mitigated PVR-LCBL) and paucicellular cytology) [2] 
Primary choroidal lymphoma (PCL)Primary vitreoretinal large cell B-cell lymphoma (PVR-LCBL)Ref
Definition PCL is an extranodal marginal zone B-cell lymphoma (EMZL) of the uveal tract without evidence of systemic lymphoma PVR-LCBL is the most common high-grade lymphoma that arises in the eye and is frequently accompanied by involvement of the central nervous system (CNS) [2] 
Pathogenesis Morphological, immunohistological, and genetic alterations are similar to those of EMZL at other locationsClonal expansion of post-germinal center (GC) B cellst(11; 18) (q21; q21)MYD88 mutationsDysregulation of the NF-?B, BCR, and PI3K signaling pathways PVR-LCBL cells demonstrate similarity to LCBL of immunoprivileged sites (IP-LCBL)Genetic alterations that enable immune escape and downregulation of specific immune reactionsFeatures corresponding to mature GC-exit B cells, which have undergone a prolonged GC reaction with evidence of ongoing somatic hypermutation in their rearranged immunoglobulin (IG) genesRearrangement in the IGHV4-34 gene (55%)Concordant MYD88 (L265P) and CD79B mutations (likely early events)The genomic signature resembles the C5/MCD/MYD88 signature found in diffuse large B-cell lymphoma NOSOther mutations: PIM1 (71%), IGLL5 (52%), TBL1XR1 (48%), and ETV6 (45%)Translocations in IG and BCL6 genesFrequent genetic imbalances, particularly 9p21/CDKN2A deletions (75%) [107–122, 126, 127, 129] 
Histopathology (enucleation, tissue biopsy) Diffuse infiltration of choroid by small-to-medium lymphocytes with mildly irregular nuclear contours, small-to-inconspicuous nucleoli, typically with expansion of small centrocyte-like lymphocytes, and varying numbers of plasmacytoid and monocytoid B cellsLymphoid follicles with GCs variably colonized by neoplastic cellsThe infiltrate may extend to ciliary body, iris and/or sub-conjunctival tissue, or even through Bruch’s membrane Large lymphocytes with irregular nuclear contours and prominent nucleoli, with brisk apoptosis/necrosis in the vitreous, neural retina, and/or beneath the RPEThe highly characteristic sub-RPE infiltrates (of variable thickness) consist of neoplastic B cells accumulating anterior to the Bruch membraneThere is perivascular accumulation of atypical lymphocytes, often associated with occlusion of retinal vessels [107, 108, 123–125, 127] 
Diagnostic cytopathology Monomorphous population of small atypical B cells Large atypical B cells in a background of apoptosis, necrosis, small T cells, and macrophages [125, 127] 
Immunohistochemistry (IHC) and flow cytometry Neoplastic cells are CD20+, CD79a+, PAX5+, BCL2+, IgM+, CD43+/-, MUM1+, CD5-, CD23-, CCND1-, CD10-, frequent light chain restriction, with a low Ki67 index Neoplastic cells are CD20+, CD79a+, PAX5+, BCL2+, BCL6+, MUM1/IRF4+, IgM+, CD10- and have a high Ki67 index [107][121] 
Diagnostic molecular pathology Clonality analysis (IGH and/or IGK PCR) demonstrates clonal B-cell population Clonality analysis of IGH genes demonstrates clonal B-cell populationIn rare cases where a PVR T-cell lymphoma is suspected, clonality analysis of the T-cell receptor (TCR) geneMutational analysis: MYD88 and CD79B hotspot mutations (particularly useful in low cellularity cases) [108, 111, 116–119, 123, 125, 128] 
Essential diagnostic criteria Diffuse infiltrate of small- to medium-sized lymphoid cells in the uveal tissueExpression of B lineage markersExclusion of systemic lymphoma with secondary ocular involvement Large B-cell lymphoma primarily confined to the vitreous or retina at presentation, with or without concurrent CNS lymphomaExclusion of secondary involvement by a systemic DBCL (after clinicopathological correlation) [2] 
Desirable diagnostic criteria Demonstration of light chain restriction or clonal IG gene rearrangement Post-GC B-cell phenotype (CD20+; CD79a+; MUM1+; BCL6+; CD10-)Demonstration of a high Ki67 growth fractionAbsence of Epstein-Barr virusDemonstration of clonal B-cell population or a MYD88 and/or CD79B hotspot mutations in cases in which histology is not definitive (e.g., corticosteroid-mitigated PVR-LCBL) and paucicellular cytology) [2] 

Soft Tissue and Bone Tumors (Table 17)

This chapter provides salient clinical, radiologic, etiological, pathogenic, diagnostic, and prognostic information on each ocular and ocular adnexal soft tissue tumor, with an emphasis on site-specific characteristics. Vascular lesions classified as benign neoplasms of blood vessels, in accordance with the ISSVA nomenclature [4], such as infantile hemangioma and lobular capillary hemangioma, are covered. Vascular malformations are discussed in the chapter dedicated to the tumors of the orbit. The material is coordinated with the 5th edition of WHO Classification of the Soft Tissue and Bone Tumours [133].

Table 17.

WHO Eye5: soft tissue and bone tumors [2]

Soft tissue tumors 
Fibrous, fibroblastic tumors 
Conjunctival stromal tumor and ocular surface fibroma 
Fibrous histiocytoma 
Nodular and proliferative fasciitis, proliferative myositis 
Solitary fibrous tumor 
Inflammatory myofibroblastic tumor 
Dermatofibrosarcoma protuberans 
Angiomyxoma 
Adipocytic tumors 
Benign lipomatous lesions 
Malignant adipocytic tumors 
Skeletal muscle tumors 
Rhabdomyosarcoma family 
Smooth muscle tumors 
Leiomyoma 
Mesectodermal leiomyoma 
Leiomyosarcoma 
Peripheral nerve sheath tumors 
Schwannoma 
Neurofibroma 
Solitary circumscribed neuroma 
Granular cell tumor 
Malignant peripheral nerve sheath tumor 
Vascular tumors 
Masson tumor 
Infantile hemangioma 
Lobular capillary hemangioma 
Lymphangiectasia 
Epithelioid hemangioma 
Hemangioblastoma 
Angiosarcoma 
Perivascular tumors 
Glomus tumor 
Myopericytoma, including myofibroma 
Tumors of uncertain derivation 
PEComa 
Epithelioid sarcoma 
Synovial sarcoma 
Alveolar soft part sarcoma 
Tumors of orbital bone 
Osteogenic tumors of orbital bone 
Osteoma 
Osteoblastoma 
Osteosarcoma 
Chondrogenic tumors of orbital bone 
Soft tissue chondroma 
Mesenchymal chondrosarcoma 
Fibrous, fibroblastic tumors of orbital bone 
Fibrous dysplasia 
Vascular tumors of orbital bone 
Hemangioma of bone 
Epithelioid hemangioendothelioma 
Angiosarcoma of bone 
Osteoclast-rich giant cell tumors of orbital bone 
Aneurysmal bone cyst 
Giant cell tumor of bone 
Other tumors of orbital bone 
Ewing sarcoma 
Soft tissue tumors 
Fibrous, fibroblastic tumors 
Conjunctival stromal tumor and ocular surface fibroma 
Fibrous histiocytoma 
Nodular and proliferative fasciitis, proliferative myositis 
Solitary fibrous tumor 
Inflammatory myofibroblastic tumor 
Dermatofibrosarcoma protuberans 
Angiomyxoma 
Adipocytic tumors 
Benign lipomatous lesions 
Malignant adipocytic tumors 
Skeletal muscle tumors 
Rhabdomyosarcoma family 
Smooth muscle tumors 
Leiomyoma 
Mesectodermal leiomyoma 
Leiomyosarcoma 
Peripheral nerve sheath tumors 
Schwannoma 
Neurofibroma 
Solitary circumscribed neuroma 
Granular cell tumor 
Malignant peripheral nerve sheath tumor 
Vascular tumors 
Masson tumor 
Infantile hemangioma 
Lobular capillary hemangioma 
Lymphangiectasia 
Epithelioid hemangioma 
Hemangioblastoma 
Angiosarcoma 
Perivascular tumors 
Glomus tumor 
Myopericytoma, including myofibroma 
Tumors of uncertain derivation 
PEComa 
Epithelioid sarcoma 
Synovial sarcoma 
Alveolar soft part sarcoma 
Tumors of orbital bone 
Osteogenic tumors of orbital bone 
Osteoma 
Osteoblastoma 
Osteosarcoma 
Chondrogenic tumors of orbital bone 
Soft tissue chondroma 
Mesenchymal chondrosarcoma 
Fibrous, fibroblastic tumors of orbital bone 
Fibrous dysplasia 
Vascular tumors of orbital bone 
Hemangioma of bone 
Epithelioid hemangioendothelioma 
Angiosarcoma of bone 
Osteoclast-rich giant cell tumors of orbital bone 
Aneurysmal bone cyst 
Giant cell tumor of bone 
Other tumors of orbital bone 
Ewing sarcoma 

Metastases to the Eye and Surrounding Structures

This chapter combines information from the prior edition on secondary and metastatic tumors to various ocular tissues into a separate section, Metastases to Intra-Ocular Structures[2]. Analogously, metastases to ocular adnexa are covered in a new section, Metastases to Ocular Adnexal Structures[2].

Genetic Tumor Predisposition Syndromes

This chapter synthesizes the most up-to-date clinical, epidemiologic, genetic, diagnostic, and prognostic information on the genetic tumor predisposition syndromes of particular relevance to the ophthalmologist: the retinoblastoma syndrome, BAP1 tumor predisposition syndrome, von Hippel-Lindau syndrome, Sturge-Weber syndrome, neurofibromatosis type 1, Goldenhar syndrome, and Muir-Torre syndrome [2]. The information is coordinated with the upcoming new WHO Classification of Tumours volume, on genetic tumor syndromes, which will be last of the 14 vol making up the 5th edition.

In conclusion, the WHO Eye5 provides an up-to-date comprehensive taxonomy for the range of tumors occurring in the eye and orbit with an emphasis on their site-specific features. The information presented is standardized across the 5th edition volumes. This systematic approach not only facilitates cross-referencing the information presented with other volumes within the classification, but also highlights the remaining gaps in knowledge. As such, the classification will not only serve as a valuable reference for a practicing ophthalmic oncologist and pathologist, but also drive future research bridging various oncology and pathology disciplines. It is hoped that the information conveyed in this overview will help to distill the major changes in the latest edition and will facilitate more detailed study of the volume.

The authors acknowledge the International Agency for Research on Cancer (IARC) staff, IARC editorial board, and authors, who have directly and indirectly contributed to the WHO Eye5 edition and other 5th edition WHO volumes, on whose behalf this paper has been prepared.

Participants provided written informed consent for the publication of their details and any accompanying images.

The authors have no conflicts of interest to declare.

No funding received.

Study design and data acquisition (T.M., H.E.G., I.A.C., G.G.-L., T.T.K., S.E.C., H.S.M., C.G.E., R.M.V., S.H., A.J.G., A.A.R., B.E., M.J.J., V.A.W., and J.C.H.), manuscript drafting (T.M.), and manuscript review (T.M., H.E.G., I.A.C., G.G.-L., T.M., H.E.G., I.A.C., G.G.-L., T.T.K., S.E.C., H.S.M., C.G.E., R.M.V., S.H., A.J.G., A.A.R., B.E., M.J.J., V.A.W., and J.C.H.). The content of this article represents the personal views of the authors and does not represent the views of the authors’ employers and associated institutions. Where authors are identified as personnel of the International Agency for Research on Cancer/World Health Organization, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy, or views of the International Agency for Research on Cancer/World Health Organization.

1.
International Agency for Research on Cancer [Internet]
Lyon
WHO Classification of Tumours Online
. Available from: https://tumourclassification.iarc.who.int (accessed August 28, 2022).
2.
WHO classification of tumours editorial board. Eye tumours
[Internet; beta version ahead of print].
WHO classification of tumours series
5th ed.
Lyon (France)
International Agency for Research on Cancer
2023
vol. 13
[cited]. Available from: https://tumourclassification.iarc.who.int/chapters/??.
3.
Grossniklaus
HE
,
Eberhart
CG
,
Kivelä
TT
WHO classifiction of tumours of the eye
4th ed.
Lyon
International Agency for Research on Cancer
2018
.
4.
International Society for the Study of Vascular Anomalies [Internet]
.
2018 ISSVA classification of vascular anomalies [cited 2018, may]
. Available from: issva.org/classification.
5.
Bruford
EA
,
Braschi
B
,
Denny
P
,
Jones
TEM
,
Seal
RL
,
Tweedie
S
.
Guidelines for human gene nomenclature
.
Nat Genet
.
2020 Aug
52
8
754
8
.
6.
den Dunnen
JT
,
Dalgleish
R
,
Maglott
DR
,
Hart
RK
,
Greenblatt
MS
,
McGowan-Jordan
J
.
HGVS recommendations for the description of sequence variants: 2016 update
.
Hum Mutat
.
2016 Jun
37
6
564
9
.
7.
Cree
IA
,
Tan
PH
,
Travis
WD
,
Wesseling
P
,
Yagi
Y
,
White
VA
.
Counting mitoses: SI(ze) matters!
.
Mod Pathol
.
2021 Sep
34
9
1651
7
.
8.
Price
HN
.
Congenital melanocytic nevi: update in genetics and management
.
Curr Opin Pediatr
.
2016 Aug
28
4
476
82
.
9.
Roh
MR
,
Eliades
P
,
Gupta
S
,
Tsao
H
.
Genetics of melanocytic nevi
.
Pigment Cell Melanoma Res
.
2015 Nov
28
6
661
72
.
10.
Pollock
PM
,
Harper
UL
,
Hansen
KS
,
Yudt
LM
,
Stark
M
,
Robbins
CM
.
High frequency of BRAF mutations in nevi
.
Nat Genet
.
2003 Jan
33
1
19
20
.
11.
Francis
JH
,
Grossniklaus
HE
,
Habib
LA
,
Marr
B
,
Abramson
DH
,
Busam
KJ
.
BRAF, NRAS, and GNAQ mutations in conjunctival melanocytic nevi
.
Invest Ophthalmol Vis Sci
.
2018 Jan
59
1
117
21
.
12.
Goldenberg-Cohen
N
,
Cohen
Y
,
Rosenbaum
E
,
Herscovici
Z
,
Chowers
I
,
Weinberger
D
.
T1799A BRAF mutations in conjunctival melanocytic lesions
.
Invest Ophthalmol Vis Sci
.
2005 Sep
46
9
3027
30
.
13.
Gonzalez-H Leon
A
,
Chavez
Y
,
Kamil
ZS
,
Ghazarian
D
,
Krema
H
.
Diagnosis of the origin of an epibulbar melanocytic tumor with molecular genomics
.
Ophthalmic Genet
.
2022 Aug
43
4
518
521
.
14.
Yeh
I
,
Lang
UE
,
Durieux
E
,
Tee
MK
,
Jorapur
A
,
Shain
AH
.
Combined activation of MAP kinase pathway and β-catenin signaling cause deep penetrating nevi
.
Nat Commun
.
2017 Sep 21
8
1
644
.
15.
Šekoranja
D
,
Vergot
K
,
Hawlina
G
,
Pižem
J
.
Combined deep penetrating nevi of the conjunctiva are relatively common lesions characterised by BRAFV600E mutation and activation of the beta catenin pathway: a clinicopathological analysis of 34 lesions
.
Br J Ophthalmol
.
2020 Jul
104
7
1016
21
.
16.
van Ipenburg
JA
,
Damman
J
,
Paridaens
D
,
Verdijk
RM
.
Histopathological and molecular features of a conjunctival caruncular deep penetrating nevus
.
Ocul Oncol Pathol
.
2020 Aug
6
4
293
6
.
17.
Eiger-Moscovich
M
,
Eagle
RC
,
Milman
T
,
Acid-Schiff
P
.
β-Catenin and periodic acid-schiff distinguish granular cell nevus from deep penetrating nevus
.
Arch Pathol Lab Med
.
2021 Dec 1
145
12
1475
6
.
18.
Larsen
AC
,
Dahl
C
,
Dahmcke
CM
,
Lade-Keller
J
,
Siersma
VD
,
Toft
PB
.
BRAF mutations in conjunctival melanoma: investigation of incidence, clinicopathological features, prognosis and paired premalignant lesions
.
Acta Ophthalmol
.
2016 Aug
94
5
463
70
.
19.
Griewank
KG
,
Westekemper
H
,
Schilling
B
,
Livingstone
E
,
Schimming
T
,
Sucker
A
.
Conjunctival melanomas harbor BRAF and NRAS mutations–response
.
Clin Cancer Res
.
2013 Nov 15
19
22
6331
2
.
20.
Scholz
SL
,
Cosgarea
I
,
Süßkind
D
,
Murali
R
,
Möller
I
,
Reis
H
.
NF1 mutations in conjunctival melanoma
.
Br J Cancer
.
2018 May
118
9
1243
7
.
21.
Gardrat
S
,
Houy
A
,
Brooks
K
,
Cassoux
N
,
Barnhill
R
,
Dayot
S
.
Definition of biologically distinct groups of conjunctival melanomas according to etiological factors and implications for precision medicine
.
Cancers
.
2021 Jul 30
13
15
3836
.
22.
van Poppelen
NM
,
van Ipenburg
JA
,
van den Bosch
Q
,
Vaarwater
J
,
Brands
T
,
Eussen
B
.
Molecular genetics of conjunctival melanoma and prognostic value of TERT promoter mutation analysis
.
Int J Mol Sci
.
2021 May 28
22
11
5784
.
23.
Lally
SE
,
Milman
T
,
Orloff
M
,
Dalvin
LA
,
Eberhart
CG
,
Heaphy
CM
.
Mutational landscape and outcomes of conjunctival melanoma in 101 patients
.
Ophthalmology
.
2022 Jun
129
6
679
93
.
24.
Kenawy
N
,
Kalirai
H
,
Sacco
JJ
,
Lake
SL
,
Heegaard
S
,
Larsen
AC
.
Conjunctival melanoma copy number alterations and correlation with mutation status, tumor features, and clinical outcome
.
Pigment Cell Melanoma Res
.
2019 Jul
32
4
564
75
.
25.
Mudhar
HS
,
Smith
K
,
Talley
P
,
Whitworth
A
,
Atkey
N
,
Rennie
IG
.
Fluorescence In Situ Hybridisation (FISH) in histologically challenging conjunctival melanocytic lesions
.
Br J Ophthalmol
.
2013 Jan
97
1
40
6
.
26.
Milman
T
,
Zhang
Q
,
Ang
S
,
Elder
D
,
Ida
CM
,
Salomao
DR
.
Conjunctival nevi and melanoma: multiparametric immunohistochemical analysis, including p16, SOX10, HMB45, and Ki-67
.
Hum Pathol
.
2020 Sep
103
107
19
.
27.
María Moral
R
,
Monteagudo
C
,
Muriel
J
,
Moreno
L
,
María Peiró
A
.
Fluorescent In Situ Hybridization (FISH): a useful diagnostic tool for childhood conjunctival melanoma
.
Eur J Ophthalmol
.
2021 Jul 9
32
6
NP13
9
. Epub ahead of print.
28.
Milman
T
,
Eiger-Moscovich
M
,
Henry
RK
,
Folberg
R
,
Coupland
SE
,
Grossniklaus
HE
.
Validation of the newly proposed World Health Organization classification system for conjunctival melanocytic intraepithelial lesions: a comparison with the C-MIN and PAM classification schemes
.
Am J Ophthalmol
.
2021 Mar
223
60
74
.
29.
Damato
B
,
Coupland
SE
.
Conjunctival melanoma and melanosis: a reappraisal of terminology, classification and staging
.
Clin Exp Ophthalmol
.
2008 Nov
36
8
786
95
.
30.
Mudhar
HS
,
Milman
T
,
Zhang
PJL
,
Shields
CL
,
Eagle
RC
,
Lally
SE
.
Conjunctival “mucoepidermoid carcinoma” revisited: a revision of terminology, based on morphologic, immunohistochemical and molecular findings of 14 cases, and the 2018 WHO Classification of Tumours of the Eye
.
Mod Pathol
.
2020 Jul
33
7
1242
55
.
31.
Agni
M
,
Raven
ML
,
Bowen
RC
,
Laver
NV
,
Chevez-Barrios
P
,
Milman
T
.
An update on endocrine mucin-producing sweat gland carcinoma: clinicopathologic study of 63 cases and comparative analysis
.
Am J Surg Pathol
.
2020 Aug
44
8
1005
16
.
32.
Yonekawa
Y
,
Jakobiec
FA
,
Zakka
FR
,
Fay
A
.
Sebaceoma of the eyelid
.
Ophthalmology
.
2012 Dec
119
12
2645.e1
4
.
33.
Misago
N
,
Mihara
I
,
Ansai
S
,
Narisawa
Y
.
Sebaceoma and related neoplasms with sebaceous differentiation: a clinicopathologic study of 30 cases
.
Am J Dermatopathol
.
2002 Aug
24
4
294
304
.
34.
Tetzlaff
MT
,
Singh
RR
,
Seviour
EG
,
Curry
JL
,
Hudgens
CW
,
Bell
D
.
Next-generation sequencing identifies high frequency of mutations in potentially clinically actionable genes in sebaceous carcinoma
.
J Pathol
.
2016 Sep
240
1
84
95
.
35.
Peterson
C
,
Moore
R
,
Hicks
JL
,
Morsberger
LA
,
De Marzo
AM
,
Zou
Y
.
NGS analysis confirms common TP53 and RB1 mutations, and suggests MYC amplification in ocular adnexal sebaceous carcinomas
.
Int J Mol Sci
.
2021 Aug 6
22
16
8454
.
36.
Tetzlaff
MT
,
Curry
JL
,
Ning
J
,
Sagiv
O
,
Kandl
TL
,
Peng
B
.
Distinct biological types of ocular adnexal sebaceous carcinoma: HPV-driven and virus-negative tumors arise through nonoverlapping molecular-genetic alterations
.
Clin Cancer Res
.
2019 Feb 15
25
4
1280
90
.
37.
Saro
F
,
Clua
A
,
Esteva
E
,
Carreras
A
,
Adán
A
,
Lerma
E
.
Cytologic diagnosis of ocular melanocytoma: a case report
.
Acta Cytol
.
2008 Jan-Feb
52
1
87
90
.
38.
Grossniklaus
HE
.
Fine-needle aspiration biopsy of the iris
.
Arch Ophthalmol
.
1992 Jul
110
7
969
76
.
39.
Char
DH
,
Kemlitz
AE
,
Miller
T
,
Crawford
JB
.
Iris ring melanoma: fine needle biopsy
.
Br J Ophthalmol
.
2006 Apr
90
4
420
2
.
40.
Hussain
RN
,
Kalirai
H
,
Groenewald
C
,
Kacperek
A
,
Errington
RD
,
Coupland
SE
.
Prognostic biopsy of choroidal melanoma after proton beam radiation therapy
.
Ophthalmology
.
2016 Oct
123
10
2264
5
.
41.
Jager
MJ
,
Shields
CL
,
Cebulla
CM
,
Abdel-Rahman
MH
,
Grossniklaus
HE
,
Stern
MH
.
Uveal melanoma
.
Nat Rev Dis Primers
.
2020 Apr 9
6
1
24
.
42.
Abbott
DW
,
Simons
K
,
Giorgadze
T
.
Choroidal melanoma diagnosed by aspiration cytology: a case report with cyto-histologic correlation and review of the literature
.
Ann Diagn Pathol
.
2019 Oct
42
39
41
.
43.
Solomon
DA
,
Ramani
B
,
Eiger-Moscovich
M
,
Milman
T
,
Uludag
G
,
Crawford
JB
.
Iris and ciliary body melanocytomas are defined by solitary GNAQ mutation without additional oncogenic alterations
.
Ophthalmology
.
2022 Dec
129
12
1429
39
.
44.
Niffenegger
JH
,
Soltero
A
,
Niffenegger
JS
,
Yang
S
,
Adamus
G
.
Prevalence of Hepatocyte Growth Factor and autoantibodies to α-HGF as a new etiology for bilateral diffuse uveal melanocytic proliferation masquerading as neovascular age-related macular degeneration
.
J Clin Exp Ophthalmol
.
2018
;
9
(
4
):
740
.
45.
Mudhar
HS
,
Bata
BM
,
Quhill
H
,
Milman
T
,
Salvi
SM
,
Perivascular
P
.
Uveal melanoma and paraneoplastic perivascular dermal melanocytic proliferation in the setting of bilateral diffuse uveal melanocytic proliferation: the potential role of the hepatocyte growth factor/c-met Axis in their pathogenesis
.
Ocul Oncol Pathol
.
2021 Dec
7
6
418
27
.
46.
Dimaras
H
,
Khetan
V
,
Halliday
W
,
Orlic
M
,
Prigoda
NL
,
Piovesan
B
.
Loss of RB1 induces non-proliferative retinoma: increasing genomic instability correlates with progression to retinoblastoma
.
Hum Mol Genet
.
2008 May 15
17
10
1363
72
.
47.
Eagle
RC
Jr
.
High-risk features and tumor differentiation in retinoblastoma: a retrospective histopathologic study
.
Arch Pathol Lab Med
.
2009 Aug
133
8
1203
9
.
48.
Skalet
AH
,
Gombos
DS
,
Gallie
BL
,
Kim
JW
,
Shields
CL
,
Marr
BP
.
Screening children at risk for retinoblastoma: consensus report from the American association of ophthalmic oncologists and pathologists
.
Ophthalmology
.
2018 Mar
125
3
453
8
.
49.
Rushlow
DE
,
Mol
BM
,
Kennett
JY
,
Yee
S
,
Pajovic
S
,
Thériault
BL
.
Characterisation of retinoblastomas without RB1 mutations: genomic, gene expression, and clinical studies
.
Lancet Oncol
.
2013 Apr
14
4
327
34
.
50.
Kim
ME
,
Polski
A
,
Xu
L
,
Prabakar
RK
,
Peng
CC
,
Reid
MW
.
Comprehensive somatic copy number analysis using aqueous humor liquid biopsy for retinoblastoma
.
Cancers
.
2021 Jul 3
13
13
3340
.
51.
Gerrish
A
,
Jenkinson
H
,
Cole
T
.
The impact of cell-free DNA analysis on the management of retinoblastoma
.
Cancers
.
2021 Mar 29
13
7
1570
.
52.
Abramson
DH
.
Cell free DNA (cfDNA) in the blood of retinoblastoma patients the robert M. Ellsworth lecture
.
Ophthalmic Genet
.
2022 Apr 5
43
6
731
735
. Epub ahead of print.
53.
Singh
G
,
Gupta
R
,
Kakkar
A
,
Iyer
VK
,
Kashyap
S
,
Bakhshi
S
.
Fine needle aspiration cytology of metastatic ocular medulloepithelioma
.
Cytopathology
.
2011 Oct
22
5
343
5
.
54.
Mahdjoubi
A
,
Cassoux
N
,
Levy-Gabriel
C
,
Desjardins
L
,
Klos
J
,
Caly
M
.
Adult ocular medulloepithelioma diagnosed by transscleral fine needle aspiration: a case report
.
Diagn Cytopathol
.
2017 Jun
45
6
561
4
.
55.
Saunders
T
,
Margo
CE
.
Intraocular medulloepithelioma
.
Arch Pathol Lab Med
.
2012
;
136
(
2
):
212
6
.
56.
Babu
N
,
Dey
P
.
Medulloepithelioma of ciliary body diagnosed by fine needle aspiration cytology
.
Cytopathology
.
2003 Apr
14
2
93
4
.
57.
Mudhar
HS
,
Milman
T
,
Eagle
RC
Jr
,
Sanderson
T
,
Pheasey
L
,
Paine
S
.
Usefulness of PAX8 immunohistochemistry in adult intraocular tumor diagnosis
.
Ophthalmology
.
2021 May
128
5
765
78
.
58.
Char
DH
,
Miller
TR
,
Crawford
JB
.
Cytopathologic diagnosis of benign lesions simulating choroidal melanomas
.
Trans Am Ophthalmol Soc
.
1991
;
112
(
1
):
235
44
; discussion 70–75.
59.
Mori
T
,
Sukeda
A
,
Sekine
S
,
Shibata
S
,
Ryo
E
,
Okano
H
.
SOX10 expression as well as BRAF and GNAQ/11 mutations distinguish pigmented ciliary epithelium neoplasms from uveal melanomas
.
Invest Ophthalmol Vis Sci
.
2017
;
58
(
12
):
5445
51
.
60.
Shields
JA
,
Eagle
RC
Jr
,
Dutton
J
,
Ehya
H
,
Shields
CL
.
Adenocarcinoma of the retinal pigment epithelium: clinicopathologic correlation with paradoxical immunohistochemical findings
.
JAMA Ophthalmol
.
2014 Oct
132
10
1249
52
.
61.
Benson
JC
,
Giannini
C
,
Cohen Cohen
S
,
Van Gompel
J
,
Kim
DK
,
Port
J
.
Optic nerve choristoma mimicking a neurenteric cyst
.
AJNR Am J Neuroradiol
.
2021 Jan
42
2
228
32
.
62.
Harrison
W
,
Pittman
P
,
Cummings
T
.
Optic nerve choristoma
.
Saudi J Ophthalmol
.
2018 Jan-Mar
32
1
90
1
.
63.
WHO Classification of Tumours Editorial Board
World Health organization classification of tumours of the central nervous system
5th ed.
Lyon
International Agency for Research on Cancer
2021
.
64.
Andreasen
S
,
von Holstein
SL
,
Homøe
P
,
Heegaard
S
.
Recurrent rearrangements of the PLAG1 and HMGA2 genes in lacrimal gland pleomorphic adenoma and carcinoma ex pleomorphic adenoma
.
Acta Ophthalmol
.
2018 Nov
96
7
e768
71
.
65.
Harrison
W
,
Pittman
P
,
Cummings
T
.
Pleomorphic adenoma of the lacrimal gland: a review with updates on malignant transformation and molecular genetics
.
Saudi J Ophthalmol
.
2018 Jan-Mar
32
1
13
6
.
66.
Mendoza
PR
,
Jakobiec
FA
,
Krane
JF
.
Immunohistochemical features of lacrimal gland epithelial tumors
.
Am J Ophthalmol
.
2013 Dec
156
6
1147
58
. e1.
67.
Jakobiec
FA
,
Stagner
AM
,
Eagle
RC
Jr
,
Lally
SE
,
Krane
JF
.
Unusual pleomorphic adenoma of the lacrimal Gland: immunohistochemical demonstration of PLAG1 and HMGA2 oncoproteins
.
Surv Ophthalmol
.
2017 Mar-Apr
62
2
219
26
.
68.
Shah
SS
,
Chandan
VS
,
Wilbur
DC
,
Khurana
KK
.
Glial fibrillary acidic protein and CD57 immunolocalization in cell block preparations is a useful adjunct in the diagnosis of pleomorphic adenoma
.
Arch Pathol Lab Med
.
2007 Sep
131
9
1373
7
.
69.
Curran
AE
,
Allen
CM
,
Beck
FM
,
Damm
DD
,
Murrah
VA
.
Distinctive pattern of glial fibrillary acidic protein immunoreactivity useful in distinguishing fragmented pleomorphic adenoma, canalicular adenoma and polymorphous low grade adenocarcinoma of minor salivary glands
.
Head Neck Pathol
.
2007 Sep
1
1
27
32
.
70.
Tom
A
,
Bell
D
,
Ford
JR
,
Debnam
JM
,
Guo
Y
,
Frank
SJ
.
Malignant mixed tumor (carcinoma ex pleomorphic adenoma) of the lacrimal gland
.
Ophthalmic Plast Reconstr Surg
.
2020 Sep
36
5
497
502
.
71.
Neerukonda
VK
,
Carruth
B
,
Estopinal
MDV
.
Invasive carcinoma ex-pleomorphic adenoma of the lacrimal gland with a cystadenocarcinoma component: a case report and review of the literature
.
Case Rep Pathol
.
2020
;
2020
:
6482837
.
72.
Andreasen
S
,
Heegaard
S
,
Grauslund
M
,
Homøe
P
.
The interleukin-6/Janus kinase/STAT3 pathway in pleomorphic adenoma and carcinoma ex pleomorphic adenoma of the lacrimal gland
.
Acta Ophthalmol
.
2016 Dec
94
8
798
804
.
73.
Katabi
N
,
Xu
B
,
Jungbluth
AA
,
Zhang
L
,
Shao
SY
,
Lane
J
.
PLAG1 immunohistochemistry is a sensitive marker for pleomorphic adenoma: a comparative study with PLAG1 genetic abnormalities
.
Histopathology
.
2018 Jan
72
2
285
93
.
74.
Zhang
P
,
Tang
LJ
,
Gao
HH
,
Zhang
WX
,
Lin
JX
,
Yang
HS
.
Immunohistochemical features of carcinoma ex pleomorphic adenoma and pleomorphic adenoma in the lacrimal gland
.
Int J Ophthalmol
.
2019 Aug 18
12
8
1238
42
.
75.
von Holstein
SL
,
Fehr
A
,
Persson
M
,
Therkildsen
MH
,
Prause
JU
,
Heegaard
S
.
Adenoid cystic carcinoma of the lacrimal gland: MYB gene activation, genomic imbalances, and clinical characteristics
.
Ophthalmology
.
2013 Oct
120
10
2130
8
.
76.
Chen
TY
,
Keeney
MG
,
Chintakuntlawar
AV
,
Knutson
DL
,
Kloft-Nelson
S
,
Greipp
PT
.
Adenoid cystic carcinoma of the lacrimal gland is frequently characterized by MYB rearrangement
.
Eye
.
2017 May
31
5
720
5
.
77.
Sant
DW
,
Tao
W
,
Field
MG
,
Pelaez
D
,
Jin
K
,
Capobianco
A
.
Whole exome sequencing of lacrimal gland adenoid cystic carcinoma
.
Invest Ophthalmol Vis Sci
.
2017 May 1
58
6
BIO240
6
.
78.
Bell
D
,
Sniegowski
MC
,
Wani
K
,
Prieto
V
,
Esmaeli
B
.
Mutational landscape of lacrimal gland carcinomas and implications for treatment
.
Head Neck
.
2016 Apr
38
Suppl 1
E724
9
.
79.
Duarte
AF
,
Alpuim Costa
D
,
Caçador
N
,
Boavida
AM
,
Afonso
AM
,
Vilares
M
.
Adenoid cystic carcinoma of the palpebral lobe of the lacrimal gland - case report and literature review
.
Orbit
.
2022 Oct
41
5
605
10
.
80.
Andreasen
S
,
Grauslund
M
,
Heegaard
S
.
Lacrimal gland ductal carcinomas: clinical, Morphological and Genetic characterization and implications for targeted treatment
.
Acta Ophthalmol
.
2017 May
95
3
299
306
.
81.
Kubota
T
,
Moritani
S
,
Ichihara
S
.
Clinicopathologic and immunohistochemical features of primary ductal adenocarcinoma of lacrimal gland: five new cases and review of literature
.
Graefes Arch Clin Exp Ophthalmol
.
2013 Aug
251
8
2071
6
.
82.
Zhu
MM
,
Cui
HG
,
Teng
XD
.
GCDFP-15, AR, and Her-2 as biomarkers for primary ductal adenocarcinoma of the lacrimal gland: a Chinese case and literature review
.
Onco Targets Ther
.
2015 May 11
8
1017
24
.
83.
See
TRO
,
Stålhammar
G
,
Tang
T
,
Manusow
JS
,
Jordan
DR
,
Nerad
JA
.
Primary ductal adenocarcinoma of the lacrimal gland: a review and report of five cases
.
Surv Ophthalmol
.
2020 May-Jun
65
3
371
80
.
84.
Tripathy
D
,
Agarwal
S
,
Biala
A
,
Rath
S
,
Mittal
R
.
Primary De novo ductal adenocarcinoma of the lacrimal gland
.
Ann Diagn Pathol
.
2021 Feb
50
151651
.
85.
Hyrcza
MD
,
Andreasen
S
,
Melchior
LC
,
Tucker
T
,
Heegaard
S
,
White
VA
.
Primary secretory carcinoma of the lacrimal gland: report of a new entity
.
Am J Ophthalmol
.
2018 Sep
193
178
83
.
86.
Bortz
JG
,
Zhang
PJL
,
Eagle
RC
Jr
,
Yong
JJ
,
Milman
T
.
Secretory carcinoma of the lacrimal gland: a rare case report
.
Ophthalmic Plast Reconstr Surg
.
2018 Sep
34
5
e154
7
.
87.
Bao
Y
,
Li
J
,
Zhu
Y
.
Mammary analog secretory carcinoma with ETV6 rearrangement arising in the conjunctiva and eyelid
.
Am J Dermatopathol
.
2018 Jul
40
7
531
5
.
88.
Armijos
PO
,
Uhlenhake
E
,
Milman
T
.
Secretory carcinoma of the eyelid arising in an adnexal gland
.
Ophthalmology
.
2022 Jun 23
129
10
1218
6420
.
89.
Ito
Y
,
Ishibashi
K
,
Masaki
A
,
Fujii
K
,
Fujiyoshi
Y
,
Hattori
H
.
Mammary analogue secretory carcinoma of salivary glands: a clinicopathologic and molecular study including 2 cases harboring ETV6-X fusion
.
Am J Surg Pathol
.
2015 May
39
5
602
10
.
90.
Skálová
A
,
Vanecek
T
,
Simpson
RH
,
Laco
J
,
Majewska
H
,
Baneckova
M
.
Mammary analogue secretory carcinoma of salivary glands: molecular analysis of 25 ETV6 gene rearranged tumors with lack of detection of classical ETV6-NTRK3 fusion transcript by standard RT-PCR: report of 4 cases harboring ETV6-X gene fusion
.
Am J Surg Pathol
.
2016 Jan
40
1
3
13
.
91.
Skalova
A
,
Vanecek
T
,
Martinek
P
,
Weinreb
I
,
Stevens
TM
,
Simpson
RHW
.
Molecular profiling of mammary analog secretory carcinoma revealed a subset of tumors harboring a novel ETV6-RET translocation: report of 10 cases
.
Am J Surg Pathol
.
2018 Feb
42
2
234
46
.
92.
Rooper
LM
,
Karantanos
T
,
Ning
Y
,
Bishop
JA
,
Gordon
SW
,
Kang
H
.
Salivary secretory carcinoma with a novel ETV6-MET fusion: expanding the molecular spectrum of a recently described entity
.
Am J Surg Pathol
.
2018 Aug
42
8
1121
6
.
93.
Guilmette
J
,
Dias-Santagata
D
,
Nosé
V
,
Lennerz
JK
,
Sadow
PM
.
Novel gene fusions in secretory carcinoma of the salivary glands: enlarging the ETV6 family
.
Hum Pathol
.
2019 Jan
83
50
8
.
94.
Drilon
A
,
Li
G
,
Dogan
S
,
Gounder
M
,
Shen
R
,
Arcila
M
.
What hides behind the MASC: clinical response and acquired resistance to entrectinib after ETV6-NTRK3 identification in a mammary analogue secretory carcinoma (MASC)
.
Ann Oncol
.
2016 May
27
5
920
6
.
95.
Drilon
A
.
TRK inhibitors in TRK fusion-positive cancers
.
Ann Oncol
.
2019 Nov
30
Suppl 8
viii23
30
.
96.
Skálová
A
,
Stenman
G
,
Simpson
RHW
,
Hellquist
H
,
Slouka
D
,
Svoboda
T
.
The role of molecular testing in the differential diagnosis of salivary gland carcinomas
.
Am J Surg Pathol
.
2018 Feb
42
2
e11
27
.
97.
Chen
Z
,
Ni
W
,
Li
JL
,
Lin
S
,
Zhou
X
,
Sun
Y
.
The CRTC1-MAML2 fusion is the major oncogenic driver in mucoepidermoid carcinoma
.
JCI Insight
.
2021 Apr 8
6
7
e139497
.
98.
Morita
M
,
Murase
T
,
Okumura
Y
,
Ueda
K
,
Sakamoto
Y
,
Masaki
A
.
Clinicopathological significance of EGFR pathway gene mutations and CRTC1/3-MAML2 fusions in salivary gland mucoepidermoid carcinoma
.
Histopathology
.
2020 Jun
76
7
1013
22
.
99.
Urano
M
,
Nakaguro
M
,
Yamamoto
Y
,
Hirai
H
,
Tanigawa
M
,
Saigusa
N
.
Diagnostic significance of HRAS mutations in epithelial-myoepithelial carcinomas exhibiting a broad histopathologic spectrum
.
Am J Surg Pathol
.
2019 Jul
43
7
984
94
.
100.
Nakaguro
M
,
Nagao
T
.
Epithelial-Myoepithelial carcinoma
.
Surg Pathol Clin
.
2021 Mar
14
1
97
109
.
101.
Bishop
JA
,
Westra
WH
.
MYB translocation status in salivary gland epithelial-myoepithelial carcinoma: evaluation of classic, variant, and hybrid forms
.
Am J Surg Pathol
.
2018 Mar
42
3
319
25
.
102.
Seethala
RR
,
Barnes
EL
,
Hunt
JL
.
Epithelial-myoepithelial carcinoma: a review of the clinicopathologic spectrum and immunophenotypic characteristics in 61 tumors of the salivary glands and upper aerodigestive tract
.
Am J Surg Pathol
.
2007 Jan
31
1
44
57
.
103.
Nakaguro
M
,
Tanigawa
M
,
Hirai
H
,
Yamamoto
Y
,
Urano
M
,
Takahashi
RH
.
The diagnostic utility of RAS Q61R mutation-specific immunohistochemistry in epithelial-myoepithelial carcinoma
.
Am J Surg Pathol
.
2021 Jul 1
45
7
885
94
.
104.
Haller
F
,
Bieg
M
,
Will
R
,
Körner
C
,
Weichenhan
D
,
Bott
A
.
Enhancer hijacking activates oncogenic transcription factor NR4A3 in acinic cell carcinomas of the salivary glands
.
Nat Commun
.
2019 Jan 21
10
1
368
.
105.
Haller
F
,
Skálová
A
,
Ihrler
S
,
Märkl
B
,
Bieg
M
,
Moskalev
EA
.
Nuclear NR4A3 immunostaining is a specific and sensitive novel marker for acinic cell carcinoma of the salivary glands
.
Am J Surg Pathol
.
2019 Sep
43
9
1264
72
.
106.
Wong
KS
,
Mariño-Enriquez
A
,
Hornick
JL
,
Jo
VY
.
NR4A3 immunohistochemistry reliably discriminates acinic cell carcinoma from mimics
.
Head Neck Pathol
.
2021 Jun
15
2
425
32
.
107.
Coupland
SE
,
Foss
HD
,
Hidayat
AA
,
Cockerham
GC
,
Hummel
M
,
Stein
H
.
Extranodal marginal zone B cell lymphomas of the uvea: an analysis of 13 cases
.
J Pathol
.
2002 Jul
197
3
333
40
.
108.
Coupland
SE
,
Damato
B
.
Understanding intraocular lymphomas
.
Clin Exp Ophthalmol
.
2008 Aug
36
6
564
78
.
109.
Martinez-Lopez
A
,
Curiel-Olmo
S
,
Mollejo
M
,
Cereceda
L
,
Martinez
N
,
Montes-Moreno
S
.
MYD88 (L265P) somatic mutation in marginal zone B-cell lymphoma
.
Am J Surg Pathol
.
2015 May
39
5
644
51
.
110.
Lue
JK
,
O’Connor
OA
,
Bertoni
F
.
Targeting pathogenic mechanisms in marginal zone lymphoma: from concepts and beyond
.
Ann Lymphoma
.
2020 Sep 30
4
7
.
111.
Coupland
SE
,
Hummel
M
,
Müller
HH
,
Stein
H
.
Molecular analysis of immunoglobulin genes in primary intraocular lymphoma
.
Invest Ophthalmol Vis Sci
.
2005 Oct
46
10
3507
14
.
112.
Belhouachi
N
,
Xochelli
A
,
Boudjoghra
M
,
Lesty
C
,
Cassoux
N
,
Fardeau
C
.
Primary vitreoretinal lymphomas display a remarkably restricted immunoglobulin gene repertoire
.
Blood Adv
.
2020 Apr 14
4
7
1357
66
.
113.
Bonzheim
I
,
Sander
P
,
Salmerón-Villalobos
J
,
Süsskind
D
,
Szurman
P
,
Gekeler
F
.
The molecular hallmarks of primary and secondary vitreoretinal lymphoma
.
Blood Adv
.
2022 Mar 8
6
5
1598
607
.
114.
Riemersma
SA
,
Jordanova
ES
,
Schop
RF
,
Philippo
K
,
Looijenga
LH
,
Schuuring
E
.
Extensive genetic alterations of the HLA region, including homozygous deletions of HLA class II genes in B-cell lymphomas arising in immune-privileged sites
.
Blood
.
2000 Nov 15
96
10
3569
77
.
115.
Booman
M
,
Douwes
J
,
Glas
AM
,
Riemersma
SA
,
Jordanova
ES
,
Kok
K
.
Mechanisms and effects of loss of human leukocyte antigen class II expression in immune-privileged site-associated B-cell lymphoma
.
Clin Cancer Res
.
2006 May 1
12
9
2698
705
.
116.
Pulido
JS
,
Salomao
DR
,
Frederick
LA
,
Viswanatha
DS
.
MyD-88 L265P mutations are present in some cases of vitreoretinal lymphoma
.
Retina
.
2015 Apr
35
4
624
7
.
117.
Bonzheim
I
,
Giese
S
,
Deuter
C
,
Süsskind
D
,
Zierhut
M
,
Waizel
M
.
High frequency of MYD88 mutations in vitreoretinal B-cell lymphoma: a valuable tool to improve diagnostic yield of vitreous aspirates
.
Blood
.
2015 Jul 2
126
1
76
9
.
118.
Hiemcke-Jiwa
LS
,
Ten Dam-van Loon
NH
,
Leguit
RJ
,
Nierkens
S
,
Ossewaarde-van Norel
J
,
de Boer
JH
.
Potential diagnosis of vitreoretinal lymphoma by detection of MYD88 mutation in aqueous humor with ultrasensitive droplet digital polymerase chain reaction
.
JAMA Ophthalmol
.
2018 Oct 1
136
10
1098
104
.
119.
Yonese
I
,
Takase
H
,
Yoshimori
M
,
Onozawa
E
,
Tsuzura
A
,
Miki
T
.
CD79B mutations in primary vitreoretinal lymphoma: diagnostic and prognostic potential
.
Eur J Haematol
.
2019 Feb
102
2
191
6
.
120.
Lee
J
,
Kim
B
,
Lee
H
,
Park
H
,
Ho Byeon
S
,
Choi
JR
.
Whole exome sequencing identifies mutational signatures of vitreoretinal lymphoma
.
Haematologica
.
2020 Sep 1
105
9
e458
60
.
121.
Lacy
SE
,
Barrans
SL
,
Beer
PA
,
Painter
D
,
Smith
AG
,
Roman
E
.
Targeted sequencing in DLBCL, molecular subtypes, and outcomes: a Haematological Malignancy Research Network report
.
Blood
.
2020 May 14
135
20
1759
71
.
122.
Balikov
DA
,
Hu
K
,
Liu
CJ
,
Betz
BL
,
Chinnaiyan
AM
,
Devisetty
LV
.
Comparative molecular analysis of primary central nervous system lymphomas and matched vitreoretinal lymphomas by vitreous liquid biopsy
.
Int J Mol Sci
.
2021 Sep 16
22
18
9992
.
123.
Coupland
SE
,
Joussen
A
,
Anastassiou
G
,
Stein
H
.
Diagnosis of a primary uveal extranodal marginal zone B-cell lymphoma by chorioretinal biopsy: case report
.
Graefes Arch Clin Exp Ophthalmol
.
2005 May
243
5
482
6
.
124.
Mashayekhi
A
,
Shukla
SY
,
Shields
JA
,
Shields
CL
.
Choroidal lymphoma: clinical features and association with systemic lymphoma
.
Ophthalmology
.
2014 Jan
121
1
342
51
.
125.
Fend
F
,
Ferreri
AJ
,
Coupland
SE
.
How we diagnose and treat vitreoretinal lymphoma
.
Br J Haematol
.
2016 Jun
173
5
680
92
.
126.
Arai
A
,
Takase
H
,
Yoshimori
M
,
Yamamoto
K
,
Mochizuki
M
,
Miura
O
.
Gene expression profiling of primary vitreoretinal lymphoma
.
Cancer Sci
.
2020 Apr
111
4
1417
21
.
127.
Tan
WJ
,
Wang
MM
,
Ricciardi-Castagnoli
P
,
Chan
ASY
,
Lim
TS
,
Diagnostics
M
.
Cytologic and molecular diagnostics for vitreoretinal lymphoma: current approaches and emerging single-cell analyses
.
Front Mol Biosci
.
2020
;
7
:
611017
.
128.
Tanaka
R
,
Kaburaki
T
,
Taoka
K
,
Karakawa
A
,
Tsuji
H
,
Nishikawa
M
.
More accurate diagnosis of vitreoretinal lymphoma using a combination of diagnostic test results: a prospective observational study
.
Ocul Immunol Inflamm
.
2021 Apr 1
30
6
1354
60
.
129.
Wang
X
,
Su
W
,
Gao
Y
,
Feng
Y
,
Wang
X
,
Chen
X
.
A pilot study of the use of dynamic analysis of cell-free DNA from aqueous humor and vitreous fluid for the diagnosis and treatment monitoring of vitreoretinal lymphomas
.
Haematologica
.
2022 Sep 1
107
9
2154
62
.
130.
Khoury
JD
,
Solary
E
,
Abla
O
,
Akkari
Y
,
Alaggio
R
,
Apperley
JF
.
The 5th edition of the World Health organization classification of haematolymphoid tumours: myeloid and histiocytic/Dendritic Neoplasms
.
Leukemia
.
2022 Jul
36
7
1703
19
.
131.
Alaggio
R
,
Amador
C
,
Anagnostopoulos
I
,
Attygalle
AD
,
Araujo
IBd O
,
Berti
E
.
The 5th edition of the World Health Organization classification of haematolymphoid tumours: lymphoid neoplasms
.
Leukemia
.
2022 Jul
36
7
1720
48
.
132.
WHO Classification of Tumours Editorial Board
World Health organization classification of hematolymphoid tumors
5th ed
Lyon
International Agency for Research on Cancer
; in press.
133.
WHO Classification of Tumours Editorial Board
World Health organization classification of soft tissue and bone tumours
5th ed
Lyon
International Agency for Research on Cancer
2020
.