134
NASOPHARYNGEAL CARCINOMA STAGING BY COMPUTED TOMOGRAPHY AND MAGNETIC RESONANCE IMAGING Thesis Submitted to the Faculty of Medicine Alexandria University In partial fulfillment of the requirements for the degree of Master of Radiodiagnosis By Islam Mohamed El Gezeiry MBBCh, University of Alexandria Faculty of Medicine Alexandria University 2014

Nasopharyngeal Carcinoma Staging by CT and MRI

  • Upload
    -

  • View
    222

  • Download
    1

Embed Size (px)

DESCRIPTION

thesis by Dr. Islam El GezeiryHead and Neck Radiology

Citation preview

  • NASOPHARYNGEAL CARCINOMA

    STAGING BY COMPUTED TOMOGRAPHY AND

    MAGNETIC RESONANCE IMAGING

    Thesis

    Submitted to the Faculty of Medicine

    Alexandria University

    In partial fulfillment of the requirements for the degree of

    Master of Radiodiagnosis

    By

    Islam Mohamed El Gezeiry

    MBBCh, University of Alexandria

    Faculty of Medicine

    Alexandria University

    2014

  • NASOPHARYNGEAL CARCINOMA

    STAGING BY COMPUTED TOMOGRAPHY AND

    MAGNETIC RESONANCE IMAGING

    Presented by

    Islam Mohamed El Gezeiry

    For the Degree of

    Master of Radiodiagnosis

    Examiners Committee:

    Approved

    Prof. Dr. Shadia Abou Seif Helmy

    Professor of Radiodiagnosis

    Faculty of Medicine

    University of Alexandria

    Prof. Dr. Mahmoud Lotfy El-Sheikh

    Professor of Radiodiagnosis

    Faculty of Medicine

    University of Alexandria

    ....

    Prof. Dr. Ahmed Abdel Khalek Abdel Razek

    Professor of Radiodiagnosis

    Faculty of Medicine

    University of Mansoura

    Date: / /

  • SUPERVISORS

    Prof. Dr. Shadia Abou Seif Helmy

    Professor of Radiodiagnosis

    Faculty of Medicine

    University of Alexandria

    Prof. Dr. Mohamed Basiouny Atalla

    Professor of Otorhinolaryngology

    Faculty of Medicine

    University of Alexandria

    .

    Ass. Prof. Dr. Mohamed Eid Ibrahim

    Assistant Professor of Radiodiagnosis

    Faculty of Medicine

    University of Alexandria

  • ACKNOLEDGEMENT

    Praise to Allah, the Most Gracious and the Most Merciful

    Who Guides me to the right way

    First and foremost, my thanks are directed to Professor Dr.

    Shadia Abou Seif Helmy, Professor of Radiodiagnosis, Faculty of

    Medicine, University of Alexandria, for her unlimited help and

    continuous insistence on perfection, without her constant supervision, this

    thesis could not have achieved its present form.

    Many thanks and appreciation to Ass. Prof. Dr. Mohamed Eid

    Ibrahim, Assistant Professor in Radiodiagnosis, Faculty of Medicine,

    University of Alexandria, for his supervision and encouragement and for

    his kindness throughout the work.

    I am greatly indebted to Prof. Dr. Mohamed Basiouny Atalla,

    Professor of Otorhinolaryngology, Faculty of Medicine, University of

    Alexandria, for fruitful suggestions and wise guidance created this thesis.

    Last but not the least, special thanks to my parents and my wife

    for their continuous encouragement and kind support during the progress

    of this work, to whom I owe a lot of things more than I can count.

  • CONTENTS

    LIST OF ABBREVIATIONS -------------------- I

    LIST OF TABLES---------------------------------- II

    LIST OF FIGURES-------------------------------- III

    INTRODUCTION----------------------------------- 1

    AIM OF THE WORK------------------------------ 34

    PATIENTS AND METHODS--------------------- 35

    RESULTS--------------------------------------------- 38

    DISCUSSION---------------------------------------- 69

    SUMMARY------------------------------------------ 84

    CONCLUSION-------------------------------------- 87

    REFERENCES-------------------------------------- 88

    ARABIC SUMMARY-----------------------------

  • Abbreviations

    i

    ABBREVIATIONS

    NPC Nasopharyngeal Carcinoma

    EBV Epstein-Barr virus

    PPS Parapharyngeal space

    PMS Pharyngeal mucosal space

    MS Masticator space

    PS Parotid space

    CS Carotid space

    BS Buccal space

    RPS Retropharyngeal space

    DS Danger space

    PVS Perivertebral space

    LRP Lateral retropharyngeal

    LN Lymph nodes

    WHO World Health Organization

    AJCC American Joint Committee on Cancer

    CN Cranial nerve

    RT Radiotherapy

    IMRT Intensity Modulated Radiotherapy

    CRT Combined chemotherapy and radiotherapy

    RPLN Retropharyngeal lymph nodes

    PPF Pterygopalatine fossa

    PNS Perineural spread

  • ii

    LIST OF TABLES

    Table Page

    (1) Distribution of studied cases according to demographic data

    38

    (2) Distribution of studied cases according to side involved 38

    (3) Distribution of studied cases according to neck spaces involved

    38

    (4) Distribution of studied cases according to extension pattern

    39

    (5) Distribution of studied cases according to paranasal sinus involvement

    39

    (6) Distribution of studied cases according to pterygopalatine fossa involvement

    40

    (7) Distribution of studied cases according to skull base bone involvement pattern

    40

    (8) Distribution of studied cases according to foramina 40

    (9) Distribution of studied cases according to perineural spread

    41

    (10) Distribution of studied cases according to lymph nodal involvement

    41

    (11) Distribution of studied cases with cervical nodal metastases according to criteria of involvement

    41

    (12) Distribution of studied cases according to primary tumor T-stage

    42

    (13) Distribution of studied cases according to lymph nodes involvement N-stage

    42

    (14) Distribution of studied cases according to TNM stage 42

  • iii

    LIST OF FIGURES

    Figure Page

    (1) Graphic of the nasopharyngeal mucosal space seen from

    behind.

    3

    (2) A graphic of skull base from below shows spaces of

    suprahyoid neck relationships to skull base with emphasis on

    the pharyngeal mucosal space.

    4

    (3) Axial graphic of the nasopharyngeal mucosal space. 4

    (4) Mid-line sagittal graphic of the nasopharynx. 5

    (5) Lateral radiograph of the nasopharynx showing enlarged

    adenoids.

    5

    (6) A graphic showing the lateral wall structures of the

    nasopharynx.

    6

    (7) Spaces related to the nasopharynx. 8

    (8) A graphic of the neck as seen from left anterior view showing specific

    margins of the levels of the imaging-based classification for the lymph

    nodes of the neck

    10

    (9) A lateral radiograph of mildly enlarged adenoid. 12

    (10) The superior end of the para-pharyngeal space just before it

    abuts the skull base.

    13

    (11) Axial T2w image at the level of the opening of the Eustachian

    tube.

    15

    (12) Axial T1w image of the pharyngeal mucosal space at the

    level of the Eustachian tube opening.

    15

    (13) Sagittal T1w image of the pharynx. 16

    (14) Coronal enhanced fat-saturated T1 MR image. 16

    (15) Coronal enhanced fat-saturated T1w MR Image. 17

  • iv

    Figure Page

    (16) Axial T2w image of the nasopharynx with demonstration of

    the related spaces.

    17

    (17) Shaded triangular area corresponding to the supraclavicular

    fossa used in staging carcinoma of the nasopharynx.

    22

    (18) (Right)Axial T2wI MR shows large right NP mass.

    (Left) Axial bone CT showing enlarged right foramen ovale.

    24

    (19) Patient presenting with a left nasopharyngeal tumor. 25

    (20) Axial TSE T2-weighted image showing left nasopharyngeal

    tumor extending to the pterygo-palatine fossa.

    26

    (21) Contrast-enhanced SE T1-weighted MR images with fat

    saturation illustrating different pathways of extension in a

    patient suffering nasopharyngeal tumor.

    27

    (22) Contrast-enhanced T1-weighted MR images in a patient

    presenting with direct lateral extension through the

    pharyngobasilar fascia to the prestyloid compartment of the

    parapharyngeal space, and the infratemporal fossa, with

    infiltration of the pterygoid muscles.

    28

    (23) Spread of an advanced nasopharyngeal tumor. 29

    (24) (a) Non-enhanced T1-weighted MR image without fat

    saturation of a nasopharyngeal tumor infiltrating the clivus

    bone marrow.

    (b) Enhanced T1-weighted image with fat saturation, the

    tumor extends laterally to the jugular foramen and the

    hypoglossal canal.

    29

  • v

    Figure Page

    (25) Patient presenting with a nasopharyngeal tumor (a) revealed

    a serous otitis. (b) Posterior spread to the retropharyngeal

    space and parapharyngeal space. (c) A left retropharyngeal

    node and inferior extension to the oropharynx.

    30

    (26) (a) CT images illustrate a nasopharyngeal tumor extending to

    the foramen lacerum.

    (b) Note the enlargement of the foramen lacerum.

    31

    (27) (a) CT depicts small skull base erosions.

    (b) MRI non-enhanced T1-weighted sequence without fat

    saturation shows infiltration of sphenoid bone marrow.

    31

  • Introduction

    1

    INTRODUCTION

    Epidemiology (1)

    Nasopharyngeal carcinoma (NPC) is a rare malignancy in most

    parts of the world, with an incidence well under 1 per 100,000 person-

    years. Populations with elevated rates include the natives of Southeast

    Asia, the natives of the Arctic region, and the Arabs of North Africa

    and parts of the Middle East. (1)

    Sex and Age Distributions:

    In almost all populations, the incidence of NPC is 2- to 3- folds

    higher in males than in females. (1)

    In most low-risk populations, NPC incidence increases

    monotonically with increasing age. In contrast, in high-risk groups, the

    incidence peaks around ages 50 to 59 years and declines thereafter.

    Risk factors:

    1. Epstein-Barr virus:

    Primary EBV infection is typically subclinical; the virus is

    associated with later development of several malignancies,

    including NPC. (2)

    NPC patients were found to express antibodies

    against EBV. Antibody against EBV capsid antigen is now

    established as the basis of a screening test for NPC in high-risk

    populations. (3-8)

  • Introduction

    2

    2. Salt-Preserved Fish and Other Foods:

    NPC risk is also elevated in association with salt preserved

    fish and other preserved food items, including meats, eggs, fruits,

    and vegetables (excluding type I NPC). (9)

    3. Tobacco, and Other Smoke:

    The majority of case-control studies examining cigarette

    smoking and risk of NPC in a variety of populations reported an

    increased risk of 2- to 6-fold. In one U.S. study, an estimated two

    thirds of type I NPC was attributable to smoking, but risk of type II

    or III NPC was not associated with smoking. (10-20)

    4. Occupational Exposures:

    Occupational exposure to fumes, smokes, dusts, or

    chemicals overall was associated with a 2- to 6-folds higher risk of

    NPC in some studies. (15, 18, 21, 22)

    5. Other Exposures:

    Most studies investigating prior chronic ear, nose, throat, and

    lower respiratory tract conditions found that they approximately

    doubled the risk of NPC. (11-13)

    6. Familial Clustering:

    Familial aggregation of NPC has been widely documented in

    high-incidence, intermediate-incidence, and low-incidence

    populations. (23-39)

  • Introduction

    3

    ANATOMY OF THE NASOPHARYNX AND

    RELATED SPACES (40-42)

    The nasopharynx extends from the base of the skull to the lower

    border of the soft palate. The rigid pharyngobasilar fascia keeps it from

    collapsing at the back and sides. At the front the upper part communicates

    with the nose through the choanae, while below this the soft palate forms

    its anterior wall. The space between the lower border of the soft palate

    and the posterior pharyngeal wall through which the nasopharynx joins

    the oral part of the pharynx is the oropharyngeal isthmus. The soft palate

    becomes a mobile floor, like a trap door, when elevated during

    swallowing to meet the posterior wall, so closing the isthmus. (43)

    The nasopharynx communicates anteriorly with the posterior nasal

    choanal openings and downward with the oropharynx. (Fig. 1)

    Fig. 1 Graphic of the nasopharyngeal mucosal space/surface seen from behind shows

    communication of the nasopharyngeal mucosal space anteriorly with the posterior

    nasal choanal openings. (41)

    The roof and posterior margins are formed by the sphenoid bone,

    the clivus and the insertion of the prevertebral muscles into the skull base.

  • Introduction

    4

    Fig. 2 A graphic of skull base from below shows spaces of suprahyoid neck

    relationships to skull base with emphasis on the pharyngeal mucosal space. Notice the

    pharyngeal mucosal space abuts a broad area of the sphenoid and occipital bones. The

    foramen lacerum, the cartilaginous floor to the anteromedial horizontal petrous

    internal carotid artery canal, is within this abutment area. Malignant tumors of the

    nasopharyngeal mucosal space can access the intracranial compartment via the

    foramen lacerum. (41)

    Fig. 3 Axial graphic of the nasopharyngeal mucosal space (in blue) shows the

    superior pharyngeal constrictor and levator veli palatine muscles are within the space.

    The middle layer of the deep cervical fascia provides a deep margin to the space. The

    retropharyngeal space is behind and the parapharyngeal space is lateral to the

    pharyngeal mucosal space. (41)

  • Introduction

    5

    This roof shows downward slopping and is formed, cranially-to-

    caudally, by the basisphenoid, the basiocciput, and the anterior aspect of

    the first two cervical vertebrae. On this wall a prominence produced by a

    mass of lymphoid tissue, more prominent in childhood, is known as

    pharyngeal tonsils (adenoids). (40)

    (Fig. 4 & Fig. 5)

    Fig. 4 Mid-line sagittal graphic of the nasopharynx.

    Fig. 5 Lateral radiograph of the nasopharynx showing enlarged adenoids. (44)

    Prominent adenoids

  • Introduction

    6

    The lateral margins are made up by the pharyngeal constrictors and

    the torus tubaris, in the center of which is the opening of the Eustachian

    tube. (Fig. 4 & Fig. 6) The Eustachian tube enters the nasopharynx

    through the sinus of Morgagni, a defect in the anterior portion of the

    pharyngobasilar fascia, which is above the superior pharyngeal

    constrictor muscle and along the upper posterior border of the medial

    pterygoid plate. The levator veli palatini muscle also enters through the

    sinus of Morgagni.

    Fig. 6 A graphic showing the lateral wall structures of the nasopharynx. (45)

    Behind the ostium of the Eustachian tube is a deep recess, the

    pharyngeal recess (fossa of Rosenmller). The fossa of Rosenmller is

    the most common site of origin in nasopharyngeal carcinoma (NPC).

    The inverted J-shape of the torus tubaris explains why the fossa of

    Rosenmller appears posterior (on axial images) and superior (on coronal

    images) to the Eustachian tube orifice.

  • Introduction

    7

    The inferior margin of the nasopharynx is the level of the hard

    palate and Passavants muscle. This muscle is composed of fibers that

    arise laterally from the palatopharyngeus muscle and the lateral aspect of

    the posterior border of the hard palate. The fibers encircle the pharynx

    inside the superior constrictor muscle.

    The lateral nasopharyngeal walls are supported by the margins of

    the superior constrictor muscle and the pharyngobasilar fascia.

    Spaces related to the nasopharynx

    In the suprahyoid neck, three layers of deep cervical fascia are

    detected. These fascias are: (41)

    1- Superficial layer (investing fascia)

    2- Middle layer (buccopharyngeal fascia)

    3- Deep layer (prevertebral fascia)

    Spaces related to the nasopharynx are defined by these three layers

    of deep cervical fascia. (Fig. 7)

  • Introduction

    8

    Fig. 7 (PPS) parapharyngeal space, (PMS) pharyngeal mucosal space, (MS)

    masticator space, (PS) parotid space, (CS) carotid space, (BS) buccal space, (RPS)

    retropharyngeal space, (DS) danger space, (PVS) perivertebral space. (41)

    1. Parapharyngeal space (41)

    A slit-like space lateral to the nasopharynx extending down from

    the base of the skull. Potential space filled with loose connective tissue.

    The space is pyramidal in shape with apex directed towards the lesser

    cornu of the hyoid bone and the base towards the skull base. It extends

    from skull base to mid-oropharynx. It is lined medially by the superior

    constrictor muscles of the pharynx, tensor and levator veli palatini

    muscles. Laterally is lined by the mandible, the deep part of the parotid

    gland, and medial pterygoid muscle. Anteriorly lying is the buccinator

    muscle, the pterygoid, and the mandible. Posteriorly is the carotid sheath.

    The parapharyngeal space contains fat, ascending pharyngeal and internal

    maxillary arteries, pharyngeal venous plexus, and branches of the

    mandibular nerve.

  • Introduction

    9

    2. Retropharyngeal space and the prevertebral spaces (41)

    Lie between the nasopharynx and the vertebral bodies. The

    retropharyngeal space extends as a potential space from the skull base to

    about the level of T4 vertebral body and it serves as a conduit through

    which infections spread from the neck to the mediastinum. It contains fat

    and lymph nodes (lateral nodes of Rouvier and medial nodes).

    3. Nasopharyngeal masticator space (41)

    Lies lateral to the nasopharynx behind the posterior wall of the

    maxilla and extends from the base of the skull to the hyoid bone. It

    contains medial and lateral pterygoid muscles. No fascia defines this

    space which was previously named as infratemporal fossa. This term was

    used to describe the area between the pterygopalatine fossa and

    zygomatic arch. Medial to this, the roof is formed by the inferior surface

    of the middle cranial fossa and is pierced by the foramen ovale and

    foramen spinosum.

    Lymphatic drainage of the nasopharynx

    Lymphatic drainage is abundant in the nasopharynx, as evidenced

    by the high rate of nodal metastases found by the time of diagnosis of

    nasopharyngeal carcinomas. Three main groups of submucosal collecting

    pathways drain the pharynx, the superior, the middle, and the inferior

    pathways. The superior pathway drains the oropharynx, soft palate,

    Eustachian tube, fossa of Rosenmller, tympanic cavity, and nasal fossae.

    (45)

  • Introduction

    10

    Fig. 8 A graphic of the neck as seen from left anterior view. Drawing shows specific

    margins of the levels of the imaging-based classification for the lymph nodes of the

    neck. Note that the line of separation between levels I and II is the posterior margin of

    the submandibular gland. Separation between levels II and III and level V is the

    posterior edge of the sternocleidomastoid muscle. The line of separation between

    levels IV and V is the oblique line extending from the posterior edge of the

    sternocleidomastoid muscle to the posterior edge of the anterior scalene muscle.

    Posterior edge of internal jugular vein separates level IIA and IIB nodes. Carotid

    arteries separate levels III and IV from level VI. Top of manubrium separates levels

    VI and VII. (46, 47)

    Within the retropharyngeal space there are lateral retropharyngeal

    (LRP) lymph nodes of Rouvire. These nodes are the first nodes in the

    lymphatic drainage of the nasopharynx and maybe identified as discrete

    3-5 mm nodules. (48)

  • Introduction

    11

    The adenoids, or pharyngeal tonsils, are lymphatic tissue located in

    the midline roof of the nasopharynx. Prominent adenoids are typically

    present in children, and of such adenoids are not identified, the patient is

    either in an immune deficiency state or has immune deficiency syndrome.

    The maximal size of the adenoids occurs at about 5 years of age, around

    the time of puberty, gradual adenoidal involution normally begins. The

    majority of individuals have lost most this adenoidal tissues by 30 years

    of age. (49-51)

  • Introduction

    12

    RADIOLOGICAL ANATOMY OF THE

    NASOPHARYNX (41, 52)

    Plain X-ray film (48)

    Fig. 9 A lateral radiograph of mildly enlarged adenoid. (48)

    Conventional radiographs are used to evaluate patients with stridor,

    suspected retropharyngeal abscess or adenoid hypertrophy.

    Lateral soft tissue neck radiography may be helpful in making the

    diagnosis of nasopharyngeal masses. Perform the study during inspiration

    with neck held in normal extension. (53)

    The posterior wall of the pharynx forms a soft-tissue shadow

    curving posteroinferiorly below the body of the sphenoid and anterior to

    the cervical vertebrae. This shadow thins as it passes down anterior to the

    upper cervical vertebrae, measuring 3mm anterior to C4. Below this the

    wall is thicker but should not exceed the AP diameter of the cervical

  • Introduction

    13

    vertebrae. In children, lymphoid tissue results in a relatively thicker

    posterior wall, measuring up to 5 mm anterior to C4 and up to 12 mm

    anterior to C6 (Fig. 9). (48)

    Widening of the soft tissues observed between the radiolucent

    airway and the spine is pathologic until otherwise proven.

    CT anatomy of the nasopharynx (41)

    Axial contrast enhanced CT (CECT) of the nasopharynx:

    Fig. 10 The superior end of the parapharyngeal space just before it abuts the skull

    base, Notice the 4 major spaces surrounding the parapharyngeal space, the pharyngeal

    mucosal, masticator, parotid and carotid spaces. (41)

    CT evaluation of the nasopharynx is achieved with axial images

    with the patient lying supine. The head should be aligned carefully with

    the cranio-caudal axis, usually with the hard palate perpendicular to the

    table top and a scan plane parallel to the inferior orbital meatal plane.

    Poor positioning may result in an appearance that either simulates

    pathology or occasionally make pathology difficult to see. (54)

  • Introduction

    14

    At CT the tissue density of the fascia itself is inseparable from that

    of the adjacent musculature. The normal fat content of surrounding

    spaces compounded by associated muscle atrophy in the elderly patients

    will produce low density regions permitting a CT identification of the

    fascial planes. (48)

    The fat content of the paranasopharyngeal space allows one to

    easily identify it as a low density tissue plane lying between the pterygoid

    and pharyngeal musculature. Inferiorly the buccopharyngeal fascia is

    continuous with the covering of the nasopharynx and esophagus. The

    infratemporal fossa lies lateral to the paranasopharyngeal space. The

    infratemporal fossa is bounded laterally by the zygomatic arch. Within

    this space is most of the mandible, pterygoid, masseter, and parts of the

    temporalis muscle and deep lobe of the parotid gland. (42, 55)

    Other spaces defined by these fascial planes are important because

    their contents determine the cell of origin of some tumors. A potential

    space, the retropharyngeal space, exists between the pharyngobasilar

    fascia and the prevertebral fascia. This space contains the chains of lymph

    nodes lying to either side of the midline posteriorly. (42)

    Laterally the carotid sheath forms a posterolateral boundary to the

    retropharyngeal space. Within the carotid sheath lie the carotid vessels,

    sympathetic chains, and the vagus and proximal parts of XI and XII

    cranial nerves together with major deep lymphatic chains intimately

    associated with the jugular vein. (42)

  • Introduction

    15

    Normal MRI anatomy of the nasopharynx (41)

    The routine MR examination after obtaining scout images, sagittal,

    axial and coronal T1-weighted images, and axial T2-weighted images,

    with post-contrast (gadolinium-DTPA) injection T1-weighted images are

    obtained. Comparison of the pre- and post-contrast images is made to

    determine the areas of enhancement and to differentiate these areas from

    fat.

    Axial T2w Image of the nasopharyngeal mucosal space: (41)

    Fig. 11 Axial T2w image at the level of the opening of the Eustachian tube

    Axial T1w image of the nasopharyngeal mucosal space: (49)

    Fig. 12 Axial T1w image of the pharyngeal mucosal space at the level of the

    Eustachian tube opening.

  • Introduction

    16

    Sagittal T1w image of the pharynx:

    Fig. 13 Sagittal T1w image of the pharynx (48)

    Coronal T1w images of the nasopharynx: (41)

    Fig. 14 Coronal enhanced fat-saturated T1 MR image shows the pharyngeal mucosal

    space surface enhances.

    1. Soft palate 2. Adenoids 3. Middle turbinate 4. Inferior turbinate 5. Hard palate 6. Intrinsic muscle of

    tongue

    7. Genioglossus 8. Mandible 9. Myelohyoid muscle 10. Hyoid bone 11. Epiglottis 12. Vocal cord 13. Thyroid cartilage 14. Nasopharynx 15. Oropharynx 16. Corniculate cartilage 17. Arytenoid cartilage 18. Cricoid cartilage

    1

    2

  • Introduction

    17

    Fig. 15 Coronal enhanced fat-saturated T1w MR image reveals the enhancing sheet of

    mucosa with the torus tubarius (cartilaginous Eustachian tube) and lateral pharyngeal

    recess.

    Fig. 16 Axial T2w image of the nasopharynx with demonstration of the related

    spaces. (41)

    The superficial nasopharyngeal landmarks and deep fascial planes

    of the nasopharynx are normally bilaterally symmetrical. The most

    prominent of these is the torus tubaris, the cartilaginous part of the

    Eustachian tube, usually seen on MR as a medium- to high-intensity

    protuberance projecting into the aerated nasopharyngeal cavity. (42)

  • Introduction

    18

    In the mid- to upper nasopharynx, the tensor veli palatini and

    levator veli palatini muscle bundles are routinely shown by MR as they

    descend from their origin at the base of the skull to their insertion in the

    soft palate. At the transition from the nasopharynx to the oropharynx, the

    soft palate, tensor and levator palate, and pharyngeal constrictor muscles

    blend together, producing a low-intensity signal which surrounds the

    airway. (42)

    Although the tonsils may normally be quite large, they should not

    cause a mass effect involving the airway or deep soft tissue planes. A U-

    shaped ring of high-intensity tissue near the base of the tongue,

    corresponding to the lingual tonsil, is also routinely demonstrated on long

    TR sequences. (56)

    Below the nasopharyngeal mucosa and pharyngobasilar fascia,

    symmetrical fatty parapharyngeal spaces extend bilaterally from the base

    of the skull to the oropharynx. (42)

  • Introduction

    19

    PATHOLOGY OF THE NASOPHARYNGEAL

    CARCINOMA

    Normal histology of the nasopharynx

    The anterior and cranial portions of the nasopharynx are lined by

    respiratory mucosa with ciliated columnar epithelium with goblet cells

    and foci of metaplastic squamous epithelium. Squamous mucosa

    predominates in the lower nasopharynx adjacent to the oropharynx. Small

    seromucinous glands and aggregates of lymphoid tissue are present in the

    submucosa throughout the nasopharynx as a normal finding without

    qualifying as chronic inflammation. (57)

    Pathology of nasopharyngeal carcinoma (NPC)

    Three subtypes of NPC are recognized in the World Health Organization

    (WHO) classification 2005: (58-60)

    1. Keratinizing squamous cell carcinoma (type I)

    2. Non-keratinizing carcinoma:

    a) undifferentiated (type II)

    b) differentiated (type III)

    3. Basaloid squamous cell carcinoma

    Most cases in childhood and adolescence are type III, with a few

    type II cases. Type II and III are associated with elevated Epstein-Barr

    virus titers, but type I is not. Types II and III may be accompanied by an

    inflammatory infiltrate of lymphocytes, plasma cells, and eosinophils,

    which are abundant, giving rise to the term lymphoepithelioma. (61, 62)

  • Introduction

    20

    Staging

    The tumor, node, metastasis (TNM) classification of the American

    Joint Committee on Cancer is usually used to determine the tumor staging

    This latest TNM classification (AJCC 7th

    ed.) takes into account Hos

    modifications for NPC which utilizes the prognostic importance of

    affected nodes extending into the lower cervical and supraclavicular

    areas. (63)

    Definition of TNM

    Primary Tumor (T)

    TX Primary tumor cannot be assessed

    T0 No evidence of primary tumor

    Tis Carcinoma in situ

    T1 Tumor confined to the nasopharynx, or tumor extends to oropharynx

    and/or nasal cavity without parapharyngeal extension*

    T2 Tumor with parapharyngeal extension*

    T3 Tumor involves bony structures of skull base and/or paranasal

    sinuses

    T4 Tumor with intracranial extension and/or involvement of cranial

    nerves, hypopharynx, orbit, or with extension to the infratemporal

    fossa/masticator space

    *Note: Parapharyngeal extension denotes posterolateral infiltration of tumor.

  • Introduction

    21

    Regional Lymph Nodes (N)

    The distribution and the prognostic impact of regional lymph nodes

    spread from nasopharynx cancer, particularly of the undifferentiated type,

    are different from those of other head and neck mucosal cancers and

    justify the use of a different N classification scheme.

    NX Regional lymph nodes cannot be assessed

    N0 No regional lymph node metastasis

    N1 Unilateral metastasis in cervical lymph node(s), 6 cm or less

    in greatest dimension, above the supraclavicular fossa,

    and/or unilateral or bilateral, retropharyngeal lymph nodes,

    6 cm or less, in greatest dimension*

    N2 Bilateral metastasis in cervical lymph node(s), 6 cm or less

    in greatest dimension, above the supraclavicular fossa*

    N3 Metastasis in a lymph node(s)* >6 cm and/or to

    supraclavicular fossa*

    N3a Greater than 6 cm in dimension

    N3b Extension to the supraclavicular fossa**

    *Note: Midline nodes are considered ipsilateral nodes.

    **Note: Supraclavicular zone or fossa is relevant to the staging of

    nasopharyngeal carcinoma and is the triangular region originally

    described by Ho. It is defined by three points (Fig. 17):

    1. The superior margin of the sternal end of the clavicle.

    2. The superior margin of the lateral end of the clavicle.

    3. The point where the neck meets the shoulder.

    Note that this would include caudal portions of levels IV and VB. All

    cases with lymph nodes (whole or part) in the fossa are considered N3b.

  • Introduction

    22

    Fig. 17 Shaded triangular area corresponding to the supraclavicular fossa used in

    staging carcinoma of the nasopharynx. (64)

    Distant Metastasis (M)

    M0 No distant metastasis

    M1 Distant metastasis

    Stage grouping

    Stage 0 Tis N0 M0

    Stage I T1 N0 M0

    Stage II T1

    T2

    T2

    N1

    N0

    N1

    M0

    M0

    M0

    Stage III T1

    T2

    T3

    T3

    T3

    N2

    N2

    N0

    N1

    N2

    M0

    M0

    M0

    M0

    M0

    Stage IVA T4

    T4

    T4

    N0

    N1

    N2

    M0

    M0

    M0

    Stage IVB Any T N3 M0

    Stage IVC Any T Any N M1

  • Introduction

    23

    Presentation (41)

    Early stage NPC is difficult to diagnose clinically because of its

    hidden localization in the nasopharynx, and most patients present with

    advanced stage of the disease.

    Asymmetric neck swelling due to lymphadenopathy.

    Nasal symptoms: Epistaxis, bloody, rhinorhea, nasal

    obstruction.

    Ear symptoms: infection (Recurrent otitis media), deafness,

    and tinnitus.

    Ophthalmic symptoms: Diplopia, visual loss, squint, Ptosis.

    Headache.

    Blood in saliva.

    Facial numbness.

    Cranial nerve palsies; CN 9-12.

  • Introduction

    24

    IMAGING OF THE NASOPHARYNGEAL

    CARCINOMA

    Fig. 18 (Right) Axial T2w MR image shows large right NP mass, extending into

    pterygoid muscle (arrow), & posterior to surround ICA (open arrow). Mastoid fluid

    (curved arrow) due to Eustachian tube obstruction. (Left) Axial bone CT image

    showing enlarged right foramen ovale (arrow) from perineural V3 NPC spread with

    adjacent skull base destruction (open arrows). (Curved arrow) normal foramen ovale.

    (41)

    Aggressive mass centered in lateral pharyngeal recess of the

    nasopharynx (fossa of Rosenmller) with deep extension and cervical

    adenopathy. It arises from the lateral nasopharynx + posterolateral nasal

    cavity. It is usually several centimeters when diagnosed. (41)

    Morphology:

    Poorly marginated nasopharyngeal mucosal space mass with deep

    extension and invasion. (41)

  • Introduction

    25

    Extension patterns of the nasopharyngeal carcinoma: (65, 66)

    As explained, nasopharyngeal tumors spread along well-defined routes.

    1. Anterior spread

    Nasopharyngeal tumors often spread to the nasal fossa, which is

    not separated from the nasopharynx by any anatomic barrier (Fig. 19).

    From the nasal fossa, the tumor may easily infiltrate the pterygopalatine

    fossa through the sphenopalatine foramen (Fig. 20). The earliest sign of

    involvement of the pterygopalatine fossa is replacement of its normal fat

    content by tumoral tissue (Fig. 21). (66)

    Fig. 19 Patient presenting with a left nasopharyngeal tumor (anterior arrow), showing

    intermediate signal intensity on T2-weighted MR image. Note the anterior extension

    to the left choana (arrowhead). Associated serous otitis (posterior arrow). (66)

  • Introduction

    26

    Fig. 20 Axial TSE T2-weighted image showing left nasopharyngeal tumor extending

    to the pterygopalatine fossa (arrow) (66)

    Once tumor gains access to the pterygopalatine fossa, it can spread

    into (Fig. 21): (66)

    o The foramen rotundum along the maxillary nerve (V2)

    o The inferior orbital fissure and further the orbital apex, from where

    the tumor can extend intracranially through the superior orbital

    fissure.

    o The infratemporal fossa, where the masticator muscles are at risk

    of invasion. Erosion of the pterygoid process may occur. Perineural

    extension along the mandibular nerve (V3) into the foramen ovale

    and the endocranium is also possible (Fig. 22)

    o The vidian canal along the pterygoidien nerve and further to the

    petrous apex.

  • Introduction

    27

    Fig. 21 Contrast-enhanced SE T1-weighted MR images with fat saturation illustrating

    different pathways of extension in a patient suffering nasopharyngeal tumor. (a)

    Extension through the sinus of Morgagni, weakest point of the pharyngobasilar fascia

    (arrow). (b) Extension into the pterygopalatine fossa (arrow), neural crossroad within

    the skull base. (c) From the pterygopalatine fossa, the tumor extends to the inferior

    orbital fissure (arrow). (d) Extension to the infratemporal fossa (arrow) and to the

    pterygoid canal with perineural spread along the vidian nerve (arrowhead). (e)

    Perineural spread along the mandibular nerve (V3) extending to the foramen ovale

    (arrow) and the cavernous sinus (arrowhead) (66)

  • Introduction

    28

    2. Lateral spread

    Lateral extension to the parapharyngeal spaces can occur directly

    through the pharyngobasilar fascia (Fig. 22), or indirectly through the

    sinus of Morgagni, the fascias point of weakness. Further lateral spread

    involves the infratemporal fossa and the masticator space infiltrating the

    pterygoid muscles. From the masticator space, perineural extension along

    the mandibular nerve (V3) may occur, leading to infiltration of the

    foramen ovale and the cavernous sinus (Fig. 23). (66)

    Fig. 22 Contrast-enhanced T1-weighted MR images in a patient presenting with direct

    lateral extension through the pharyngobasilar fascia to the prestyloid compartment of

    the parapharyngeal space (a, arrow), and the infratemporal fossa, with infiltration of

    the pterygoid muscles (b, arrow) (66)

    3. Posterior spread

    Nasopharyngeal tumors can extend posteriorly to the

    retropharyngeal space and the prevertebral muscles (Fig. 23). Destruction

    of vertebral bodies is occasionally seen in very advanced tumors.

    Posterolateral extension may involve the jugular foramen and the

    hypoglossal canal (Fig. 24), with possible but rare spread to the posterior

    fossa. This posterior extension may result in hypoglossal nerve (XII)

    palsy. (66)

  • Introduction

    29

    Fig. 23 Spread of an advanced nasopharyngeal tumor. (a) Posterior extension to the

    retropharyngeal space and prevertebral muscles (arrow). (b) Lateral extension to the

    retrostyloid compartment of the parapharyngeal space, with encasement and

    narrowing of the internal carotid artery (arrow). (c) Extension to the infratemporal

    fossa (arrow) with intracranial spread into the cavernous sinus through the foramen

    ovale (arrowhead). (66)

    Fig. 24 Non-enhanced T1-weighted MR image of a nasopharyngeal tumor extending

    posteriorly, infiltrating the clivus bone marrow (a, arrowhead), well identified on this

    sequence by signal loss within the normally hyperintense bone marrow. (b) On the

    enhanced T1-weighted fat saturation image, the tumor is seen to extend laterally to the

    jugular foramen (anterior arrow) and the hypoglossal canal (XII) (posterior arrow). (66)

  • Introduction

    30

    4. Inferior spread

    Some nasopharyngeal tumors present with submucosal spread into

    the oropharynx, involving the tonsillar fossa (Fig. 24). This extension

    may take place submucosally and thus escape detection by endoscopy,

    although not detection by imaging. (66)

    Fig. 25 Patient presenting with a nasopharyngeal tumor, clinically revealed by a

    serous otitis (a, arrowhead) without lateral extension but a posterior spread to the

    retropharyngeal space (arrow) and posterior parapharyngeal space (b, arrow). (c) A

    left retropharyngeal node (arrow). Note the inferior extension to the oropharynx

    (arrowhead). (66)

    5. Superior spread

    Nasopharyngeal tumor can spread through the foramen lacerum,

    even if it is contained by the pharyngobasilar fascia. If the tumor extends

    to the tough fibrous cartilage which closes the foramen lacerum,

    intracranial extension may occur (Fig. 26).

    Superior spread with erosion of the clivus and the sphenoid sinus is

    also possible leading to intracranial extension (Fig. 27). (66)

  • Introduction

    31

    Fig. 26 (a) Coronal reconstruction of contrast enhanced CT image illustrating a

    nasopharyngeal tumor extending to the foramen lacerum. (b) Coronal bone window

    CT image. Note the enlargement of the foramen lacerum. (66)

    Fig. 27 Patient presenting with a nasopharyngeal tumor showing direct superior

    extension and infiltration of the sphenoid bone. (a) CT depicts small skull base clival

    erosions, (b) whereas MRI, in particular the non-enhanced T1-weighted sequence

    without fat saturation, shows a much more important infiltration of sphenoid bone

    marrow. (66)

    Intracranial extension of nasopharyngeal tumors is possible via

    different pathways such as the foramen lacerum, the foramen ovale and

    erosion of the skull base. Many studies have illustrated the good

    sensitivity of MRI to detect such extension, which is usually perineural.

    The frequency of intracranial abnormalities on MRI is 30%. (67)

    Nasopharyngeal tumors with intracranial extension are classified as T4

    tumors according to the TNM staging system. (67, 68)

    Intracranial spread is

    usually extra-axial, resulting in involvement of the cavernous and

    temporal meninges. (66)

  • Introduction

    32

    Staging and Treatment: (69)

    Taking into account the various TNM features, NPC patients are

    then staged accordingly from Stage 0 to Stage IV. Several features of

    note are:

    a) T3 disease indicates a patient is at least Stage III

    b) T4 disease places the patient at Stage IV

    c) N3 disease (i.e. single node >6 cm in size; supraclavicular

    nodes) indicates a patient is at least Stage IVb

    d) M1 disease places the patient at stage IVc.

    Correct staging enables the clinician to determine which treatment

    modality is best for the patient. A detailed discussion of the treatment

    options is beyond the scope of this paper. In brief, radiotherapy (RT) is

    the mainstay of treatment for NPC, as the differentiated and

    undifferentiated non-keratinizing squamous cell carcinomas (formerly

    named type II and III) are very radiosensitive. (70, 71)

    Conventional

    external beam RT was the traditional method of treatment. However, the

    tumor could not be maximally irradiated without damaging adjacent

    structures such as the parotid glands. With the advent of conformal

    techniques, and in particular, intensity-modulated radiotherapy(IMRT),

    doses of up to 70 Grays may be delivered with relative sparing of the

    adjacent soft tissues. Limitations still remain with very large tumors, for

    example, T4 tumors, where NPC may be so close to vital structures such

    as the optic chiasm that the latter cannot be spared if the full RT dose

    were to be administered.

    Combined chemotherapy using platinum-based drugs and

    radiotherapy (CRT) is given for patients with T3 disease and nodal

  • Introduction

    33

    disease >N1. Patients with T4 and N3 disease may receive neoadjuvant

    chemotherapy with platinum-based combination chemotherapy followed

    by definitive RT with concurrent chemotherapy. (71)

    Patients with T1/T2

    N1 disease are also treated with CRT although this is a controversial topic

    and beyond the scope of this article.

  • Aim of the work

    34

    AIM OF THE WORK

    The aim of this work is to describe the role of state of the art cross

    sectional imaging computed tomography and magnetic resonance

    imaging (CT & MRI) in the staging of nasopharyngeal carcinoma.

  • Patients and methods

    35

    PATIENTS AND METHODS

    This study included 20 patients presenting with pathologically

    proven nasopharyngeal carcinoma referred to the Radiodiagnosis

    Department at the Alexandria Main University Hospital.

    All the studied patients were subjected to the following:

    1. Complete history taking.

    2. Thorough clinical ENT examination.

    3. The medical ethics were considered. The patient was aware of the

    examination, patient's approval was obtained.

    4. Multi-detector Computed Tomography examination especially bone

    algorithm as well as post contrast sequences.

    Patients lying supine were instructed to take shallow breaths and

    refrain from swallowing during scanning. MDCT was performed on a 16-

    MDCT scanner (Philips MX16, Philips Healthcare) with tube voltage,

    120 kV; effective tube current, 150 mAs; collimation, 0.75 mm; table

    feed, 12 mm/rotation; and rotation time, 0.5 second. The effective

    radiation dose for a typical scanning range of 250 mm was 3.6 mSv for

    men and 4.1 mSv for women. A non-ionic contrast agent ULTRAVIST,

    Bayer (each ml of injection contains 769 mg iopromide, equivalent to 370

    mg iodine) was injected at a flow rate of 1 ml/sec. for 50 seconds then a

    waiting time for 50 seconds then inject 50 ml at 2.5 ml/sec. and start scan

    at the end of injection. The scanning range started from top of the frontal

    sinus base to the tracheal bifurcation. Data set was reconstructed using a

    standard soft-tissue (B 40) convolution kernel with a slice thickness of 1

    mm (0.7-mm reconstruction increment). For the assessment of bone and

  • Patients and methods

    36

    cartilage, additional data set was reconstructed using a sharp (bone)

    convolution kernel (B 70).

    5. MRI examination:

    a. Axial: T1, T2 and T2 Fat Saturation images.

    b. Coronal: T1 and T2 Fat Saturation images.

    c. Sagittal: T2w images.

    d. Post-Contrast T1w Fat Saturation images in 3 planes.

    MR imaging was performed with a 1.5 Tesla whole-body MR imaging

    system (Philips Achieva 1.5T, the Netherlands), by using a 4-channel

    phased array head and neck coil. The following parameters were used:

    Pre-contrast axial and coronal T1-images were obtained with SE

    450/15, 90, 2 excitations, a 22-cm field of view (FOV), a 256 256

    matrix, a 3-mm-thick section and a 0.9-mm gap.

    Axial and sagittal T2-images were obtained with SE 4500/88, 180,

    3 excitations, a 22-cm field of view (FOV), a 256 256 matrix, a 3-

    mm-thick section and a 0.9-mm gap.

    Axial and coronal fat-suppressed T2-weighted sequence obtained

    with SE 2500 ms/100 ms; echo-train length, 15; 22-cm field of

    view (FOV), a 256 256 matrix, 4-mm-thick section, with no

    intersection gap; and 256 256 matrix size.

    Post-contrast medium (Dotarem ,Guerbet (0.1 mmol/kg)) Axial,

    coronal, and sagittal T1w scan with fat suppression images were

    obtained with SE 500/22 ms, 2 excitations, a 22-cm field of view

    (FOV), a 256 256 matrix, 3-mm section thickness and 0.5-mm

    gap.

  • Patients and methods

    37

    6. Endoscopic examination and biopsy from the suspected area with

    routine histopathologic examination.

    7. Correlation with pathological data.

  • Results

    38

    RESULTS

    Table (1): Distribution of studied cases according to demographic

    data (n=20)

    No. %

    Age (years)

    20 >30 3(15%) 15.0

    30 >40 3(15%) 15.0

    40 >50 6(30%) 30.0

    50 >60 3(15%) 15.0

    60 >70 5(25%) 25.0

    Sex

    Male 12(60%) 60.0

    Female 8(40%) 40.0

    Table (2): Distribution of studied cases according to side (n=20)

    No. %

    Side

    Right 8(40%) 40.0

    Left 11(55%) 55.0

    Diffuse 1(5%) 5.0

    Table (3): Distribution of studied cases according to neck spaces

    involvement (n=20)

    No.

    % CT MRI

    Space

    Retropharyngeal space (RPLN) 4(20%) 8(40%) 40.0

    Carotid sheath 1(5%) 1(5%) 5.0

    Parapharyngeal space 7(35%) 8(40%) 40.0

    Masticator space 3(15%) 3(15%) 15.0

  • Results

    39

    Table (4): Distribution of studied cases according to extension

    pattern (n=20)

    Extension No.

    % CT MRI

    Anteriorly

    Nasal choana 8(40%) 8(40%) 40.0

    Inferiorly

    Oropharynx 5(25%) 7(35%) 35.0

    Superiorly

    Intracranial extension 7(35%) 8(40%) 40.0

    Perineural spread 6(30%) 10(50%) 50.0

    Posteriorly

    Retropharyngeal space (RPLN) 4(20%) 8(40%) 30.0

    Posterolaterally

    Carotid sheath 1(5%) 1(5%) 5.0

    Laterally

    Parapharyngeal space 7(35%) 8(40%) 40.0

    Masticator space 3(15%) 3(15%) 15.0

    Table (5): Distribution of studied cases according to paranasal

    sinuses involvement (n=20)

    No. of cases

    % CT MRI

    Paranasal sinus involvement 4 6 30.0

    Sphenoid sinus 4(20%) 6(30%) 30.0

    Maxillary antrum 1(5%) 1(5%) 5.0

  • Results

    40

    Table (6): Distribution of studied cases according to

    pterygopalatine fossa involvement (n=20)

    No.

    % CT MRI

    Pterygopalatine fossa 7(35%) 9(45%) 45.0

    Right 2(10%) 3(15%) 15.0

    Left 5(25%) 6(30%) 30.0

    Table (7): Distribution of studied cases according to skull base bone

    involvement pattern (n=20)

    No. of cases

    % CT MRI

    Bone 18 17

    Lytic 7(40%) 8(40%) 40.0

    Sclerotic 5(25%) 3(15%) 25.0

    Mixed sclerosis and erosion 6(30%) 6(30%) 30.0

    Table (8): Distribution of studied cases according to neural

    foraminal involvement (n=20)

    No. %

    Foramina 17(85%) 85.0

    Ovale 8(40%) 40.0

    Lacerum 9(45%) 45.0

    Jugular 1(5%) 5.0

    Sphenopalatine 1(5%) 5.0

    Rotundum 1(5%) 5.0

  • Results

    41

    Table (9): Distribution of studied cases according to perineural

    spread (n=20)

    No.

    % CT MRI

    Perineural spread

    Along V3

    Along V2

    Along vidian nerve

    Facial nerve(VII)

    8(40%)

    5(25%)

    2(10%)

    3(5%)

    0(0%)

    11(55%)

    6(30%)

    3(15%)

    4(5%)

    1(5%)

    50.0

    30.0

    15.0

    20.0

    5.0

    Table (10): Distribution of studied cases according to lymph nodal

    involvement (n=20)

    No.

    % CT MRI

    Supraclavicular lymph nodes 5(25%) 5(25%) 25.0

    Retropharyngeal lymph nodes (RPLN) 4(20%) 8(40%) 60.0

    Cervical lymph nodes 14(70%) 14(70%) 70.0

    Parotid LN 3(15%) 3(15%) 15.0

    Table (11): Distribution of studied cases with cervical lymph nodal

    metastases according to criteria of involvement (n=20)

    No. %

    CT MRI

    Size enlargement 16 16 80.0

    Necrosis 5 5 25.0

    Extra-capsular extension 3 3 15.0

  • Results

    42

    Table (12): Distribution of studied cases according to primary tumor

    T-stage (n=20)

    No. %

    Primary tumor

    T1 0(0%) 0.0

    T2 0(0%) 0.0

    T3 5(25%) 25.0

    T4 15(75%) 75.0

    Table (13): Distribution of studied cases according to lymph nodes

    involvement N-stage (n=0)

    No. %

    Lymph nodes

    N0 4(20%) 20.0

    N1 8(40%) 40.0

    N2 3(15%) 15.0

    N3a 0(0%) 0.0

    N3b 5(25%) 25.0

    Table (14): Distribution of studied cases according to TNM stage

    (n=20)

    No. %

    TNM Stage

    I 0(0%) 0.0

    II 0(0%) 0.0

    III 3(15%) 15.0

    IVa 11(55%) 55.0

    IVb 3(15%) 15.0

    IVc 3(15%) 15.0

  • Results

    43

    Case 1:

    (A) (B)

    (C) (D)

  • Results

    44

    (E) (F)

    (G) (H)

  • Results

    45

    Fig. 28 Case 1: 28 years old male patient with right side

    nasopharyngeal carcinoma.

    (A and B) Axial T2w and T2w fat suppression images: A right

    nasopharyngeal iso-intense heterogeneous mass (white arrow)

    obliterating the parapharyngeal space fat and the levator veli palatini

    muscle, and crossing the midline along the posterior pharyngeal wall.

    (C and D) Pre- and post-contrast axial T1w images: The mass

    lesion fills anteriorly the scaphoid fossa (white block arrow) of the

    pterygoid process. Moderate post-contrast enhancement of the mass is

    noted (D image).

    (E and F) Axial CT bone window and MR T1w post-contrast

    images: In the (E) image (orange curved arrow) represents the tumor

    entering through the sphenopalatine foramen into the pterygopalatine

    fossa (red arrow head), and hence reaching into the vidian canal (black

    arrow). In the (F image) the moderately enhancing tumor filling the

    pterygopalatine fossa (red arrow head) and reaching the vidian canal

    (black arrow).

    (G and H) Pre- and post-contrast sagittal T1w images: The lesion

    extends anteriorly to the ipsilateral choana, and inferiorly reaches the

    junction of the naso- and oro-pharynx (yellow arrow).

  • Results

    46

    Case 2:

    (A) (B)

    (C) (D)

  • Results

    47

    (E)

    Fig. 29 Case 2: 50 years old female patient with left sided

    nasopharyngeal carcinoma.

    (A) Axial CT bone window image: showing lytic lesion eroding the

    clivus (red arrow) and bony borders of the vidian canal (black arrow) and

    foramen rotundum (white arrow).

    (B) Axial T2w image: showing intermediate signal mass lesion with

    extension to the left sphenoidal sinus which showed retained secretions

    (blue star), near total encasement of the petrous segment of the internal

    carotid artery (yellow arrow).

    (C) Axial T1w image: showing a nasopharyngeal carcinoma of the

    pterygopalatine fossa invasion (green arrow).

  • Results

    48

    (D) Post-contrast sagittal T1w fat saturation image: showing

    posterior invasion of the clivus with intracranial extension effacing the

    pre-pontine cistern (blue arrow).

    (E) Enhanced coronal T1w fat saturation image: The

    nasopharyngeal mass reaches the cavernous sinus through the vidian

    canal and the foramen rotundum (vidian and maxillary (V2) nerves

    Perineural spread) with encasement of the internal carotid artery (red

    arrow head) and enhancement of the maxillary nerve (yellow arrow

    head).

  • Results

    49

    Case 3:

    (A) (B)

    (C) (D)

  • Results

    50

    (E)

    Fig. 30 Case 3: a 30 years old female patient with left sided

    nasopharyngeal carcinoma.

    (A) Axial CT bone window image: It shows widening of the petro-

    clival fissure (black arrow) and the foramen lacerum (white arrow).

    (B) Axial T1w image: a mucosal based mass lesion obliterating the left

    fossa of Rosenmller showing T1 intermediate signal.

    (C) Post-contrast axial 3D GRE T1w image: The moderately

    enhancing tumor reaches along the lateral wall of the nasopharynx to the

    pterygopalatine fossa (red arrow head) then along the vidian canal

    representing perineural spread along the vidian nerve (red arrow).

    (D) Post-contrast axial T1w fat saturation image: (yellow arrow)

    Left sided level II enlarged metastatic lymph node with foci of necrosis

    showing post-contrast enhancement.

  • Results

    51

    (E) Post-contrast sagittal T1w fat saturation image: Superior

    extension of the tumor to the floor of the sphenoid sinus with minimal

    intra-sinus extension (green arrow).

  • Results

    52

    Case 4:

    (A) (B)

    (C) (D)

  • Results

    53

    (E)

    Fig. 31 Case 4: a 40 years old male patient with left sided

    nasopharyngeal carcinoma.

    (A) Axial T1w image: A mass lesion is noted growing and expanding

    the left lateral nasopharyngeal recess (Fossa of Rosenmller), showing

    intermediate to low T1 signal (white arrow).

    (B) Axial 3D GRE T1w image: The nasopharyngeal carcinoma grows

    anteriorly to reach the pterygopalatine fossa (yellow arrow) and laterally

    to reach the widened foramen ovale (red arrow) and grows along the

    mandibular division of the trigeminal nerve V3 (perineural spread) in the

    foramen ovale to reach intracranial cavity.

    (C and D) Axial T2w and post-contrast T1w fat saturation

    images: demonstrates the intracranial extra-axial left temporal

    component of the nasopharyngeal carcinoma (blue arrow). Involvement

  • Results

    54

    of the cavernous sinus with total encasement of the still patent internal

    carotid artery (black arrow).

    (E) Post-contrast coronal T1w fat saturation image: The

    nasopharyngeal lesion (white arrow) extends superiorly through the

    widened foramen ovale (orange arrow) into the intracranial extra-axial

    temporal mass lesion (green arrow).

  • Results

    55

    Case 5:

    (A) (B)

    (C) (D)

  • Results

    56

    (E)

    Fig. 32 Case 5: a 48 years old male patient with left sided

    nasopharyngeal carcinoma.

    (A) Sagittal reconstruction bone window CT image: Sclerosis and

    infiltration of the bone marrow of the clivus (black arrow).

    (B) Axial T1w image: Perineural spread along the mandibular division

    of the left trigeminal nerve (V3), through the foramen ovale (yellow

    arrow) and infiltration of the clival bone marrow (black arrow).

    (C) Axial T2w image: An intracranial extra-axial temporal component

    (green arrow), reaching the cavernous sinus encasing the patent internal

    carotid artery siphon (blue arrow).

    (D) Post-contrast coronal T1w fat saturation image: Shows the

    nasopharyngeal lesion with perineural spread along the V3 division of the

    mandibular nerve through the foramen ovale (yellow arrow) widening the

  • Results

    57

    foramen and extending superiorly to an extra-axial temporal mass (green

    arrow) reaching the region of the trigeminal ganglion.

    (E) Post-contrast sagittal T1w image: shows infiltration of the clival

    bone marrow with post-contrast enhancement (black arrow).

  • Results

    58

    Case 6:

    (A) (B)

    (C) (D)

  • Results

    59

    Fig. 33 Case 6: a 65 years old male patient with right side

    nasopharyngeal carcinoma.

    (A and B) Axial T2w and post-contrast T1w fat saturation

    images: A right-sided nasopharyngeal carcinoma (black arrow) with

    anterior extension to the pterygoid plates and scaphoid fossa (yellow

    arrow).

    (C) Axial Diffusion weighted image: Restricted diffusion of right

    nasopharyngeal mass (white arrow)

    (D) Axial T2w image: bilateral jugular chain lymph nodes (red arrows)

    with necrosis in the left one.

  • Results

    60

    Case 7:

    (A) (B)

    (C) (D)

  • Results

    61

    (E) (F)

    Fig. 34 Case 7: a 38 years old male patient with diffuse bilateral

    nasopharyngeal carcinoma.

    (A) Sagittal reconstruction CT bone window image: shows full

    thickness moth eaten erosion of the clivus (white arrows).

    (B) Contrast-enhanced axial CT image: shows a nasopharyngeal

    heterogeneously enhancing mass lesion obliterating fossae of

    Rosenmller bilaterally and filling the nasopharyngeal cavity (red

    arrows).

    (C) Axial T2w image: shows anterior extension of the hyperintense

    nasopharyngeal lesion through both choanae into nasal cavities (orange

    arrows)

    (D and E) Axial T2w and Diffusion weighted images: show

    enlarged hyperintense right sided retropharyngeal lymph node which

    showed restricted diffusion (green arrows).

  • Results

    62

    (F) Post-contrast sagittal T1w image: shows the enhancing pre-clival

    nasopharyngeal mass (blue arrow) infiltrating into the clivus (red arrow)

    with pre-pontine enhancing component (yellow arrow).

  • Results

    63

    Case 8:

    (A) (B)

    (C) (D)

  • Results

    64

    (E)

    Fig. 35 Case 8: A 62 years old female patient of left sided

    nasopharyngeal carcinoma

    (A) Axial T2w image: shows a left sided iso-intense nasopharyngeal

    mass lesion obliterating the left fossa of Rosenmller (orange arrow).

    (B) Post-contrast axial T1w fat saturation image: shows superior

    extension of the moderately enhancing tumor with total encasement of the

    internal carotid artery (blue arrow).

    (C) Post-contrast axial T1w fat saturation image: shows inferior

    extension of the enhancing nasopharyngeal tumor along the lateral

    pharyngeal wall reaching the oropharynx (red arrow).

    (D and E) Pre-contrast sagittal T1w and post-contrast T1w fat

    saturation images: shows hypo-intense infiltration of the tumor in the

    bone marrow of the clivus with post contrast enhancement (yellow

    arrow).

  • Results

    65

    Case 9:

    (A) (B)

    (C) (D)

  • Results

    66

    Fig. 36 Case 9: A 50 years old male patient with left sided

    nasopharyngeal carcinoma.

    (A) Axial T2w image: shows a left sided isointense nasopharyngeal

    mass lesion (white arrow) with anterior extension through the left choana

    into the left nasal cavity (red arrow).

    (B) Axial T1w image: The hypointense nasopharyngeal mass extends

    anteriorly to the pterygopalatine fossa with obliteration of the

    pterygopalatine fossa fat signal (orange arrow).

    (C) Post-contrast sagittal T1w image: shows moderately enhancing

    nasopharyngeal lesion (green arrow) with superior extension infiltrating

    the clival marrow (yellow arrow) and the sphenoid sinus (blue arrow).

    (D) Axial T2w image: shows a left sided lateral retropharyngeal

    enlarged lymph node showing hyper-intense signal (red arrow).

  • Results

    67

    Case 10

    (A) (B)

    (C) (D)

  • Results

    68

    Fig. 37 Case 10: A 65 years old male patient of right sided

    nasopharyngeal carcinoma.

    (A and B) Axial T2w and post-contrast T1w images: show a right

    sided nasopharyngeal mass lesion showing T2 iso-intensity with

    moderate post-contrast enhancement which infiltrates the levator veli

    palatini and medial pterygoid muscles with obliteration of the

    parapharyngeal fat (white arrow). The tumor infiltrates posteriorly the

    anterior surface of the right prevertebral muscle (yellow arrow).

    (C) Post-contrast axial T1w image: shows enhancing intracranial

    extra-axial temporal component (red arrow).

    (D) Post-contrast coronal T1w fat saturation image: The

    moderately enhancing nasopharyngeal lesion (blue arrow) extends

    superiorly through the right foramen ovale (black arrow) with intracranial

    extra-axial component reaching the cavernous sinus and encasing the

    internal carotid artery (red arrow).

  • Discussion

    69

    DISCUSSION

    Nasopharyngeal carcinoma (NPC) is a rare malignancy in most parts

    of the world, with an incidence well under 1 per 100,000 person-years.

    Populations with elevated rates include the natives of Southeast Asia, the

    natives of the Arctic region, and the Arabs of North Africa and parts of the

    Middle East. (1)

    The present study included 20 patients with pathologically proven

    NPC as 12 (60%) males and 8 (40%) females with a mean age of 45.9 years.

    Parkin DM et al (2002) stated that in almost all populations surveyed,

    the incidence of NPC is 2- to 3-folds higher in males than in females. (72)

    In

    our study, male to female ratio was of about 1.5 folds higher in males than in

    females.

    In most low-risk populations, NPC incidence increases monotonically

    with increasing age. (73-75)

    In the contrary, in high-risk groups, the incidence

    peaks around ages 50 to 59 years and declines thereafter. (76, 77)

    In our study,

    patients age ranged from 24 to 65 years with bimodal peaks of 6 and 5 cases

    for the 5th

    and 7th

    decades of life respectively.

    Of the studied NPC cases, 8 cases (40%) were seen on the right side,

    11 cases (55%) on the left side, and 1 case (5%) diffusely infiltrating both

    sides and crossing the midline.

  • Discussion

    70

    Since NPC is diagnosed by endoscopy, the foremost role of CT or

    MRI is to determine the extent of primary tumor and the presence of

    metastatic adenopathy. (78)

    Accurate assessment of the disease extent facilitates appropriate

    treatment planning and prognosis. (79)

    NPC is generally iso-dense to muscle on non-enhanced CT. It is

    usually hypo- to iso-intense and relatively hyper-intense to muscles on T1-

    weighted and T2-weighted MR images, respectively. Mild to moderate

    tumor enhancement is evident following intravenous contrast injection on

    both CT and MRI.

    Ng SH et al (1997) stated that CT and MRI findings were essentially

    in agreement in patients whose disease was limited to the nasopharyngeal

    cavity, but not those with tumor spreading beyond the boundaries of the

    nasopharynx. (80)

    The pharyngobasilar fascia, the medial border of the parapharyngeal

    space, is normally seen on MRI and not on CT. Involvement of the

    parapharyngeal space denotes at least T2 stage of the tumor. (81-84)

    In NPC,

    parapharyngeal space involvement can be assessed directly by MRI, which

    shows tumor displacement or infiltration of the pharyngobasilar fascia or

    extension through the sinus of Morgagni. (81, 85)

    In contrast, involvement of

    the parapharyngeal space by CT is inferred indirectly by an abnormal soft

    tissue deforming the parapharyngeal fibro-fatty tissue plane or by outward

    bulging of an imaginary line between the medial pterygoid plate and the

    lateral border of the carotid artery. (84, 86)

  • Discussion

    71

    In our study the involvement of the parapharyngeal space was

    demonstrated in 7 cases by CT and in 8 cases by MRI. King AD et al (2000)

    found that CT scanning suggested the presence of parapharyngeal tumor

    extension more frequently than MRI because of its inability to distinguish

    the primary tumor from lateral retropharyngeal nodes, and direct tumor

    invasion of the parapharyngeal region from tumor compression. (87)

    Xie C et

    al (2004) in a study on 69 patients found that there was no difference

    between CT and MRI in demonstrating the invasion of the parapharyngeal

    space. (88)

    Further laterally, the tumor may spread into the masticator space.

    Anatomic masticator space involvement affects the overall survival and local

    relapse-free survival of patients with NPC. The frequency of masticator

    space involvement in NPC is 19.7% as declared by Abdel Khalek Abdel

    Razek A. et al (2012). (89)

    Infiltration of the medial and lateral pterygoid

    muscles, infratemporal fat, and temporalis muscle is found when tumors

    extend laterally from the parapharyngeal space, pterygoid base, or the

    pterygo-maxillary fissure. (66, 90)

    When the muscles of mastication (notably

    the medial and lateral pterygoid muscles) are involved, the patient often

    complains of trismus (Chong VF 1997). (91)

    The mandibular nerve within the

    masticator space may also be infiltrated, resulting in denervation atrophy of

    the muscles of mastication. MRI features of denervation atrophy of these

    muscles appear as T2 hyperintensity with asymmetrically reduced bulk of

    the muscle of the affected side compared to the normal side (Chong VF et al

    2008). (92)

    In our study, the masticator space was involved in 3 cases (15%) and

    it was as well seen in both CT and MRI. 2 cases showed involvement of the

  • Discussion

    72

    pterygoid fossa and the pterygo-maxillary fissure, and in the third one

    showed denervation changes involving the ipsilateral masticator muscles,

    sequel to mandibular nerve affection in the masticator space.

    Further posterolateral spread may also involve the carotid space and

    encase the carotid artery. (93)

    Carotid artery encasement is defined as tumor

    tissue surrounding >270o of the vessel circumference.

    (69) This becomes

    important in the follow-up setting, where surgical resection (e.g.

    nasopharyngectomy or lymph node dissection) may be contemplated. The

    patient is deemed inoperable if this is present, as the surgeon cannot remove

    all the tumor tissue. Other potential issues that may result from encasement

    include vascular invasion and potential carotid artery blow-outs post-

    radiotherapy. Different criteria for detecting carotid artery involvement on

    CT are suggested, and accordingly differs the sensitivity and accuracy in

    detecting the vascular encasement. The use of loss of the fat plane between

    the tumor and the carotid artery leads to very high false positive rates but

    with a sensitivity of 100%. (94-96)

    Others, suggest the use of tumor in contact

    with one-half of the circumference of the artery and loss of the tissue planes,

    with a much less false positive rates. (95, 97)

    Other imaging characteristics are

    carotid artery deformation, compression and segmental obliteration of the

    fat.

    Kraus DH et al (1992) declared that MRI was superior to CT in

    determining carotid artery involvement. (82)

    Yousem DM et al (1995), in a

    study of carotid artery encasement, sensitivity of MRI was 100% and

    specificity 88%. (98)

    Sarvanan K et al (2002) studied encasement of >270

    degrees and loss of fat planes. Sensitivity reached 75% and specificity

    100%. (99)

  • Discussion

    73

    In our study we depicted only 1 case (5%) of carotid space

    involvement by total encasement of the still patent carotid artery by NPC,

    and that was seen by CT and by MRI as well.

    In patients with NPC, paranasal sinuses involvement denotes a tumor

    of at least stage T3. Paranasal sinus opacity is a common finding seen on

    CT; it is occasionally difficult to differentiate whether it represents tumor

    invasion or sinonasal secretions. On MRI, hydrated secretions within the

    obstructed paranasal sinuses are of increased signal on T2-weighted images.

    Thus, high-signal secretions can be differentiated from an intermediate

    signal-intensity tumor. (82)

    Desiccated or mixed sinonasal secretions may

    exhibit signal characteristics similar to those of tumor on both T1- and T2-

    weighted images; contrast-enhanced MRI is then helpful because tumor

    within the sinus enhances whereas sinonasal secretions do not enhance and

    are surrounded by a rim of strongly enhancing sinus mucosa. (100, 101)

    In late

    involvement of the sinuses, erosion of the sinus walls is a straightforward

    exercise except in early cases. CT is superior to MRI in visualizing erosions

    of paranasal sinuses floor. (53, 102)

    In our study, we found that paranasal sinuses involvement was

    demonstrated in the sphenoid sinus in 6 cases while in CT 4 cases showed

    invasion of the sinus floor as erosions and the other 2 cases were equivocal

    regarding the discrimination of the paranasal sinus secretions from tumor

    tissue infiltration. The remaining 2 cases were only discriminated as

    sphenoid sinus involvement in the enhanced MRI series. The maxillary sinus

    involvement could be seen equally in CT and MRI in one case. In

    agreement, Chong VF et al (1998) and Ng SH et al (1997) stated that

    enhanced MRI excels on CT in detection and discrimination of involvement

  • Discussion

    74

    of NPC in the sphenoid and ethmoidal sinuses from inflammatory paranasal

    secretions, and that both CT and MRI are equal in the detection and

    discrimination of the tumor involvement of the maxillary sinus from

    inflammatory secretions. (53, 80)

    The significance of pterygopalatine fossa (PPF) involvement by

    NPC is that once the tumor gains access to the pterygopalatine fossa, it gains

    a route of spread to the orbit, infratemporal fossa, nasal cavity, and middle

    cranial fossa. Earliest indication of tumor infiltration of the pterygopalatine

    fossa is the replacement of the normal fat content. Widening of the fossa and

    erosion of the bony margins are late signs. As expected, bony abnormality is

    best seen on CT. However, direct visualization of tumor or replacement of

    fat is more elegantly demonstrated on T1-weighted MRI. (103)

    In our study, Pterygopalatine fossa invasion was demonstrated in 9

    cases by MRI and by CT in 7 cases only and 2 cases were seen by MRI only

    as obliteration of fossa fat signal by the tumor. This agrees with Tomura N.

    et al (1999) where CT didnt depict the abnormalities in the pterygopalatine

    fossa in five patients (17%) of a total of 30 patients with pterygopalatine

    fossa involvement, while unenhanced T1w MR images depicted the tumoral

    invasion in all patients. (104)

    In agreement, also Chong VF et al (1995 and

    1997) stated that direct visualization of fat replacement by the tumor is

    better seen on T1w images. (103, 105)

    Detection of skull base bone involvement is based on either direct

    visualization of tumor infiltration or detection of the reaction of bone to the

    malignant process. MRI can identify early involvement of bone marrow. CT,

    which depends mainly on bone destruction, provides detailed bone

  • Discussion

    75

    morphology. Both cortical and trabecular bone components are well defined

    by CT. Based on the balance between the osteoclastic and osteoblastic

    processes, the radiologic appearance of a bone involvement may be lytic,

    sclerotic (blastic), or mixed. (106)

    Rapidly growing aggressive metastases tend

    to be lytic, whereas sclerosis is considered to indicate a slower tumor growth

    rate. Sclerosis may also be a sign of repair after treatment. (107-109)

    CT is not

    sensitive for assessment of malignant marrow infiltration. (106, 110)

    In NPC with skull base invasion, CT can directly determine the extent

    of cortical bone destruction and/or remodeling by cancer. (82, 85, 86, 111, 112)

    On

    the other hand, MRI can show tumor involvement of the skull base as a

    lesion with different signal intensities encroaching on the signal-void bone

    cortex or replacing the marrow. (82, 85)

    Contrast-enhanced fat-suppressed MRI

    provides a better delineation of tumor extension into the clivus and allows

    discrimination of tumor invasion from edema of the marrow. (113)

    The clivus,

    pterygoid bones, body of the sphenoid and apices of the petrous temporal

    bones are most commonly invaded. (114)

    In our study skull base bony involvement was seen in 18 cases (90%)

    by CT and in 17 cases (85%) by MRI. In 53 patients with NPC studied by

    Olmi P et al, CT showed skull base erosion in 12 patients and MRI in 8. (112)

    Ng SH et al (1997) stated that skull base destruction was revealed in 27 of

    67 cases (40.3 %) on CT and in 40 cases (59.7 %) on MRI, and that there

    was no case in which the skull-base invasion was not visible on MRI, while

    there were 13 cases (19.4 %) in which it was detected only by MRI. (80)

    CT demonstrated lytic bone invasion in 7 cases, while 8 cases were

    seen by MRI. MRI, mainly the enhanced T1w fat suppressed images,

  • Discussion

    76

    excelled over CT and demonstrated 1 case of lytic infiltration of the skull

    base bone that couldnt be seen by CT. In contrast, CT better demonstrated

    sclerotic bone involvement in 5 cases but 3 cases were seen only by MRI.

    We acknowledge that bone window CT excels over MRI in visualizing tiny

    bony erosions by the tumor without involvement of the bone marrow. In

    mixed sclerotic and erosive invasion of the bone, both CT and MRI

    demonstrated 6 cases as well. To be noted that bone sclerosis alone, can be

    either bone infiltration by osteoblastic tumor, or remodeling activity due to

    nearby tumor.

    The skull base foramina and fissures which include the foramen

    rotundum (V2 nerve), the vidian canal (vidian nerve), the foramen ovale (V3

    nerve), and foramen lacerum should be examined. The foramen ovale and

    lacerum are common routes of tumor extension into the intracranial cavity.

    (114) Lederman M (1961) stated that foramen lacerum is the most frequently

    invaded foramen due to its close proximity to the lateral pharyngeal recess.

    (115) While the skull base foramina present an unobstructed route for tumor

    spread, direct invasion of the bone bordering these foramina is also a

    common finding. The skull base foramina are best assessed on coronal

    images. Less common findings include inferior spread of tumor to involve

    the hypoglossal nerve canal (XII nerve) and jugular foramen (IX-XI nerves).

    (114)

    Nerves are resistant to tumor, and perineural tumor spread (PNS) is

    an insidious and often asymptomatic process by which NPC can invade

    upward and backward through the skull base to the cavernous sinus and

    middle cranial fossa and invade CN II to VI (upper CN palsy). Cranial

    nerves involvement indicates a tumor T-stage of T4. It may also involve the

  • Discussion

    77

    carotid space, where it may compress or invade CN XII as it exits through

    the hypoglossal canal, CN IX to XI as they emerge from the jugular

    foramen. (116, 117)

    CN involvement on MRI is seen when there is asymmetric

    enlargement, asymmetric enhancement on gadolinium-enhanced T1-

    weighted images, obliteration of perineural fat planes, denervation changes

    in end organs supplied by the nerves, and at last widening of foramina and

    foraminal wall affection. (118, 119)

    Skip lesions may also be noted. (120)

    Widening of a foramen or fissure that an involved nerve normally

    traverses is an indirect sign of perineural spread, and this is best appreciated

    on CT scan using bone algorithm. Features such as obliteration of juxta-

    foraminal fat pads and fat planes along the path of a CN are seen well on

    both modalities. Expansion of the cavernous sinus and soft tissue

    enhancement of Meckels cave, which is normally fluid filled, are other

    indicators of PNS on CT and MRI. (118)

    In our study, skull base foraminal involvement was seen by bone

    algorithm CT and the enhanced MR T1w fat saturated images as well in 17

    cases (85%). The foramen ovale was involved in 8 cases (40%), foramen

    lacerum in 9 cases (45%), jugular foramen in 1 case (5%), sphenopalatine

    foramen in 1 case (5%), and foramen rotundum in 1 case (5%). Of these 17

    cases, only 11 (55%) cases showed perineural spread as demonstrated by

    MRI, and 3 cases of perineural spread were missed by CT. Perineural spread

    was seen along the mandibular nerve (CN V3) in 6 cases by MRI and of

    these cases, one case was missed by CT. Maxillary (CN V2) was involved in

    3 cases by MRI, one of them was missed by CT. The vidian nerve

  • Discussion

    78

    involvement was demonstrated in 4 cases by MRI, while CT missed one

    case. The facial nerve (CN VII) was involved in 1 case by MRI and was

    missed in CT. Thus we declare that CT and MRI, especially the 3D gradient-

    echo T1w post-contrast series, were as well in depicting the involvement of

    the bony foramina, and MRI excelled over CT in detecting perineural

    spread. In agreement, King AD et al (1999) acknowledged that perineural

    spread would be underestimated unless enhanced fat saturated images are

    obtained. (121)

    Maxillary and mandibular nerve involvement is best seen on

    coronal T1-weighted contrast-enhanced MRI with fat saturation. (118)

    MRI is

    generally more sensitive than CT in detecting all features of perineural

    spread except for enlargement and destruction of bony foraminal boundaries.

    Also Caldemeyer KS et al (1998), Liao XB et al (2008); Ng SH et al (2009),

    Sakata K et al (1999) agreed in that MRI was superior in identifying

    perineural spread. (122-125)

    Intracranial extension occurs either from direct extension or through

    skull base foramina (direct invasion, or perineural spread). Intracranial

    extension denotes T4, stage IV tumors. Features denoting intracranial

    extension include cavernous sinus involvement, meningeal involvement

    (especially if seen as nodular enhancing masses), and less frequently masses

    within the middle and/or posterior cranial fossa, respectively according to

    the frequency of involvement. (69, 79)

    Anterior cranial fossa invasion by NPC

    is rarely seen. To be noted that posterior cranial fossa is seen more readily

    with MRI due to its pluri-directional scanning and does not show beam-

    hardening artifact from the dense bone of the skull base. (80)

    In our study, intracranial extension was detected by CT in 7 cases

    (35%) and by MRI in 8 cases (40%). MRI excelled over CT in detection of

  • Discussion

    79

    intracranial extension in one case, where minor meningeal involvement was

    seen in contrast enhanced T1w MR images. All of the studies we could

    reach highlighted the superiority of MRI over CT, revealing intracranial

    invasion, especially in conjunction with the contrast-enhanced fat-

    suppression technique. (69, 78, 82, 85, 113, 123, 126-129)

    Nasopharyngeal carcinoma tumor may infiltrate submucosally

    inferiorly to involve the oropharynx or even the hypopharynx. On imaging,

    oropharyngeal extension is readily noted on coronal or sagittal MR

    imaging as tumor that has extended inferiorly past the plane of palate. On

    axial sections, the oropharynx is considered involved when tumor is seen

    inferior to the C1/C2 junction. (66, 120)

    In our study, MRI excelled over CT in detection of oropharyngeal

    infiltration by NPC. Oropharyngeal carcinoma involvement was seen in 7

    cases (35%) by CT. By MRI, 2 cases of these 7 cases seen by CT were

    interpreted to be retropharyngeal lymph nodal involvement rather than true

    oropharyngeal tumoral involvement. MRI detected 7 cases (35%) of

    oropharyngeal infiltration by the nasopharyngeal carcinoma. Soft palate was

    involved in 2 case, 3 cases showed creeping on the posterior and lateral

    walls of the naso- and oro-pharynx, and 2 cases showed palatine tonsils and

    pillars involvement. Retropharyngeal lymph nodal involvement in the

    retropharyngeal space can be misinterpreted by CT as oropharyngeal

    posterior and/or lateral walls involvement as described formerly. In

    agreement, Ng SH (1997) in a study, Of 18 patients, in whom CT suggested

    oropharyngeal involvement, seven actually had retropharyngeal adenopathy

    disclosed by MRI, with subsequent down-staging of the cases. (80)

    Although

    MR was better in the assessment of the oropharynx, the exam can be non-

  • Discussion

    80

    diagnostic secondary to excessive swallowing artifact, which is not

    uncommon if the patient has pooling of saliva and a large tumor. (130)

    On the

    other side, MRI avoids CTs dental amalgam artifact. (131)

    Lymphadenopathy has very important prognostic implications. Up to

    60-90% of NPC patients will have nodal metastases at presentation

    (Glastonbury, 2007; Goh and Lim, 2009). (69, 132)

    Positive neck nodal disease

    in NPC is associated with an increased risk of local recurrence and distant

    metastases (Goh and Lim, 2009). (69)

    The presence of a single nodal

    metastasis reduces the patients survival rate by 50%. Bilateral

    lymphadenopathy further reduces the survival rate by another 50%. Patients

    with nodes showing necrosis and extra-nodal spread and fixation have a very

    poor prognosis with a further 50% decreased 5-years survival rate. (130, 133, 134)

    NPC generally follows a very orderly pathway of nodal spread,

    beginning with the lateral retropharyngeal lymph nodes (RPLN) - located

    medial to the carotid artery - before involving the nodal groups along the

    internal jugular chain (levels II to IV), spinal accessory chain (Va and Vb),

    as well as supraclavicular nodes (Glastonbury C, 2007; King AD et al,

    2004). (132, 135)

    Nodal disease in the submandibular and parotid/periparotid

    regions is a rare occurrence (Chong & Fan, 2000; King & Bhatia, 2010) (114,

    136) Although the RPLN are generally considered the first echelon of

    metastatic spread, studies have shown that this is not true in all cases and

    that RPLN may be bypassed allowing direct spread to level IIa and IIb

    nodes, which are the most common site for non-retropharyngeal nodal

    involvement (Liu et al, 2006; Mao et al, 2008; Ng et al, 2004; Wang et al,

    2009; King et al, 2000 and 2004). (135, 137-141)

    As medial retropharyngeal

    nodes are usually not visible, any medial retropharyngeal nodes detected on

  • Discussion

    81

    MRI are highly suspicious of metastatic involvement (Wang et al, 2009).

    (142) In addition, Ng SH et al also reported skip metastases in the lower neck

    lymph nodes and the supraclavicular fossa, and distant metastases to thoracic

    and abdominal nodes. (137)

    Several criteria are used in the evaluation of lymph nodes. Size is the

    most commonly used. The measurements are taken using the shortest trans-

    axial diameter and are considered suspicious when the shortest axis is >5

    mm for RPLN, >1.5 cm for levels I and II, and >1 cm for levels IV-VII (Goh

    and Lim, 2009; King and Bhatia, 2010). (69, 114)

    A cluster of 3 or more lymph nodes borderline in size, rounded nodes

    with loss of the fatty hilum, and necrosis are also suggestive of metastatic

    disease (King and Bhatia, 2010). (114)

    However, nodes may still be of normal

    size and harbor malignant cells. The ratio of the longest longitudinal to axial

    dimensions has also been proposed; if the ratio is less than 2, this

    suggests metastatic carcinoma. Normal nodes should have a ratio greater

    than 2. (69)

    If identified, necrosis is considered 100% specific. However, due to

    resolution restrictions, necrosis can only be reliably identified in tumor foci

    greater than 3 mm, of which approximately one-third reportedly have nodal

    necrosis (Goh and Lim, 2009; Som and Brandwein, 2003; Yousem et al,

    1992). (69, 143, 144)

    Necrosis or cystic change is hypo-intense on T1-weighted

    images with rim enhancement after contrast injection, and hyper-intense on

    T2-weighted images. In CT images, necrosis is seen as a focal area of hypo-

    attenuation with or without post-contrast rim enhancement. (110)