Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines
®
)
Melanoma: Uveal
Version 2.2021 — June 25, 2021
Continue
NCCN.org
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
NCCN Guidelines Panel Disclosures
Continue
ϖ Dermatology
‡ Hematology/
Hematology oncology
Þ Internal medicine
† Medical oncology
۞ Ophthalmology
≠ Pathology
¥ Patient advocacy
§ Radiotherapy/
Radiation oncology
¶ Surgery/Surgical
oncology
* Discussion Section
Writing Committee
NCCN
Nicole McMillian, MS
Mai Nguyen, PhD
Brian Gastman, MD ¶
Case Comprehensive Cancer Center/
University Hospitals Seidman Cancer
Center and Cleveland Clinic Taussig
Cancer Institute
Kenneth Grossmann, MD, PhD †
Huntsman Cancer Institute
at the University of Utah
Samantha Guild ¥
AIM at Melanoma
Ashley Holder, MD ¶
O'Neal Comprehensive
Cancer Center at UAB
Douglas Johnson, MD, MSCI †
Vanderbilt-Ingram Cancer Center
Richard W. Joseph, MD ‡ †
Mayo Clinic Cancer Center
Giorgos Karakousis, MD ¶
Abramson Cancer Center at the
University of Pennsylvania
Kari Kendra, MD, PhD †
The Ohio State University Comprehensive
Cancer Center - James Cancer Hospital
and Solove Research Institute
Julie R. Lange, MD, ScM ¶
The Sidney Kimmel Comprehensive
Cancer Center at Johns Hopkins
Ryan Lanning, MD, PhD §
University of Colorado Cancer Center
Kim Margolin, MD †
City of Hope National Medical Center
*Susan M. Swetter, MD/Chair ϖ
Stanford Cancer Institute
*John A. Thompson, MD ‡ †/Vice-Chair
Fred Hutchinson Cancer Research Center/
Seattle Cancer Care Alliance
Mark R. Albertini, MD †
University of Wisconsin
Carbone Cancer Center
Christopher A. Barker, MD §
Memorial Sloan Kettering Cancer Center
Joel Baumgartner, MD ¶
UC San Diego Moores Cancer Center
Genevieve Boland, MD, PhD ¶
Massachusetts General Hospital
Cancer Center
Bartosz Chmielowski, MD, PhD ‡ †
UCLA Jonsson Comprehensive
Cancer Center
Dominick DiMaio, MD ≠
Fred & Pamela Buffett Cancer Center
Alison Durham, MD ϖ
University of Michigan
Rogel Cancer Center
Ryan C. Fields, MD ¶
Siteman Cancer Center at Barnes-
Jewish Hospital and Washington
University School of Medicine
Martin D. Fleming, MD ¶
The University of Tennessee
Health Science Center
Anjela Galan, MD ≠
Yale Cancer Center/
Smilow Cancer Hospital
Miguel Materin, MD ۞
Duke Cancer Institute
Anthony J. Olszanski, MD, RPh †
Fox Chase Cancer Center
Patrick A. Ott, MD, PhD † ‡ Þ
Dana-Farber/Brigham and Women's
Cancer Center
*P. Kumar Rao, MD ۞
Siteman Cancer Center at Barnes-
Jewish Hospital and Washington
University School of Medicine
Ramesh Rengan, MD, PhD §
Fred Hutchinson Cancer Research Center/
Seattle Cancer Care Alliance
Merrick I. Ross, MD ¶
The University of Texas
MD Anderson Cancer Center
April K. Salama, MD †
Duke Cancer Institute
Rohit Sharma, MD ¶
UT Southwestern Simmons
Comprehensive Cancer Center
Joseph Skitzki, MD ¶
Roswell Park Cancer Institute
Jeffrey Sosman, MD ‡
Robert H. Lurie Comprehensive Cancer
Center of Northwestern University
Katy Tsai, MD †
UCSF Helen Diller Family Comprehensive
Cancer Center
Evan Wuthrick, MD §
Moffitt Cancer Center
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
Continue
NCCN Guidelines Panel Disclosures
*P. Kumar Rao, MD/Lead ۞
Siteman Cancer Center at Barnes-Jewish
Hospital and Washington University School of
Medicine
Christopher A. Barker, MD §
Memorial Sloan Kettering Cancer Center
Samantha Guild ¥
AIM at Melanoma
Richard Joseph, MD ‡ †
Mayo Clinic Cancer Center
Miguel Materin, MD ۞
Duke Cancer Institute
Ramesh Rengan, MD, PhD §
Fred Hutchinson Cancer Research Center/
Seattle Cancer Care Alliance
Jeffrey Sosman, MD ‡
Robert H. Lurie Comprehensive Cancer
Center of Northwestern University
*Susan Swetter, MD ϖ
Stanford Cancer Institute
*John A. Thompson, MD ‡ †
Fred Hutchinson Cancer Research
Center/Seattle Cancer Care Alliance
Evan Wuthrick, MD §
Moffitt Cancer Center
ϖ Dermatology
‡ Hematology/Hematology oncology
† Medical oncology
۞ Ophthalmology
¥ Patient advocacy
§ Radiotherapy/Radiation oncology
* Discussion Section Writing Committee
UVEAL MELANOMA SUBCOMMITTEE
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
NCCN Uveal Melanoma Panel Members
NCCN Uveal Melanoma Subcommittee Members
Summary of the Guidelines Updates
Clinical Presentation, Workup and Diagnosis, Clinical Staging (UM-1)
Workup and Staging, Tumor Size, Primary Treatment (UM-2)
Additional Primary Treatment (UM-3)
Systemic Imaging Based on Risk Stratication (UM-4)
Treatment for Recurrence (UM-5)
Treatment of Metastatic Disease (UM-6)
Risk Factors for Development of Uveal Melanoma (UM-A)
Principles of Radiation Therapy (UM-B)
Systemic Therapy for Distant Metastatic Disease (UM-C)
Staging (ST-1)
Clinical Trials: NCCN believes that
the best management for any patient
with cancer is in a clinical trial.
Participation in clinical trials is
especially encouraged.
Find an NCCN Member Institution:
https://www.nccn.org/home/member-
institutions.
NCCN Categories of Evidence and
Consensus: All recommendations
are category 2A unless otherwise
indicated.
See NCCN Categories of Evidence
and Consensus.
NCCN Categories of Preference:
All recommendations are considered
appropriate.
See NCCN Categories of Preference.
The NCCN Guidelines
®
are a statement of evidence and consensus of the authors regarding their views of currently accepted approaches to
treatment. Any clinician seeking to apply or consult the NCCN Guidelines is expected to use independent medical judgment in the context of individual
clinical circumstances to determine any patient’s care or treatment. The National Comprehensive Cancer Network
®
(NCCN
®
) makes no representations
or warranties of any kind regarding their content, use or application and disclaims any responsibility for their application or use in any way. The NCCN
Guidelines are copyrighted by National Comprehensive Cancer Network
®
. All rights reserved. The NCCN Guidelines and the illustrations herein may not
be reproduced in any form without the express written permission of NCCN. ©2021.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
UPDATES
UM-1
• Workup and Diagnosis, First bullet:
First arrow sub-bullet revised: "H&P, including personal/family
history of prior or current cancers (outside the eye)" and footnote
b added.
Third arrow sub-bullet revised: Ocular ultrasound of eye and orbit
.
• Footnote d revised: "...Biopsy of the primary tumor does not impact
outcome, but may provide prognostic information that can help
inform frequency of follow-up and may be needed for eligibility
for clinical trials. If biopsy is performed, molecular/chromosomal
testing for prognostication (chromosome analysis or gene
expression proling [GEP]) is preferred over cytology alone..."
(Also for UM-2A)
UM-2
• Tumor size: The largest diameter was revised as follows:
Largest diameter 5–18
5–19 mm and thickness <2.5 mm
Largest diameter ≤18
≤19 mm and thickness 2.5–10 mm
Largest diameter >18
>19 mm [any thickness] or Thickness >10
mm...
UM-2A
• Footnote u is new: Pathologic evaluation should follow the uveal
melanoma synoptic report recommendations by the College of
American Pathologists. Available at: https://documents.cap.org/
protocols/cp-uveal-melanoma-17protocol-4000.pdf
• Footnote p revised: "...The largest commercially available
brachytherapy plaque is 22
23 mm in diameter; thus, plaque
brachytherapy is recommended only for tumors with largest basal
diameter ≤18
≤19 mm.
UM-3
• Footnote v regarding "Extraocular extension at the time of
enucleation" is new: This is a relatively rare occurrence; data are
limited for these recommendations.
UM-4
• First column recommendation revised: Systemic imaging ± blood
tests based on risk stratication by genetic testing ± tumor size and
histology (at presentation).
• Risk of Distant Metastasis: Under High risk, the following were
removed:
Extraocular extension
Ciliary body involvement
• Footnote dd revised: "... Additional imaging modalities may include
chest/abdominal/pelvic CT with contrast, or dual energy subtraction
chest x-ray. However, screening should..."
• Footnote ee is new: 8q gain, especially when numerous copies are
found portends greater risk for metastasis.
UM-6
• Treatment of Metastatic Disease:
After "No evidence of disease" revised: Clinical trial, if available
(preferred)
.
After "Residual or progressive disease" the arrow was redrawn for
clarity.
Continued
Updates in Version 1.2021 of the NCCN Guidelines for Melanoma: Uveal from Version 3.2020 include:
General
• The Guideline name changed from Uveal Melanoma to Melanoma: Uveal.
Updates in Version 2.2021 of the NCCN Guidelines for Melanoma: Uveal from Version 1.2021 include:
MS-1
The Discussion has been updated to reect the changes in the algorithm.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
UM-A Risk Factors for Development of Uveal Melanoma
• First bullet:
Third arrow sub-bullet revised: Familial uveal melanoma (eg,
germline mutations in BAP1 mutation,
PALB-2, MBD4, or NF-1
(neurobromatosis) [NF-1], dysplastic nevus
syndrome [BK-mole])
Three new arrow sub-bullets added:
Higher numbers of atypical cutaneous nevi, common cutaneous
nevi, and/or cutaneous freckles
Light skin color, propensity to sunburn, and/or light eye (iris)
color
Strong personal or family history of cancer
• Footnote b is new:
Evaluate for evidence of hereditary syndrome and refer for genetic
counseling and testing if indicated:
Early age of diagnosis (<30 years of age)
History of other primary cancers in the patient
Family or personal history of other cancers known to be
associated with a hereditary syndrome:
BAP1: RCC, mesothelioma, cutaneous melanoma,
cholangiocarcinoma, meningioma
BRCA, PALB2: breast, ovarian, or pancreatic cancers
• New references were added.
UM-B Principles of Radiation Therapy
1 of 3
• Treatment Information
Second arrow sub-bullet revised: Plaque brachytherapy is
appropriate for patients with tumors ≤18
≤19 mm in largest base
diameter, ≤10 mm in thickness
Fifth arrow sub-bullet revised: "...MRI or CT may be used for
preoperative planning."
Sixth arrow sub-bullet revised: Round or custom plaques are most
commonly used,
. although non-round plaques (eg, notched) can
be considered for tumors in specic locations (eg, peripapillary).
Custom plaques, such as notched plaques, are commonly used for
tumors in specic locations (peripapillary).
Treatment Dosing Information, rst arrow sub-bullet revised: "...The
largest commercially available brachytherapy plaque is 22
23 mm
in diameter; thus, plaque brachytherapy is recommended only for
tumors with largest basal diameter ≤18
≤19 mm.
2 of 3
Radioembolization, rst bullet revised: Selective internal radiation
therapy for patients with liver metastases using yttrium-90 has been
reported in retrospective studies and in one prospective study.
3 of 3
• References updated.
UM-C Systemic Therapy for Distant Metastatic Disease
• Preferred regimens revised:
Clinical trial
When available and clinically appropriate, enrollment
in a clinical trial is recommended.
• Footnote a revised: When available and clinically appropriate,
enrollment in a clinical trial is recommended. The literature is not
directive regarding the specic systemic agent(s) oering superior
outcomes, but does provide evidence that uveal melanoma is
sensitive to some of the same systemic therapies used to treat
cutaneous melanoma. Although there are no systemic therapies
that have reliably improved the overall survival in patients with
metastatic uveal melanoma, individual patients may derive
substantial
benet on occasion. Given the lack of positive phase III
studies, clinical trials are preferred.
• New references were added for nivolumab/ipilimumab.
UPDATES
Updates in Version 1.2021 of the NCCN Guidelines for Melanoma: Uveal from Version 3.2020 include:
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
• Clinical evaluation, including:
H&P, including personal/family history of prior
or current cancers (outside the eye)
b
Color fundus photography
Ocular ultrasound
Comprehensive eye exam: Examine the front
and back of eye (biomicroscopy)
Dilated fundus exam
(indirect ophthalmoscopy)
Measure visual acuity
Measure and document location and the size
of the tumor (diameter, thickness), distance
from disc and fovea, and ciliary body
involvement
Assess and document if present:
Subretinal uid
– Orange pigment
• Additional testing options include:
Autouorescence of the ocular fundus
Optical coherence tomography
Retinal uorescein angiography of the ocular
fundus
Transillumination
MRI occasionally needed to conrm diagnosis
Consider biopsy if needed to conrm diagnosis
c
or for prognostic analysis for risk stratication
d
UM-1
CLINICAL
PRESENTATION
WORKUP AND DIAGNOSIS CLINICAL STAGING
a
This guideline does not include the management of iris melanoma.
b
See Risk Factors for Development of Uveal Melanoma (UM-A).
c
Biopsy is usually not necessary for initial diagnosis of uveal melanoma and
selection of first-line treatment, but may be useful in cases of uncertainty
regarding diagnosis, such as for amelanotic tumors, or retinal detachment.
d
Biopsy of the primary tumor may provide prognostic information that can help
inform frequency of follow-up and may be needed for eligibility for clinical trials.
If biopsy is performed, molecular/chromosomal testing for prognostication is
preferred over cytology alone. The risks/benefits of biopsy for prognostic analysis
should be carefully considered and discussed.
e
Risk factors for growth of small melanocytic tumors: presence of symptoms,
tumor thickness >2 mm, tumor diameter >5 mm, presence of subretinal fluid and
orange pigment, tumor margin within 3 mm of optic disk, ultrasound hollowness,
absence of halo.
f
The recommendation to "observe and re-evaluate" consists of tests listed under
"Workup and Diagnosis" that would help to clarify if there is progression and
determine the natural history of the indeterminate lesion.
g
Frequency of evaluation should depend on index of suspicion, patient age, and
medical frailty.
• Suspicious pigmented
uveal tumor of ciliary
body and/or choroid
a
Symptoms may
include:
Vision loss
Vision changes
(eg, blurred vision,
photopsia, oaters,
metamorphopsia)
May be asymptomatic
Assessment of risk
factors for developing
uveal melanoma
b
Uveal melanoma
Observe and
re-evaluate for growth or
features of malignancy
f
• Every 2–4 months
g
as
clinically indicated
• Then close follow-up
for 5 years
g
• Then annually
thereafter
See Workup and
Staging for uveal
melanoma (UM-2)
Diagnosis uncertain
and/or <3 risk factors
for growth
e
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-2
WORKUP AND STAGING
PRIMARY TREATMENT
m,n
• Ocular imaging if not previously
done:
If large tumor, close to nerve
or suspicion of extraocular
involvement, MRI of orbit with and
without IV contrast
h
• Assess and document, if present:
Ciliary body involvement
Extraocular extension
• Extraocular imaging:
Baseline imaging to screen for
distant disease
h,i,j
• Consider biopsy of primary tumor
for prognostic analysis
d
Largest diameter 5–19 mm
k
and
thickness <2.5 mm
Largest diameter ≤19 mm
k
and thickness 2.5–10 mm
Largest diameter >19 mm
k
[any thickness]
or Thickness >10 mm [any diameter]
or Thickness >8 mm with optic nerve
involvement [any diameter]
Metastasis
l
• Options:
Brachytherapy plaque
o,p,q
Particle beam radiation
o
• Other options in highly select
patients
r
Options:
• Brachytherapy plaque
o,p,q,s
• Particle beam radiation
o,s
• Enucleation
t,u
Options:
• RT
Particle beam radiation
o,s
Stereotactic radiosurgery (SRS)
o
• Enucleation
t,u
See UM-6
See
Additional
Primary
Treatment
(UM-3)
TUMOR SIZE
See Footnotes on UM-2A
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
d
Biopsy of the primary tumor may provide prognostic information that can help inform frequency of follow-up and may be needed for eligibility for clinical trials. If biopsy
is performed, molecular/chromosomal testing for prognostication is preferred over cytology alone. The risks/benefits of biopsy for prognostic analysis should be
carefully considered and discussed.
h
Unless there is a specific contraindication to the administration of IV contrast (ie, renal impairment or history of a severe allergy), all cross-sectional imaging studies
should be performed with and without IV contrast.
i
Despite lack of treatment options for patients with distant metastatic disease, NCCN favors staging before primary treatment. For small, low-risk tumors, imaging after
primary treatment can be considered.
j
The most frequent sites of metastasis are liver, lungs, skin/soft tissue, and bones. At minimum, all patients should have contrast-enhanced MR or ultrasound of the
liver, with modality preference determined by expertise at the treating institution. Additional imaging modalities may include chest/abdominal/pelvic CT with contrast.
However, screening should limit radiation exposure whenever possible.
k
The cutoff for largest basal diameter depends on the dimensions of the largest brachytherapy plaque available, so may depend on the type of plaque and isotope
selected if brachytherapy is used.
l
Patients may be considered for palliative local therapy to the primary tumor in the setting of metastatic disease. Patients who present with advanced metastatic disease
and limited life expectancy may elect to have no treatment to their primary tumor.
m
An essential feature of high-quality care is that clinical decisions are informed by a variety of case-specific factors (eg, patient characteristics and preferences like age,
status of the other eye among others, disease characteristics, medical history), such that for some patients the best clinical approach may be other than one of the
listed options.
n
For small ciliary body and iris tumors (less than 3 clock hours), surgical excision may be considered.
o
See Principles of Radiation Therapy (UM-B).
p
The plaque should cover the tumor with a ≥2-mm circumferential margin. The exception is for tumors near the optic nerve where it may be impossible to achieve
adequate coverage of the margins. The largest commercially available brachytherapy plaque is 23 mm in diameter; thus, plaque brachytherapy is recommended only
for tumors with largest basal diameter ≤19 mm.
q
Brachytherapy with scleral patch graft for cases with limited extraocular extension.
r
Consider laser ablation or enucleation for patients who are not good candidates for brachytherapy or particle beam radiation.
s
Consider additional treatment with resection, laser ablation, transpupillary thermotherapy, or cryotherapy if concerned that adequate response was not achieved from
initial radiation.
t
While there is a trend toward avoiding enucleation, it is recommended for patients with neovascular glaucoma, tumor replacing >50% of globe, or blind, painful eyes.
Consider enucleation in cases of extensive extraocular extension.
u
Pathologic evaluation should follow the uveal melanoma synoptic report recommendations by the College of American Pathologists.
Available at: https://documents.cap.org/protocols/cp-uveal-melanoma-17protocol-4000.pdf
UM-2A
FOOTNOTES
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-3
o
See Principles of Radiation Therapy (UM-B).
v
This is a relatively rare occurrence; data are limited for these recommendations.
ADDITIONAL PRIMARY TREATMENT
Extraocular
extension
at the time of
enucleation
v
All others
See Follow-up
(UM-4)
Microscopically positive or close
margins after enucleation (but no clinical,
intraoperative, or radiographic evidence of
gross residual disease in the orbit)
Observe
or
Map biopsy
and/or
Consider RT to orbit
(particle beam or photon beam)
o
Visible extraocular tumor or
suspicion of gross disease in the orbit
Biopsy extraocular tissue if possible
and consider one or more of the
following:
• Intraoperative cryotherapy
• Orbital exenteration
• RT to orbit (particle beam or
photon beam)
o
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
RISK OF DISTANT METASTASIS
bb
SYSTEMIC IMAGING BASED ON RISK STRATIFICATION
Low risk:
• Class 1A
cc
• Disomy 3
• Gain of chromosome 6p
• EIF1AX mutation
• T1 (AJCC) (See ST-1 and ST-2)
Medium risk:
• Class 1B
cc
• SF3B1 mutation
• T2 and T3 (AJCC) (See ST-1 and ST-2)
High risk:
• Class 2
cc
• Monosomy 3
• Gain of chromosome 8q
ee
• BAP1 mutation
• PRAME expression
• T4 (AJCC) (See ST-1 and ST-2)
• Imaging to evaluate signs or symptoms
as clinically indicated
• Consider surveillance imaging
dd
every
12 months
• Imaging to evaluate signs or symptoms
• Consider surveillance imaging
dd
every 6–12 months for 10 years,
then as clinically indicated
• Imaging to evaluate signs or symptoms
• Consider surveillance imaging
dd
every 3–6 months for 5 years,
then every 6–12 months for years 6–10,
then as clinically indicated
Recurrence
(See UM-5)
UM-4
Standard follow-
up for aected
eye
w,x,y
and
Systemic imaging
± blood tests
z
based on risk
stratication by
genetic testing
aa
± tumor size
(at presentation)
bb
bb
Risk stratification to determine the frequency of follow-up should be based on the highest risk factor present.
w
The affected eye should be imaged with color fundus photography and
ultrasonography every 3–6 months for 3–5 years, then every 6–12 months
thereafter, if stable. The frequency of follow-up should depend on the size
and location (eg, juxtapapillary location, ciliary body involvement) of the
tumor at presentation. Radiation-related retinopathy and other treatment-
related complications may occur at any time following treatment.
x
The contralateral eye is not at increased risk of uveal melanoma, and can
be followed with routine ophthalmologic care.
y
Additional risk factors for recurrence: Juxtapapillary location and ciliary
body involvement.
z
Liver function tests (LFTs) may be considered as part of follow-up,
although some studies showed poor sensitivity for early detection of liver
metastases.
aa
If biopsy not performed, then follow medium- or high-risk pathways,
depending on whether any high-risk features are present.
cc
Onken MD, Worley LA, Char DH, et al. Collaborative ocular oncology group report
number 1: prospective validation of a multi-gene prognostic assay in uveal melanoma.
Ophthalmology 2012;119:1596-1603.
dd
The most frequent sites of metastasis are liver, lungs, skin/soft tissue, and bones. For
patients who elect to have surveillance imaging, options include: contrast-enhanced
MR or ultrasound of the liver, with modality preference determined by expertise at
the treating institution. Additional imaging modalities may include chest/abdominal/
pelvic CT with contrast, or dual energy subtraction chest x-ray. However, screening
should limit radiation exposure whenever possible. Scans should be performed with IV
contrast unless contraindicated. Recognizing that there are limited options for systemic
recurrence, and that regular imaging may cause patient anxiety, patients should discuss
with their treating physician the benefits of surveillance imaging, and some patients may
elect to forgo surveillance imaging. Participation in a clinical trial is strongly encouraged.
ee
8q gain, especially when numerous copies are found, portends greater risk for
metastasis.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-5
Recurrence
• H&P
• Biopsy if clinically
appropriate
,gg
• Ocular orbital
imaging if not
previously done
• Imaging to evaluate
extent of local
recurrence and/or for
baseline staging
hh
Distant metastatic disease
TREATMENT FOR
RECURRENCE
WORKUP
RT (plaque brachytherapy or particle beam)
o
or
Enucleation
or
Laser ablation
ii
(See UM-6)
Intraocular recurrence
(limited to eye, no orbital
involvement)
Extraocular involvement
Surgical resection
± RT to orbit (particle beam or photon beam)
o
± cryotherapy to orbital tumor
Surgical resection
or Cryotherapy to orbital tumor
and/or
RT to orbit (particle beam or photon beam)
o
Orbital involvement in patients
with prior enucleation
o
See Principles of Radiation Therapy (UM-B).
ff
Extraocular recurrence or metastasis should be confirmed histologically whenever possible or if clinically indicated. Biopsy techniques may include FNA or core.
Obtain tissue for genetic analysis (screening for mutations that may be potential targets for treatment or determine eligibility for a clinical trial) from either biopsy of
the metastasis (preferred) or archival material if the patient is being considered for targeted therapy. Consider broader genomic profiling if the test results might guide
future decisions or eligibility for participation in a clinical trial.
gg
Intraocular recurrence can often be diagnosed and managed without a biopsy. Additional prognostic FNA biopsy may be valuable.
hh
The most frequent sites of metastasis are liver, lungs, skin/soft tissue, and bones. Imaging options include: contrast-enhanced MR or ultrasound of the liver, with
modality preference determined by expertise at the treating institution. Additional imaging may include chest/abdominal/pelvic CT with contrast and/or whole-body FDG
PET/CT; however, screening should limit radiation exposure whenever possible. Brain MRI with IV contrast may be performed if neurologic symptoms are present, but
routine CNS imaging is not recommended. Scans should be performed with IV contrast unless contraindicated.
ii
For small recurrences in patients who cannot undergo RT or surgery, transpupillary thermotherapy is recommended.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-6
Biopsy if clinically
appropriate
• Imaging
hh
for
baseline staging
and to evaluate
specic signs and
symptoms
• Consider LFTs,
including LDH
jj
WORKUP TREATMENT OF METASTATIC DISEASE
Distant
metastatic
disease
No evidence
of disease
Residual or
progressive
disease
Clinical trial,
if available
or
Observation
(See Follow-up
UM-4)
Imaging
hh
to assess
response or
progression
Clinical trial (preferred)
OR
Consider one or more of the following:
• Liver-directed therapies
Regional isolation perfusion of the liver
Embolization (chemotherapy,
radiation,
o
immunotherapy)
Ablative procedures (thermal ablation,
cryotherapy)
Consider resection and/or RT
(photon beam or SRS)
o
for limited or
symptomatic disease in the liver
ll
• Systemic therapies
Systemic therapy
kk
Consider resection and/or RT
(photon beam or SRS)
o
for limited or
symptomatic extrahepatic disease
ll
• Best supportive/palliative care
(See NCCN Guidelines for Palliative
Care)
o
See Principles of Radiation Therapy (UM-B).
ff
Extraocular recurrence or metastasis should be confirmed histologically whenever
possible or if clinically indicated. Biopsy techniques may include FNA or core. Obtain
tissue for genetic analysis (screening for mutations that may be potential targets for
treatment or determine eligibility for a clinical trial) from either biopsy of the metastasis
(preferred) or archival material if the patient is being considered for targeted therapy.
Consider broader genomic profiling if the test results might guide future decisions or
eligibility for participation in a clinical trial.
hh
The most frequent sites of metastasis are liver, lungs, skin/soft tissue, and bones.
Imaging options include: contrast-enhanced MR or ultrasound of the liver, with modality
preference determined by expertise at the treating institution. Additional imaging may
include chest/abdominal/pelvic CT with contrast and/or whole-body FDG PET/CT;
however, screening should limit radiation exposure whenever possible. Brain MRI with IV
contrast may be performed if neurologic symptoms are present, but routine CNS imaging
is not recommended. Scans should be performed with IV contrast unless contraindicated.
jj
LDH is a validated prognostic indicator in cutaneous melanoma.
However, its role in risk stratification of metastatic uveal melanoma is
unknown.
kk
In general, there are no systemic therapies that have reliably
improved the overall survival in patients with metastatic uveal
melanoma; however, individual patients may derive substantial
benefit on occasion. If disease is confined to the liver, regionally
hepatic-directed therapies such as chemoembolization,
radioembolization, or immunoembolization should be considered. See
Systemic Therapy for Metastatic or Unresectable Disease (UM-C).
ll
See Principles of Radiation for Metastatic Disease (ME-H 3 of 7) in
the NCCN Guidelines for Melanoma: Cutaneous.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-A
1 OF 2
• Patients with the following risk factors are at increased risk of developing uveal melanoma:
Choroidal nevi
a
Ocular/oculodermal melanocytosis (hyperpigmentation of episclera, uvea, and skin)
Familial uveal melanoma (eg, germline mutations in BAP1, PALB-2, MBD4, or NF-1 [neurobromatosis])
1-6
Higher numbers of atypical cutaneous nevi, common cutaneous nevi, and/or cutaneous freckles
Light skin color, propensity to sunburn, and/or light eye (iris) color
Strong personal or family history of cancer
b
• The presence of cutaneous melanoma does not increase the risk of uveal melanoma. Among patients with cutaneous melanoma, routine
screening for uveal melanoma is not required.
RISK FACTORS FOR DEVELOPMENT OF UVEAL MELANOMA
a
Risk factors for growth of small melanocytic tumors: presence of symptoms, tumor thickness >2 mm, tumor diameter >5 mm, presence of subretinal fluid and orange
pigment, tumor margin within 3 mm of optic disk, ultrasound hollowness, absence of halo.
b
Evaluate for evidence of hereditary syndrome and refer for genetic counseling and testing if indicated:
Early age of diagnosis (<30 years of age)
• History of other primary cancers in the patient
• Family or personal history of other cancers known to be associated with a hereditary syndrome:
BAP1: RCC, mesothelioma, cutaneous melanoma, cholangiocarcinoma, meningioma
BRCA, PALB2: breast, ovarian, or pancreatic cancers
References
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
1
Walpole S, Pritchard AL, Cebulla CM, et al. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide. J Natl Cancer Inst
2018;110:1328-1341.
2
Abdel-Rahman MH, Pilarski R, Cebulla CM, et al. Germline BAP1 mutation predisposes to uveal melanoma, lung adenocarcinoma, meningioma, and other cancers. J
Med Genet 2011;48:856-859.
3
Pilarski R, Carlo M, Cebulla C, Abdel-Rahman M. BAP1 Tumor Predisposition Syndrome. In: Adam MP, Ardinger HH, Pagon RA, et al. eds. GeneReviews. Seattle, WA:
University of Washington, Seattle. Copyright © 1993-2020, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington,
Seattle. All rights reserved; 2020.
4
Derrien AC, Rodrigues M, Eeckhoutte A, et al. Germline MBD4 mutations and predisposition to uveal melanoma. J Natl Cancer Inst 2021;113:80-87.
5
Abdel-Rahman MH, Sample KM, Pilarski R, et al. Whole exome sequencing identifies candidate genes associated with hereditary predisposition to uveal melanoma.
Ophthalmology 2020;127:668-678.
6
Abdel-Rahman MH, Pilarski R, Ezzat S, et al. Cancer family history characterization in an unselected cohort of 121 patients with uveal melanoma. Fam Cancer
2010;9:431-438.
7
Singh N, Singh R, Bowen RC, et al. Uveal Melanoma in BAP1 Tumor Predisposition Syndrome: Estimation of Risk. Am J Ophthalmol 2020;224:172-177.
RISK FACTORS FOR DEVELOPMENT OF UVEAL MELANOMA
REFERENCES
UM-A
2 OF 2
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-B
1 OF 3
Plaque Brachytherapy
• Treatment Information
Plaque brachytherapy is a common form of denitive radiotherapy for the primary tumor.
1
A prospective trial found no dierence in cause-
specic survival among patients with tumors 2.5–10 mm in apical height (2.5–8 mm if peripapillary) and ≤16 mm in maximum basal diameter
randomized to plaque brachytherapy or enucleation.
2
Plaque brachytherapy is appropriate for patients with tumors ≤19 mm
a
in largest base diameter, ≤10 mm in thickness.
Plaque brachytherapy is appropriate as an upfront therapy after initial diagnosis, or after local recurrence following a prior local therapy.
Plaque brachytherapy should be performed by an experienced multidisciplinary team including an ophthalmic oncologist, radiation
oncologist, and brachytherapy physicist.
3
Tumor localization for brachytherapy may be performed using indirect ophthalmoscopy, transillumination, light pipe diathermy,
and/or ultrasound (intraoperative and/or preoperative).
4
MRI or CT may be used for preoperative planning.
Round or custom plaques are most commonly used. Custom plaques, such as notched plaques, are commonly used for tumors in specic
locations (peripapillary).
• Treatment Dosing Information
Using iodine-125 Collaborative Ocular Melanoma Study (COMS) plaques, 85 Gy should be prescribed to the apex of the tumor at low dose
rate (≥0.6 Gy/h). Dose adjustments may need to be made for non-COMS plaques.
5
The plaque margin on the tumor border should be
≥2 mm when feasible (diameter of plaque ≥4 mm larger than largest base diameter of tumor). The exception is for tumors near the optic
nerve where it may be impossible to achieve adequate coverage of the margins. The largest commercially available brachytherapy plaque
is 23 mm
a
in diameter; thus, plaque brachytherapy is recommended only for tumors with largest basal diameter ≤19 mm.
Using other radioisotopes (eg, ruthenium-106, palladium-103, strontium-90, cobalt-60, cesium-131), or non-COMS iodine-125 plaques,
60–100 Gy may be prescribed at low dose rate to the tumor apex; alternatively, a minimum dose may be prescribed to the base of the tumor.
The plaque margin on the tumor border may vary for other radioisotopes.
Particle Beam Therapy
• Treatment Information
Particle beam therapy is a common form of denitive radiotherapy for the primary tumor.
1
A prospective trial found no dierence in cause-
specic survival among patients with tumors ≤15 mm in maximum basal diameter and ≤11 mm in apical height randomized to plaque
brachytherapy or particle beam therapy.
6
Particle beam therapy is appropriate as upfront therapy after initial diagnosis, after margin-positive enucleation, or for intraocular or orbital
recurrence.
Particle beam therapy should be performed by an experienced multidisciplinary team including an ophthalmic oncologist, radiation
oncologist, and particle beam physicist.
7
Tumor localization for particle beam therapy may be performed using indirect ophthalmoscopy, transillumination, and/or ultrasound
(intraoperative and/or preoperative), MRI, and/or CT.
PRINCIPLES OF RADIATION THERAPY
Continued
References
a
The cutoff for largest basal diameter depends on the dimensions of the largest brachytherapy plaque available, so may depend on the type
of plaque and isotope selected if brachytherapy is used.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
Particle Beam Therapy (continued)
• Treatment Dosing Information
For intraocular tumors:
Using protons, 50–70 cobalt Gray equivalent (CGyE) in 4–5 fractions should be prescribed to encompass the planning target volume
surrounding the tumor.
7-9
Using carbon ions, 60–85 CGyE in 5 fractions should be prescribed to encompass the planning target volume surrounding the tumor.
10
Fiducial markers (tantalum clips) are encouraged to permit eye and tumor position verication for image-guided radiotherapy delivery.
Volumetric planning in 3 dimensions (with or without CT and/or MRI) is encouraged to maximize radiation delivery to tumor and minimize
radiation delivery to organs and tissues at risk of injury from radiation.
Stereotactic Radiosurgery (SRS)
• Treatment Information
SRS is the least often used form of denitive radiotherapy for the treatment of primary or recurrent intraocular tumors.
11,12
Few prospective studies have assessed the ecacy and safety of radiosurgery.
13,14
Tumor localization, ducial marker use, and planning for SRS are generally consistent with particle beam therapy approaches.
• Treatment Dosing Information
Using fractionated SRS: 45–70 Gy in 2–5 fractions should be prescribed.
Using single-fraction SRS: 18–45 Gy in 1 fraction should be prescribed.
Photon Beam Radiotherapy
• Treatment/Dosing Information
Photon beam radiotherapy is a preferred option as an adjuvant to surgery for orbital involvement.
Adjuvant radiotherapy can be used in patients at risk for local recurrence (margin-positive enucleation or exenteration) or regional
recurrence (resected regional metastases).
– Adjuvant RT Dosing
A dose of 20–30 Gy in 5 fractions should be prescribed to the clinical target volume at risk for recurrence
15,16
using intensity-
modulated techniques with image guidance.
Photon beam radiotherapy can be used for treatment of distant metastases at risk for causing symptoms or for palliation of symptomatic
distant metastases.
– RT Dosing for Distant Metastases
Doses of 8–30 Gy in 1–10 fractions should be prescribed to the appropriate target volume
17
using appropriate 3-D or intensity-
modulated radiation therapy (IMRT) techniques with or without image guidance.
Radioembolization
• Selective internal radiation therapy for patients with liver metastases using yttrium-90 has been reported in retrospective studies and in one
prospective study.
18,19
Further study is required to determine the appropriate patients for and risks and benets of this approach.
UM-B
2 OF 3
PRINCIPLES OF RADIATION THERAPY
References
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-B
3 OF 3
1
Abrams MJ, Gagne NL, Melhus CS, Mignano JE. Brachytherapy vs. external beam radiotherapy for choroidal melanoma: Survival and patterns-of-care analyses.
Brachytherapy 2016;15:216-223.
2
Collaborative Ocular Melanoma Study Group. The COMS randomized trial of iodine 125 brachytherapy for choroidal melanoma: V. Twelve-year mortality rates and
prognostic factors: COMS report No. 28. Arch Ophthalmol 2006;124:1684-1693.
3
American Brachytherapy Society - Ophthalmic Oncology Task Force. The American Brachytherapy Society consensus guidelines for plaque brachytherapy of uveal
melanoma and retinoblastoma. Brachytherapy 2014;13:1-14.
4
Almony A, Breit S, Zhao H, et al. Tilting of radioactive plaques after initial accurate placement for treatment of uveal melanoma. Arch Ophthalmol 2008;126:65-70.
5
Rivard MJ, Chiu-Tsao ST, Finger PT, et al. Comparison of dose calculation methods for brachytherapy of intraocular tumors. Med Phys 2011;38:306-316.
6
Mishra KK, Quivey JM, Daftari IK, et al. Long-term results of the UCSF-LBNL randomized trial: Charged particle with helium ion versus iodine-125 plaque therapy for
choroidal and ciliary body melanoma. Int J Radiat Oncol Biol Phys 2015;92:376-383.
7
Hrbacek J, Mishra KK, Kacperek A, et al. Practice patterns analysis of ocular proton therapy centers: The International OPTIC Survey. Int J Radiat Oncol Biol Phys
2016;95:336-343.
8
Hartsell WF, Kapur R, Hartsell SO, et al. Feasibility of proton beam therapy for ocular melanoma using a novel 3D treatment planning technique. Int J Radiat Oncol Biol
Phys 2016;95:353-359.
9
Gragoudas ES, Lane AM, Regan S, et al. A randomized controlled trial of varying radiation doses in the treatment of choroidal melanoma. Arch Ophthalmol
2000;118:773-778.
10
Tsuji H, Ishikawa H, Yanagi T, et al. Carbon-ion radiotherapy for locally advanced or unfavorably located choroidal melanoma: a phase I/II dose-escalation study. Int J
Radiat Oncol Biol Phys 2007;67:857-862.
11
Muller K, Naus N, Nowak PJ, et al. Fractionated stereotactic radiotherapy for uveal melanoma, late clinical results. Radiother Oncol 2012;102:219-224.
12
Dunavoelgyi R, Georg D, Zehetmayer M, et al. Dose-response of critical structures in the posterior eye segment to hypofractioned stereotactic photon radiotherapy of
choroidal melanoma. Radiother Oncol 2013;108:348-353.
13
Muller K, Nowak PJ, de Pan C, et al. Effectiveness of fractionated stereotactic radiotherapy for uveal melanoma. Int J Radiat Oncol Biol Phys 2005;63:116-122.
14
Zehetmayer M, Kitz K, Menapace R, et al. Local tumor control and morbidity after one to three fractions of stereotactic external beam irradiation for uveal melanoma.
Radiother Oncol 2000;55:135-144.
15
The Collaborative Ocular Melanoma Study (COMS) randomized trial of pre-enucleation radiation of large choroidal melanoma III: local complications and observations
following enucleation COMS report no. 11. Am J Ophthalmol 1998;126:362-372.
16
Ang KK, Peters LJ, Weber RS, et al. Postoperative radiotherapy for cutaneous melanoma of the head and neck region. Int J Radiat Oncol Biol Phys 1994;30:795-798.
17
Huguenin PU, Kieser S, Glanzmann C, et al. Radiotherapy for metastatic carcinomas of the kidney or melanomas: an analysis using palliative end points. Int J Radiat
Oncol Biol Phys 1998;41:401-405.
18
Jia Z, Jiang G, Zhu C, et al. A systematic review of yttrium-90 radioembolization for unresectable liver metastases of melanoma. Eur J Radiol 2017;92:111-115.
19
Gonsalves CF, Eschelman DJ, Adamo RD, et al. A prospective phase II trial of radioembolization for treatment of uveal melanoma hepatic metastasis. Radiology
2019;293:223-231.
PRINCIPLES OF RADIATION THERAPY
REFERENCES
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-C
1 OF 3
SYSTEMIC THERAPY FOR DISTANT METASTATIC DISEASE
a
a
The literature is not directive regarding the specific systemic agent(s) offering superior outcomes, but does provide evidence that uveal melanoma is sensitive to some
of the same systemic therapies used to treat cutaneous melanoma. Although there are no systemic therapies that have reliably improved the overall survival in patients
with metastatic uveal melanoma, individual patients may derive benefit on occasion. Given the lack of positive phase III studies, clinical trials are preferred.
b
See NCCN Guidelines for Management of Immunotherapy-Related Toxicities.
c
See Management of Toxicities Associated with Targeted Therapy from the NCCN Guidelines for Melanoma: Cutaneous (ME-K).
d
The listed systemic therapy options do not cover BRAF or KIT mutated tumors. In general, uveal melanomas rarely have BRAF or KIT mutations.
Preferred Regimens
• When available and clinically appropriate,
enrollment in a clinical trial is recommended.
Other Recommended Regimens
a
• Consider one or more of the following options:
Immunotherapy
b
Anti PD-1 monotherapy
– Pembrolizumab
– Nivolumab
Nivolumab/ipilimumab
Ipilimumab
Cytotoxic Regimens
Dacarbazine
Temozolomide
Paclitaxel
Albumin-bound paclitaxel
Carboplatin/paclitaxel
Targeted Therapy
c,d
Trametinib
References
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-C
2 OF 3
Immunotherapy
Pembrolizumab and Nivolumab
• Kottschade LA, McWilliams RR, Markovic SN, et al. The use of pembrolizumab for the treatment of metastatic uveal melanoma. Melanoma Res
2016;26:300-303.
Algazi AP, Tsai KK, Shoushtari AN, et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer
2016;122:3344-3353.
Nivolumab/ipilimumab
Piulats JM, Espinosa E, de la Cruz Merino L, et al. Nivolumab Plus Ipilimumab for Treatment-Naive Metastatic Uveal Melanoma: An Open-Label,
Multicenter, Phase II Trial by the Spanish Multidisciplinary Melanoma Group (GEM-1402). J Clin Oncol 2021;39:586-598.
Pelster MS, Gruschkus SK, Bassett R, et al. Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: Results From a Single-Arm Phase II Study. J
Clin Oncol 2021;39:599-607.
Ipilimumab
Zimmer L, Vaubel J, Mohr P, et al. Phase II DeCOG-study of ipilimumab in pretreated and treatment-naive patients with metastatic uveal melanoma.
PLoS One 2015;10:e0118564.
Danielli R, Ridol R, Chiarion-Sileni V, et al. Ipilimumab in pretreated patients with metastatic uveal melanoma: safety and clinical ecacy. Cancer
Immunol Immunother 2012;61:41-48.
Luke JJ, Callahan MK, Postow MA, et al. Clinical activity of ipilimumab for metastatic uveal melanoma: a retrospective review of the Dana-Farber
Cancer Institute, Massachusetts General Hospital, Memorial Sloan-Kettering Cancer Center, and University Hospital of Lausanne experience. Cancer
2013;119:3687-3695.
SYSTEMIC THERAPY FOR METASTATIC OR UNRESECTABLE DISEASE
REFERENCES
Continued
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
Note: All recommendations are category 2A unless otherwise indicated.
Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.
NCCN Guidelines Index
Table of Contents
Discussion
UM-C
3 OF 3
Cytotoxic Regimens
Dacarbazine
• Serrone L, Zeuli M, Sega FM, et al. Dacarbazine-based chemotherapy for metastatic melanoma: thirty-year experience overview.
J Exp Clin Cancer Res 2000;19:21-34.
Temozolomide
Bedikian AY, Papadopoulos N, Plager C, et al. Phase II evaluation of temozolomide in metastatic choroidal melanoma. Melanoma Res 2003;13:303-
306.
Paclitaxel
Wiernik PH and Einzig AI. Taxol in malignant melanoma. J Natl Cancer Inst Monogr 1993;15:185-187.
Albumin-bound paclitaxel
Hersh EM, O'Day SJ, Ribas A, et al. A phase 2 clinical trial of nab-paclitaxel in previously treated and chemotherapy-naïve patients with metastatic
melanoma. Cancer 2010;116:155-163.
• Kottschade LA, Suman VJ, Amatruda T, et al. A phase II trial of nab-paclitaxel (ABI-007) and carboplatin in patients with unresectable stage iv
melanoma: a north central cancer treatment group study, N057E(1). Cancer 2011;117:1704-1710.
Paclitaxel/carboplatin
• Rao RD, Holtan SG, Ingle JN, et al. Combination of paclitaxel and carboplatin as second-line therapy for patients with metastatic melanoma. Cancer
2006;106:375-382.
Homsi J, Bedikian AY, Papadopoulos NE, et al. Phase 2 open-label study of weekly docosahexaenoic acid-paclitaxel in patients with metastatic uveal
melanoma. Melanoma Res 2010;20:507-510.
Targeted Therapy
Trametinib
• Falchook GS, Lewis KD, Infante JR, et al. Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: a phase 1 dose-escalation
trial. Lancet Oncol 2012;13:782-789.
Shoushtari AN, Kudchadkar RR, Panageas K, et al. A randomized phase 2 study of trametinib with or without GSK2141795 in patients with advanced
uveal melanoma. J Clin Oncol 2016;34:9511-9511.
SYSTEMIC THERAPY FOR METASTATIC OR UNRESECTABLE DISEASE
REFERENCES
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Used with permission of the American College of Surgeons, Chicago, Illinois. The original source for this information is the AJCC Cancer Staging Manual, Eighth Edition
(2017) published by Springer International Publishing.
American Joint Committee on Cancer (AJCC)
Denitions of TNM for Choroidal and Ciliary Melanoma (8th ed., 2017)
Table 1. Denitions for T, N, M
Choroidal and Ciliary Body Melanomas
T Primary Tumor
TX Primary tumor cannot be assessed
T0 No evidence of primary tumor
T1 Tumor size category 1
T1a Tumor size category 1 without ciliary body involvement and extraocular extension
T1b Tumor size category 1 with ciliary body involvement
T1c Tumor size category 1 without ciliary body involvement but with extraocular extension ≤5 mm in largest diameter
T1d Tumor size category 1 with ciliary body involvement and extraocular extension ≤5 mm in largest diameter
T2 Tumor size category 2
T2a Tumor size category 2 without ciliary body involvement and extraocular extension
T2b Tumor size category 2 with ciliary body involvement
T2c Tumor size category 2 without ciliary body involvement but with extraocular extension ≤5 mm in largest diameter
T2d Tumor size category 2 with ciliary body involvement and extraocular extension ≤5 mm in largest diameter
T3 Tumor size category 3
T3a Tumor size category 3 without ciliary body involvement and extraocular extension
T3b Tumor size category 3 with ciliary body involvement
T3c Tumor size category 3 without ciliary body involvement but with extraocular extension ≤5 mm in largest diameter
T3d Tumor size category 3 with ciliary body involvement and extraocular extension ≤5 mm in largest diameter
T4 Tumor size category 4
T4a Tumor size category 4 without ciliary body involvement and extraocular extension
T4b Tumor size category 4 with ciliary body involvement
T4c Tumor size category 4 without ciliary body involvement but with extraocular extension ≤5 mm in largest diameter
T4d Tumor size category 4 with ciliary body involvement and extraocular extension ≤5 mm in largest diameter
T4e Any tumor size category with extraocular extension >5 mm in largest diameter
Notes
1. Primary ciliary body and choroidal melanomas are classied according to the four tumor size categories dened in Figure 1. (See ST-3)
2. In clinical practice, the largest tumor basal diameter may be estimated in optic disc diameters (DD; average: 1 DD = 1.5 mm), and tumor thickness may be
estimated in diopters (average: 2.5 diopters = 1 mm). Ultrasonography and fundus photography are used to provide more accurate measurements.
3. When histopathologic measurements are recorded after xation, tumor diameter and thickness may be underestimated because of tissue shrinkage.
Continued
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
ST-1
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
American Joint Committee on Cancer (AJCC)
Denitions of TNM for Choroidal and Ciliary Melanoma (8th ed., 2017)
Table 1. Denitions for T, N, M (continued)
N Regional Lymph Nodes
NX Regional lymph nodes cannot be assessed
N0 No regional lymph node involvement
N1 Regional lymph node metastases or discrete tumor
deposits in the orbit
N1a Metastasis in one or more regional lymph node(s)
N1b No regional lymph nodes are positive, but there are
discrete tumor deposits in the orbit that are not contiguous
to the eye (choroidal and ciliary body).
M Distant Metastasis
M0
No distant metastasis by clinical classication
M1
Distant metastasis
M1a Largest diameter of the largest metastasis ≤3.0 cm
M1b Largest diameter of the largest metastasis 3.1–8.0 cm
M1c Largest diameter of the largest metastasis ≥8.1 cm
G Histologic Grade
GX
Grade cannot be assessed
G1
Spindle cell melanoma (>90% spindle cells)
G2
Mixed cell melanoma (>10% epithelioid cells and <90% spindle cells)
G3
Epithelioid cell melanoma (>90% epithelioid cells)
Note: Because of the lack of universal agreement regarding which proportion
of epithelioid cells classies a tumor as mixed or epithelioid, some ophthalmic
pathologists currently combine grades 2 and 3 (non-spindle, ie, epithelioid cells
detected) and contrast them with grade 1 (spindle, ie, no epithelioid cells detected).
Used with permission of the American College of Surgeons, Chicago, Illinois. The original source for this information is the AJCC Cancer Staging Manual, Eighth Edition
(2017) published by Springer International Publishing.
Table 2. AJCC Prognostic Stage Groups
T N M
Stage I T1a N0 M0
Stage IIA T1b-d N0 M0
T2a N0 M0
Stage IIB T2b N0 M0
T3a N0 M0
Stage IIIA T2c-d N0 M0
T3b-c N0 M0
T4a N0 M0
Stage IIIB T3d N0 M0
T4b-c N0 M0
Stage IIIC T4d-e N0 M0
Stage IV Any T N1 M0
Any T Any N M1a-c
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
ST-2
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Figure 1: Classication of Ciliary Body and Choroid Uveal Melanoma Based on Thickness and Diameter
Thickness (mm)
>15.0 4 4 4
12.1–15.0 3 3 4 4
9.1–12.0 3 3 3 3 3 4
6.1–9.0 2 2 2 2 3 3 4
3.1–6.0 1 1 1 2 2 3 4
≤3.0 1 1 1 1 2 2 4
≤3.0 3.1–6.0 6.1–9.0 9.1–12.0 12.1–15.0 15.1–18.0 >18.0
Largest basal diameter (mm)
Used with permission of the American College of Surgeons, Chicago, Illinois. The original source for this information is the AJCC Cancer Staging Manual, Eighth Edition
(2017) published by Springer International Publishing.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
ST-3
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
Version 2.2021, 06/25/21 © 2021 National Comprehensive Cancer Network
®
(NCCN
®
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Index
Table of Contents
Discussion
NCCN Categories of Evidence and Consensus
Category 1 Based upon high-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2A Based upon lower-level evidence, there is uniform NCCN consensus that the intervention is appropriate.
Category 2B Based upon lower-level evidence, there is NCCN consensus that the intervention is appropriate.
Category 3 Based upon any level of evidence, there is major NCCN disagreement that the intervention is appropriate.
All recommendations are category 2A unless otherwise indicated.
NCCN Categories of Preference
Preferred intervention
Interventions that are based on superior ecacy, safety, and evidence; and, when appropriate,
aordability.
Other recommended
intervention
Other interventions that may be somewhat less ecacious, more toxic, or based on less mature data;
or signicantly less aordable for similar outcomes.
Useful in certain
circumstances
Other interventions that may be used for selected patient populations (dened with recommendation).
All recommendations are considered appropriate.
CAT-1
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-1
Discussion
Table of Contents
Overview ......................................................................................... MS-2
Staging ........................................................................................ MS-2
Molecular Characteristics ............................................................. MS-2
Risk Factors for Uveal Melanoma ................................................. MS-3
Literature Search Criteria and Guidelines Update Methodology ........ MS-5
Diagnosis and Workup .................................................................... MS-5
Characteristics, Detection, and Differential Diagnosis ................... MS-5
Initial Workup of Suspicious Pigmented Uveal Lesion ................... MS-7
Observation for Uncertain Diagnosis in Patients with Low Risk ...... MS-14
Further Workup Prior to Treatment ................................................ MS-15
Treatment of Localized Primary Uveal Melanoma .......................... MS-15
Surgical Options......................................................................... MS-16
Radiation Therapy ...................................................................... MS-17
Other Ablative Techniques ......................................................... MS-22
Treatment for Extraocular/Extrascleral Extension ....................... MS-23
Treatment of Localized Uveal Melanoma .................................... MS-24
Treatment of Primary Tumor in Patients with Metastatic Disease MS-25
Follow-up ...................................................................................... MS-25
Patterns of Local Recurrence ..................................................... MS-25
Patterns of Treatment Complications .......................................... MS-27
Surveillance Methods for Local Recurrence or Complications ..... MS-30
Follow-up for the Treated Eye .................................................... MS-31
Risk in Contralateral (Fellow) Eye............................................... MS-31
Patterns of Metastases .............................................................. MS-32
Risk Factors for Metastasis ....................................................... MS-32
Surveillance Methods for Distant Metastatic Disease ................. MS-34
Risk of Developing Secondary Cancers During Follow-up .......... MS-35
Follow-up for Distant Metastasis ................................................ MS-35
Management of Recurrence .......................................................... MS-36
Workup for Recurrence ............................................................. MS-36
Treatment for Local Recurrence ................................................ MS-37
Treatment for Metastatic Disease .............................................. MS-37
References ................................................................................... MS-52
This discussion corresponds to the NCCN Guidelines for Melanoma: Uveal. Last updated: June 25
th
, 2021.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-2
Overview
Uveal melanoma is the most common type of primary intraocular
malignancy in adults, with new cases occurring in 5 to 10 people per
million per year.
1-7
Uveal melanomas can arise anywhere in the uveal
tract, with less than 75% arising in the choroid, and the remainder arising
in the iris or ciliary body.
1,3,6,8,9
The distribution of uveal melanoma by sex, race, and geography differs
from that of cutaneous melanoma.
1,3,10
Most uveal melanomas are
localized at first presentation, and only a small percentage of cases
present with metastases (<3%).
6,10-14
Risk of metastasis varies by stage at
presentation, with 5-year risk of metastasis ranging from 3% to 5% for
stage I to 44% or greater for stage III.
8,13
Large population-based analyses
have reported disease-specific survival (for uveal melanoma) between
70% to 81% at 5 years,
2,5,6,10,15-17
and this rate has remained stable over
time.
5
Disease-specific survival varies by the extent of disease at
presentation.
12,13,15,18
For those with early-stage uveal melanoma (stage I
II), 5-year melanoma-specific survival is 85% or better.
13,15,18
For those
with distant metastatic disease, most studies report estimated 5-year
survival of less than 20%,
13,15,19-22
which has not improved over the past
few decades.
23
These NCCN Guidelines include recommendations for management of
melanomas arising in the choroid or ciliary body. Recommendations for iris
melanomas are not included in these guidelines, because iris melanomas
are rare (3%–5% of uveal melanomas),
3,8,9,24,25
have a low rate of systemic
metastasis (~5% at 5 years compared with 15%–20% for ciliary body or
choroidal melanomas),
26
8,27-30
have a better prognosis than other types of
uveal melanoma,
25,30
have a different AJCC staging system,
31
and have a
different molecular signature.
32-36
Treatments for iris melanoma may differ
from other types of uveal melanoma due to these factors, as well as
anatomic considerations, ease of resection,
37-40
and the negative effects of
radiation to the iris.
41-46
Moreover, patients with iris melanomas were
excluded from many of the large randomized trials that inform treatment
recommendations for uveal melanomas.
14,47,48
Staging
The patterns of presentation and prognosis for uveal melanoma are
completely different from cutaneous melanoma, and the AJCC Staging
Manual, 8
th
Edition, includes separate staging systems for cutaneous,
uveal, and conjunctival melanoma.
31
The staging system for uveal
melanoma is further subdivided into separate T-staging for iris versus
choroidal or ciliary body melanoma.
31
Each of these staging systems is
empirically based on survival data from large epidemiologic studies, albeit
with more data independently validating the staging system for melanoma
of the choroid and ciliary body
12,13,18
compared with the iris
29
or
conjunctiva.
49,50
Molecular Characteristics
Cutaneous, uveal, and conjunctival melanomas also have different
molecular signatures.
34,51-61
Whereas BRAF, NRAS, KIT, and TERT
promoter mutations are extremely rare in uveal melanoma,
34,51-54,60,62,63
they are more common in conjunctival and cutaneous melanomas.
55-61
Most notably, BRAF mutations occur in 20% to 50% of conjunctival
melanomas, suggesting that conjunctival melanoma may be more similar
to cutaneous melanoma than to uveal melanoma.
55-58,60
Molecular markers
common in uveal melanomas (and may have prognostic significance) are
not often found in conjunctival or cutaneous melanoma. These include
chromosomal abnormalities (particularly chromosomes 3 and 8),
12,64-68
and
mutations in GNAQ or GNA11 (>80% of uveal cases),
34,51,66,69,70
BAP1,
71,72
SF3B1, and EIFAX.
36,73,74
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-3
Risk Factors for Uveal Melanoma
Studies of large populations of patients with and without uveal melanoma
have identified a number of risk factors for the development of uveal
melanoma.
Choroidal Nevi
Large population-based studies have found that choroidal nevi occur in
1.9% to 6.5% of the population, depending on the population studied.
75-77
Choroidal nevi are more common in whites (4.6%7.9% in the United
States) than in Hispanics or blacks.
75
Due to differences in prevalence
across races, it is not surprising that the reported rate also depends on
geography. Several large studies have shown that there is no association
between choroidal nevi and cutaneous melanoma.
75
Choroidal nevi can
transform into choroidal melanoma,
78,79
and one study reported that of
uveal melanomas diagnosed, 8% arose from a previously documented
nevus.
78
Others have argued that the fraction of uveal melanomas that
arise from nevi may in fact be much higher, as many patients diagnosed
with uveal melanoma have not had an ophthalmologic exam for many
years.
80
The rate of transformation from nevi to uveal melanoma is an issue of
much debate. Large population studies comparing the prevalence of
choroidal melanoma with the prevalence of choroidal nevi have estimated
the annual rate of malignant transformation of a choroidal nevus to range
from 1/4300 to 1/8845.
76,81,82
Many studies have aimed to more directly
determine the rate at which choroidal nevi transform into uveal melanoma
by evaluating changes in presumed nevi or indeterminate melanocytic
lesions over time.
83-87
Choroidal nevi can be difficult to distinguish from
choroidal melanoma, however, and there is much debate about how to
distinguish choroidal nevi from melanoma, as there is evidence from
multiple studies that many small lesions presumed to be nevi based on
size may actually be melanoma (See Diagnosis and Workup section).
88,89
Because the criteria for diagnosis of choroidal melanoma differed across
studies, and diagnosis was rarely confirmed by histology, there is concern
that in many studies some fraction of the population diagnosed with uveal
melanoma was in fact misdiagnosed. Given the debate about differential
diagnosis, the actual rate of transformation is unclear. Due to uncertainty
regarding uveal melanoma diagnosis, other analyses have looked at the
likelihood of lesion growth in patients with untreated melanocytic choroidal
lesions.
90,83,84,91-97
Growth has been correlated with risk of metastasis,
91
even though some growing choroidal nevi do not undergo malignant
transformation.
98,99
These studies have found that 13% to 36% of lesions
grew by 5 years.
83,90,94,95
The wide variability in rates across studies is
likely due to differences in the population selected for study and the
retrospective nature of data collection in some of these studies. More
importantly, these studies have also identified factors predictive of growth
(Table 1). Several analyses used tumor thickness greater than 2 mm,
tumor diameter greater than 5 mm, and tumor margin within 3 mm of the
optic disc as cutoffs for these risk factors.
94,96
Some of these
characteristics have also been associated with increased risk of tumor
metastasis: proximity to optic disc, documented growth, and tumor
thickness.
91
It is important to note that for patients with small lesions presumed to be
nevi or indeterminate, who are observed and treated upon evidence of
transformation (eg, growth, development of orange pigment, subretinal
fluid, other symptoms), the risk of metastasis is low.
91,95
The risk of death
from ocular melanoma in this situation is also low (1/2341 patients per
year),
84
but these risks increase with increasing baseline tumor size.
84
Patient characteristics that increase the likelihood of choroidal nevi growth
include Birt-Hogg-Dubé syndrome, myotonic dystrophy, and
immunocompromise. Cases of patients with uveal melanoma and
myotonic dystrophy have been reported,
100,101
and a retrospective study
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-4
found an increased risk of choroidal melanoma in patients with myotonic
dystrophy (relative to the general population).
102
Cases of choroidal
melanoma have been reported in patients with Birt-Hogg-Dubé
syndrome.
103,104
Cases of ocular melanoma have been reported in
immunocompromised patients.
105,106
Ocular/Oculodermal Melanocytosis
The rate of ocular/oculodermal melanocytosis, which causes
hyperpigmentation of the episclera, uvea, and skin, is much higher in
patients with uveal melanoma than in the general population.
107-112
Familial Uveal Melanoma
Although only a small percentage of patients with uveal melanoma have at
least one family member with uveal melanoma,
113,114
this rate is higher
than would be expected by coincidence, given the very low incidence of
uveal melanoma in the population as a whole.
115,116
Having a family
member with uveal melanoma is therefore considered a risk factor for
melanoma. Studies of families with more than one member with uveal
melanoma have shown that there are several family cancer syndromes
associated with increased risk of uveal melanoma.
BAP1 Tumor Predisposition Syndrome
Certain BAP1 germline mutations have been associated with
predisposition for uveal melanoma, malignant mesothelioma, cutaneous
melanoma, and renal cell carcinoma.
72,116-126
Some families with BAP1
tumor predisposition syndrome also tend to have atypical Spitz tumors,
which are benign/precursor melanocytic lesions that have distinctive
clinical and pathologic features.
116,120,123,127-129
In individuals with germline
BAP1 mutations associated with this syndrome, the risk of uveal
melanoma is high (up to ~30%),
123,129,130
uveal melanoma tends to develop
at a younger age,
122,129
primary lesions tend to be larger and involve the
ciliary body,
72
and the disease has a more aggressive course.
72,131
Some
individuals with this syndrome develop more than one type of primary
cancer,
129,130,132
and there is a high likelihood of BAP1-associated cancers
in first- or second-degree relatives.
116,129
PALB2
Mutations in PALB2 have been associated with increased risk for
developing breast, ovarian, and pancreatic cancer. Pathogenic variants
leading to biallelic inactivation of PALB2 were identified in tumors of two
patients with familial uveal melanoma.
133
This finding from a
retrospective case series might warrant further investigation into PALB2
as a uveal melanoma susceptibility gene.
MBD4
MBD4 deleterious mutations have been identified in uveal melanoma
tumors at increased incidence compared with the general population and
associated with high tumor mutation burden.
134,135
Questions remain
whether MBD4 germline variants or somatic loss predisposes individuals
to uveal melanoma.
136
How MBD4 inactivation might affect
immunotherapy response is also worth further investigation.
134,137
Neurofibromatosis Type 1 (NF-1)
Neurofibromatosis type 1 (NF-1): Based on case reports of uveal
melanomas developing in patients with neurofibromatosis,
138-145
this
condition is thought to be a risk factor for uveal melanoma, although
population statistics supporting this claim are lacking.
146
Other Potential Risk Factors
Other risk factors include atypical cutaneous nevi, common cutaneous
nevi, iris nevi, and cutaneous freckles. These associations are based on
moderate-quality evidence from several studies, including meta-analyses,
a systematic review, a case study of familial uveal and cutaneous
melanoma, and case-controlled studies.
147-151
Patient characteristics
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-5
associated with increased uveal melanoma risk are fair skin color, light
eye color, and propensity to sunburn.
147,152
In conclusion, the NCCN Panel recommends evaluation for evidence of
hereditary syndrome and referral for genetic counseling and testing in
case of: early age of diagnosis (<30 years of age), history of other primary
cancers in the patient, or family or personal history of other cancers known
to be associated with a hereditary syndrome (eg, BAP1renal cell
carcinoma, mesothelioma, cutaneous melanoma, cholangiocarcinoma,
meningioma; BRCA, PALB2breast, ovarian, and pancreatic cancer).
The Relationship Between Uveal and Cutaneous Melanoma
Clinic-based studies (n < 250) evaluating the likelihood of finding
concurrent cutaneous melanoma in patients with ocular melanoma
suggest a relationship between these two cancers.
151,153-155
Most large
population-based studies reveal no relationship between preexisting
cutaneous melanoma and the subsequent development of uveal
melanoma.
156-160
However, one SEER-based study revealed a potential
relationship between these two cancers.
161
Currently the NCCN Panel
does not consider cutaneous melanoma to be a risk factor for uveal
melanoma, and patients with cutaneous melanoma do not need more
frequent ocular screening than the general population.
For patients who present with uveal melanoma as their first primary
cancer, some population-based studies have shown increased risk of
subsequent cutaneous melanoma,
157-159,162
and others have not found the
risk to be significantly higher than in the general population.
163
One
analysis based on Swedish Cancer Registry data and re-analysis of
archival tissue found that some tumors originally recorded as primary
cutaneous melanomas were in fact uveal melanoma metastasis.
160
After
correcting the classification, the standardized incidence ratio (SIR) for
primary cutaneous melanoma in patients with prior uveal melanoma was
no longer significant.
160
Literature Search Criteria and Guidelines Update
Methodology
For each update to the NCCN Guidelines for Melanoma: Uveal, an
electronic search of the PubMed database was performed to obtain key
literature. The search results were narrowed by selecting studies in adult
patients published in English. Articles were also excluded if they: 1)
involved investigational agents that have not yet received FDA approval;
2) did not pertain to the disease site; 3) were clinical trial protocols; or 4)
were reviews that were not systematic reviews. The search results were
further narrowed by selecting publications reporting clinical data, meta-
analyses and systematic reviews of clinical studies, and treatment
guidelines developed by other organizations. The potential relevance of
the PubMed search results was examined by the oncology scientist and
panel chair, and a list of selected articles was sent to the panel for their
review and discussion at the panel meeting. The panel also reviewed and
discussed published materials referenced in institutional review comments
or provided with submission requests. The Discussion section was
developed based on review of data from peer-reviewed publications as
well as articles from additional sources deemed as relevant to these
guidelines and/or discussed by the panel (eg, e-publications ahead of
print, meeting abstracts). Any recommendations for which high-level
evidence is lacking are based on the panel’s review of lower-level
evidence and expert opinion.
The complete details of the development and update of the NCCN
Guidelines are available at www.NCCN.org.
Diagnosis and Workup
Characteristics, Detection, and Differential Diagnosis
The majority of uveal melanomas are symptomatic at
presentation,
9,78,164,165
but studies have reported 13% to 30% of patients
were asymptomatic at diagnosis (and are discovered by routine eye
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-6
exam).
9,78,165,166
The most common symptom is blurred vision (38%–49%);
other common symptoms include visual field defect/loss, photopsia,
irritation and pain, metamorphopsia, floaters, redness, pressure, and
change in appearance.
9,78,165
Although in patients where a lesion was
detected, current imaging techniques provide very high accuracy of
diagnosis of medium to large uveal melanomas,
167,168
real-world studies
have reported relatively high rates of delay in diagnosis and treatment
(23%–37%) due to failure to detect these lesions in the initial
ophthalmologic exam.
9,78,165,169
Another surprising result was the high rate
at which uveal melanomas were initially missed in symptomatic
patients.
9,78
These studies underscore the importance of full dilation of the
pupil and meticulous examination of the fundus in any patient presenting
with symptoms.
80
Ciliary body melanoma can be difficult to detect,
9
as it is often hidden
behind the iris,
170
and symptoms often do not develop until the tumor is
large.
9,41,46,170
Ciliary body melanoma can cause lens tilting or
displacement,
46,170
cataract development,
46,170,171
and elevated intraocular
pressure,
46
and is often associated with dilated episcleral “sentinel”
vessels,
170
but only rarely has extrascleral extension.
170
Ciliary body
melanomas usually present with a dome shape,
46,170
but occasionally have
a circumferential ring shape.
170,172
Uveal melanoma typically presents as a pigmented lesion. One study of
8033 eyes with uveal melanoma found that the lesion was pigmented in
55%, nonpigmented in 15%, and 30% had a mixture of pigmented and
nonpigmented areas.
173
Some smaller studies report that a higher
percentage of patients have lesions with pigmentation,
166
likely due to
varying clinical diagnostic practices resulting in lower detection of
nonpigmented lesions. Uveal melanoma most often presents as a dome-
shaped tumor (75%), but approximately 20% present with a mushroom
shape due to the rupture of Bruch’s membrane and growth into the sub-
retinal space.
173
A small number of cases present as a diffuse, flat,
plateau-shaped tumor (6%) or have a multinodular tapioca appearance
(<1%).
173
Subretinal fluid is present in the majority of cases (75%).
173
Uveal melanoma tumors are associated with intraocular hemorrhage in
~10% of cases, and extraocular extension is apparent at presentation in
3% of patients.
173
As described above (see section entitled Risk Factors for Uveal
Melanoma), several risk factors have been identified that may increase the
risk of uveal melanoma. Included among these are rare diseases and
family cancer syndromes. When a pigmented lesion on the ciliary body or
choroid is discovered, these factors should be evaluated, as they may
inform the index of suspicion.
Studies of lesions that were thought to be uveal melanoma but later were
assigned an alternate diagnosis showed that the most common simulating
lesion is choroidal nevus.
174-177
These studies have reported a variety of
other conditions mistaken for uveal melanoma, including congenital
hypertrophy of the retinal pigment epithelium (RPE), and choroidal
hemangioma, peripheral exudative hemorrhagic chorioretinopathy,
hemorrhagic detachment of the retina or pigment epithelium, and age-
related macular degeneration.
174-177
There is a considerable overlap in size
between small uveal melanomas and large uveal nevi, and there is
evidence from several studies that many small lesions presumed to be
nevi based on size may actually be melanoma.
88,89
Although likely but not
explored, there is the possibility that some of the treated lesions were nevi
and not melanoma. Therefore, size and appearance based on clinical
exam may not be sufficient for diagnosis.
It is also important to rule out metastasis to the uvea from other cancers
either known cancers based on patient history or occult primary cancer.
Among patients with metastases to the eye or orbit, the most common
primary cancer diagnosis is breast cancer, followed by lung cancer, which
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-7
together account for over half of the cases.
178-180
The remainder of uveal
metastases arise from a variety of cancers, each cancer type accounting
for less than 5% of the population, including cancers of the kidney,
gastrointestinal (GI) tract, skin, prostate, thyroid, pancreas, and
others.
179,180
Initial Workup of Suspicious Pigmented Uveal Lesion
Upon discovery of a suspicious pigmented lesion in the ciliary body or
choroid, clinical evaluation should be performed to determine whether the
lesion is uveal melanoma, and if so, the extent of disease and other
characteristics that should inform management. Clinical evaluation should
include a complete history and physical, including personal or family
history of prior or current cancers (outside the eye), as this may help
determine whether the lesion is primary uveal cancer or a metastasis from
another primary cancer. A wide variety of imaging and clinical exam
techniques have been tested for their utility in diagnosis and
characterization of uveal melanoma. In most cases, the diagnosis of uveal
melanoma and characterization for treatment planning (or follow-up) can
be achieved based on comprehensive exam of the front and back of the
eye including biomicroscopy and dilated fundus exam (indirect
ophthalmoscopy), along with color fundus photography, and conventional
ocular ultrasound (US).
45,46,80,181
In some cases, additional imaging may be
needed to confirm diagnosis or better characterize the tumor for treatment
planning (or monitoring). Additional imaging options that may be useful
include autofluorescence of the ocular fundus, optical coherence
tomography (OCT), retinal fluorescein angiography of the ocular fundus,
transillumination, and MRI.
Features that are essential to measure and document as part of workup
include visual acuity (VA), location and size of the tumor (ie, diameter,
thickness), distance from the tumor to the disc and to the fovea, ciliary
body involvement, and subretinal fluid and orange pigment, if present. The
reason for recording each of these features and the utility of different
techniques for diagnosis and assessment of these features is described
below.
Features to Record
Visual Acuity
It is important to measure VA during workup, as many uveal melanomas
(as well as simulating lesions) can disturb vision, and changes in VA can
be an indication of progression, response to treatment, or a side effect of
certain treatments. VA or changes in VA can contribute to the differential
diagnosis, as benign nevi rarely cause visual impairment,
76,182
whereas
visual impairment or a decline in VA is associated with malignancy.
9,78,87,96
In patients with uveal melanoma, VA in the affected eye tends to be worse
than in the fellow eye.
14
A baseline measure of VA is also important to
determine effects of uveal melanoma treatment. Radiation therapy (RT)
can cause a decline in VA, although the effects are highly variable.
183-185
Larger tumor size and tumor location near the optic disc are associated
with greater loss of VA and higher rates of local complications that result in
lower rates of improvement in VA in eyes treated with iodine-125
brachytherapy.
186-188
Size
The size of the tumor is an element that contributes to the differential
diagnosis, particularly when trying to distinguish uveal melanoma from
choroidal nevus. Although size alone does not determine diagnosis, it is
one of the features that informs the diagnosis, as uveal melanomas are
generally larger than choroidal nevi, and size (thickness >2 mm, tumor
diameter >5 mm) is predictive of growth in melanocytic lesions.
94,96
For
lesions thought to be nevi, or small lesions with uncertain diagnosis,
accurate measurements of size are important for monitoring for growth, as
rapid growth is a sign of likely malignancy.
91
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-8
As described in subsequent sections on treatment options for uveal
melanoma, tumor size (largest basal diameter and thickness) is also
important for selecting and planning treatment. For brachytherapy,
selection of the appropriate size plaque is important to ensure that the
lesion is fully covered, and alternative or additional treatment options need
to be considered for tumors that exceed the size of the largest
commercially available plaque. Selection among alternative treatment
options (eg, particle beam RT, stereotactic radiosurgery [SRS],
enucleation) should also depend on tumor size, and accurate description
of size and shape are needed for planning particle beam RT and SRS.
Accurate measurements of size and shape of the tumor are also needed
for monitoring response or progression after treatment.
As described in subsequent sections, baseline tumor size (ie, largest basal
diameter, thickness) is needed to determine the T stage of the tumor,
which is one of the elements that determines the prognostic risk category
used to inform post-treatment surveillance. (See UM-4 in the algorithm for
other determinants of the prognostic risk category).
Location, Distance from Disc and Fovea, Ciliary Body Involvement
Lesion location is one of many features that can contribute to diagnosis.
Proximity to the optic disc is considered a diagnostic feature of uveal
melanomas, as it is correlated with likelihood of growth in melanocytic
choroidal lesions.
90-92,94
As described in subsequent sections, lesion
location, including distance from disc and fovea, and ciliary body
involvement, can impact imaging results, such as the ability to detect
certain diagnostic features and the ability to accurately discern borders for
measurement of tumor size. In some cases, color fundus photography,
US, and complete clinical exam may not be sufficient for detection and/or
characterization of ciliary body involvement; this is a feature that may
require additional imaging approaches.
181
As described in subsequent
sections, lesion location may also impact the efficacy and safety of specific
treatments, and should be considered when selecting and planning
treatment. Lesion location can help explain visual symptoms, which can be
helpful for determining whether any of the treatment options are likely to
improve visual symptoms. Ciliary body involvement is also one of the
elements that determines the T stage of the tumor and prognostic risk
category used to inform post-treatment follow-up surveillance.
Subretinal Fluid
Subretinal fluid is another feature that supports the diagnosis of uveal
melanoma, as it is rare in benign choroidal nevi,
85,99
and has been shown
to develop during growth of choroidal nevi and transformation to
melanoma.
79,86
The presence of subretinal fluid has been shown to be
predictive of growth in choroidal nevi or small indeterminate melanocytic
lesions.
86,91,92,94,189
One study of a large number of uveal melanomas (n =
8033) reported that subretinal fluid was present in 75% of cases.
173
However, ocular metastases from other types of primary cancer (eg,
breast, lung) can also give rise to subretinal fluid,
179
which is why other
features must also be considered for a differential diagnosis.
Orange Pigment
Orange pigment, also called lipofuscin, can be present in benign choroidal
nevi;
85,99
however, it has been shown to develop during nevi growth and
transformation to uveal melanoma.
79,86
In nevi or indeterminate small
lesions, the presence of orange pigment is predictive of future growth and
increased likelihood of future diagnosis as a uveal melanoma.
83,86,90-96
Therefore, it is important to record the presence of orange pigment during
initial clinical exam.
Imaging Methods
Comprehensive Eye Exam (biomicroscopy)
Initial workup should include a comprehensive eye exam using
biomicroscopy to examine the front and back of the eye, including a
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-9
dilated fundus exam (indirect ophthalmoscopy). Meticulous analysis of the
fundus after full pupil dilation will allow detection of most choroidal
melanomas,
80
and can be used to record many of the relevant features
needed for diagnosis and treatment planning. Whereas ciliary body
melanomas can be difficult to detect by this method, fundus exam with
good indentation can sometimes help.
41
Color Fundus Photography
Color fundus photography is useful for documenting the clinical features of
lesions, with the advantage of giving the most identical appearance to
clinical examination.
190
Color fundus photography can be used to evaluate
the borders of the lesion, and thereby record the location and shape;
calculate lesion basal diameter and area; and be used to detect orange
pigment, drusen, and halo.
80,190-192
Areas of orange pigment seen by color
fundus photography have been shown to correlate with those seen by
fundus autofluorescence.
191,192
Obtaining a baseline image is important
because serial fundus photography helps to monitor nevi or indeterminate
lesions for growth, and for those diagnosed with uveal melanoma, helps in
monitoring response to treatment and surveillance for recurrence.
Because serial images are often compared, it is important that the
baseline color photograph includes all the tumor margins.
171
A wide-angle
camera may be needed to capture choroidal lesions that are particularly
large or peripheral.
80
It is important to note that color fundus photography
alone is not sufficient for a differential diagnosis, and diagnostic accuracy
based on color fundus photography has wide interobserver variation.
193
Ocular Ultrasound
In addition to comprehensive clinical ophthalmologic exam and color
fundus photography, US of the eye (ocular echography) is one of the most
useful tools for diagnosis of choroidal melanomas.
45,194-196
In experienced
hands, US in combination with complete ophthalmologic clinical exam
results in a high level of diagnostic accuracy (>99%) for studies of medium
to large uveal melanomas that confirmed diagnosis based on histologic
evaluation after enucleation.
167,168,197
Melanomas tend to exhibit low
internal reflectivity as well as an intrinsic acoustic quiet zone on US,
196
features that distinguish them from a variety of other intraocular
conditions. US is particularly useful when a mass cannot be visually
inspected due to opacity or pathology of structures in the anterior portion
of the eye such as corneal scars, cataract, or blood in the vitreous.
45
US can help in the detection of ciliary body melanomas that may be
missed by fundoscopy.
198
US is particularly useful for determining the
thickness of the tumor,
171,194
and has been shown to have a high level of
accuracy compared with tumor height measurements based on
histopathology of enucleated specimens.
199,200
For measurements of tumor
thickness it is important to place the caliper at the internal scleral surface,
hold the probe at right angles to the scleral and tumor surfaces, and
account for overlying retinal detachment.
171
By facilitating measurement of
tumor dimensions, US is useful for detecting lesion growth.
170,195
US can
also be used to detect extraocular extension,
45
which shows
hyporeflectivity on US.
80
There are two modes of US to evaluate the eye, A-mode and B-mode.
Uveal melanomas show low reflectivity on both A-scan and B-scan
ultrasonography, although other features differ, and these two modes have
different uses.
80
A-Scan Ultrasonography
Uveal melanomas typically show low to medium internal reflectivity on A-
mode US,
41,45,170,195,201
which further decreases toward the sclera.
45,80
Other A-scan US hallmarks specific to uveal melanoma are: 1) a regular
internal structure with similar height of the inner tumor spikes or regular
decrease in height (positive angle kappa sign); 2) solid consistency with
no aftermovement of tumor spikes; and 3) echographic sign of
vascularization with a fast, spontaneous, continuous, flickering vertical
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-10
motion of single tumor spikes.
41,201
In contrast, choroidal metastases from
other types of cancer typically show an irregular structure on A-mode
US,
201
hemangiomas show much higher reflectivity,
45,195,201
and nevi and
melanocytomas show higher reflectivity and more irregular structures than
malignant melanoma.
201
Fresh choroidal hemorrhages may have similar
structure and reflectivity as melanomas, but will show aftermovement
following small eye movements.
201
B-Scan Ultrasound
On B-scan US, posterior uveal melanomas appear as hyperchromic mass
with lower reflectivity than the surrounding choroid, thus giving an
acoustically hollow appearance.
41,45,80,170
Posterior uveal melanomas,
particularly the larger ones, show choroidal excavation and orbital
shadowing on B-scan US.
41,45,170,195
These features help confirm clinical
diagnosis, and are distinct from hemangiomas or metastases to the eye,
which typically show high reflectivity.
170,202
However, some other lesions
that may show choroidal excavation include: hemangiomas, long-standing
nevi, and choroidal metastases.
197
B-scan US is used for obtaining tumor
dimensions, extent, and shape,
41,195,202
and is useful for characterizing
larger ciliary body tumors.
41
Extraocular extension can be observed by B-
scan US as areas of hyporeflectivity compared to normal orbital
tissue.
80,170
Additional Imaging
Depending on the disease characteristics observed by clinical evaluation,
additional testing options may be needed to either confirm diagnosis or
assess the extent of disease to determine first-line treatment options.
Additional imaging options that may be considered in certain situations
include: autofluorescence of the ocular fundus, OCT, retinal fluorescein
angiography of the ocular fundus, and transillumination. In select
situations, MRI is occasionally needed to confirm diagnosis or to plan
treatment. In most cases these imaging methods are not needed, as
equivalent or better information can be obtained through standard US
combined with comprehensive clinical exam.
Autofluorescence of Ocular Fundus
Fundus autofluorescence has been proposed as a method to help in the
diagnosis and characterization of uveal melanomas.
171,191,203,204
Choroidal
melanoma generally shows slight intrinsic hyperautofluorescence and the
brightness increases with tumor pigmentation and disrupted RPE.
205,206
Orange pigment (lipofuscin) is the most highly autofluorescent uveal
melanoma feature.
203,205-210
Drusen can also be detected by
autofluorescence,
209
and have increased, normal, or decreased
autofluorescence.
203,207,210
Fibrous metaplasia also shows elevated
autofluorescence.
206,207,209
Autofluorescence can be used to distinguish
orange pigment from drusen in both pigmented and non-pigmented
tumors.
170
Unlike uveal melanoma and choroidal nevi, melanocytomas show
hypofluorescence.
211
Comparative studies have shown that the
autofluorescence pattern often matches that of orange pigment and
hyperpigmentation seen by color fundus photography.
45,192,207
Based on
comparative studies, it is not clear that fundus autofluorescence increases
the detection rate of orange pigment relative to standard ophthalmologic
exam.
212
Optical Coherence Tomography
OCT is another imaging method proposed for assisting diagnosis of uveal
melanoma
45,171
and treatment planning.
45
Spectral domain OCT (SD-OCT)
allows the detailed evaluation of the retina and RPE, changes in which are
more common in choroidal melanoma versus choroidal nevi.
45,203
OCT can
help identify overlying retinal detachment or edema, even before clinically
apparent.
203
Results differ across comparative studies regarding whether
OCT is more sensitive than standard ophthalmologic exam plus US for
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-11
detecting subretinal fluid.
212-214
OCT can detect subretinal and intraretinal
fluid, subretinal lipofuscin, retinal epithelium atrophy and degeneration,
shaggy photoreceptors, and structural loss of photoreceptors in the
neurosensory retina.
45,170,203,215,216
These features are more likely in
choroidal melanoma than in choroidal nevi, although some features are
more useful than others for differential diagnosis, especially in small
lesions.
45,214,215,217,218
In OCT, the structure of the lesion differed between choroidal nevi,
melanomas, hemangiomas, and metastases.
216
Optical density ratio based
on OCT can also be used to distinguish choroidal melanomas from
choroidal metastases.
219
Enhanced depth imaging OCT (EDI-OCT) is
particularly useful for detecting small lesions, and distinguishing small nevi
from small choroidal melanoma. It is less useful for thick tumors.
41
OCT
angiography (OCT-A) can be used to distinguish choroidal nevi from
choroidal melanoma based on margin character (well-delimited vs.
imprecise), reflectivity of choroid capillary vasculature (hyper vs. hypo),
and characteristics of lesion vasculature.
220,221
Both OCT and US have
been evaluated for characterizing iris and ciliary body tumors. US
biomicroscopy provides better overall visualization and better resolution of
the posterior margin, while OCT provides better resolution of the anterior
segment anatomy and margin.
222
Retinal Fluorescein Angiography
Retinal fluorescein angiography can be used to characterize lesion
vasculature, which can aid in diagnosis because choroidal melanomas
may have intrinsic tumor circulation, sometimes called “double circulation,
in addition to normal choroidal vasculature.
41,45,80,170,195,196,223
Observation
of the tumor vasculature is helpful in distinguishing melanomas from
choroidal nevi, hemorrhagic degeneration, or choroidal melanoma.
45,80,224
After treatment with brachytherapy, fluorescein angiography is also useful
for detection of complications such as radiation retinopathy.
45,80
Transillumination
Transillumination has been tested as a method for detecting and
measuring uveal melanomas that may be difficult to detect or fully
characterize by other techniques. Examples include large ciliary body
melanomas,
41,45,80
or tumors obscured by cataracts.
225
Compared with
tumor dimension measurements based on histology of enucleated eyes,
transillumination tends to overestimate both the thickness and diameter of
tumors, and is prone to shadowing artifacts.
200,226
Therefore, it is only used
if other methods have proven inadequate.
MRI
MRI is generally not needed for diagnosis and workup, but occasionally is
necessary in cases with features that may make other imaging methods
difficult, such as secondary vitreous hemorrhage, extensive retinal
detachment, or cataract.
45,80
MRI may be useful in determining whether
cataract is caused by an underlying ciliary body melanoma, and can help
distinguish uveal melanomas from hemangiomas.
45
Uveal melanomas
usually have high signal intensity in T1-weighted MRI images and low
signal intensity in T2-weighted images.
41,45,80,170
Although similar signal
pattern can be caused by hemorrhage or necrosis,
45,80
hemangiomas
typically have hyperintense signal on T-weighted images and T2-weighted
images are isointense with the vitreous.
45
MRI is useful for detecting and
characterizing extraocular extension,
45,80,170,194,227,228
and is also used for
RT planning.
80,170,194,229,230
Biopsy
The use of biopsy as part of workup for uveal melanoma is an issue of
some debate. Potential benefits of biopsy during workup are 1)
cytologic/histologic confirmation of diagnosis; 2) potential for molecular
analysis that may impact eligibility for current or future clinical trials; and 3)
potential for molecular analyses that may provide more accurate
prognostic assessment for risk of metastasis, which may inform future
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-12
follow-up surveillance. Potential harms of biopsy of the primary tumor
include 1) risk of complications from the procedure that damage the eye;
2) the risk of tumor seeding leading to local or distant recurrence; and 3)
the risk of inadequate sampling resulting in misdiagnoses or inaccurate or
inconclusive molecular testing results. As discussed in greater detail
below, the likelihood of each of these potential benefits and potential
harms is debated, as there are many different biopsy methods than can be
used, and the likelihood of benefit versus harm may also vary across
practitioners.
There are a wide variety of biopsy techniques that have been tested and
are sometimes used for choroidal or ciliary body tumors. Some involve a
transscleral (direct) approach, where the tumor is approached from the
outside, the needle first puncturing the sclera over the tumor, then the
tumor itself, leaving the retina intact.
231
Others use a transvitreal (indirect)
approach, with anterior entry through the pars plana opposite the tumor,
going through the vitreous body and retina to reach the tumor.
231
Tumor
location is a major determinant of which approach is likely to be successful
and safe. Table 2 provides a list of techniques (or categories of
techniques) for biopsying choroidal and ciliary body tumors that have been
used to assist with diagnosis and/or prognostication in primary uveal
melanomas. Key features of these biopsy methods are also included in
Table 2: the typical surgical approach (transscleral or
transvitreal/transretinal), tumor location(s) for which the biopsy method
was developed and/or is most often used, and the type of sample
obtained, as some of these methods provide aspirated cells that can be
analyzed by cytopathology, whereas other methods can provide tissue
samples that can be sectioned for histopathology. Table 2 also lists
representative studies that describe these biopsy methods in more detail,
report diagnostic and/or prognostic yield for these biopsy options (percent
of biopsies providing sufficient material for diagnostic or prognostic
analyses), and provide safety data, including intraoperative or
postoperative procedure-related complications and analyses aimed at
determining the risk of tumor seeding (eg, evidence of tumor cell
tracks/contamination in neighboring tissues; rates of local recurrence
during follow-up).
There is some risk of complications with any biopsy procedure. Any of
these procedures can result in the following intraocular
complications/morbidities, which if severe may require secondary
procedures or other interventions for management: hemorrhage (eg,
vitreal, subretinal, choroidal, perilesional), decrease in VA, retinal
detachment (eg, rhegmatogenous, exudative), retinal perforation,
hypotony, and endophthalmitis.
232-243
Longitudinal population-based
studies have shown that biopsy does not impact all-cause mortality or
disease-specific deathfor better or worsein patients with uveal
melanoma.
244-246
Fine-Needle Aspiration Biopsy
Fine-needle aspiration biopsy (FNAB) is the simplest, most inexpensive,
and most commonly used method for biopsy of choroid or ciliary body
tumors presumed to be melanoma.
231,247,248
Details of the technique are
described in several review articles,
231,247,248
as well as multiple primary
reports from many different centers.
88,232,233,235,249-251
FNAB can be done
via a transvitreal or transscleral approach depending on the location of the
tumor. The transvitreal approach is generally easier due to better
intraoperative illumination and visualization options.
231,248
The disadvantage to FNAB is that the amount of material obtained may be
small, and multiple passes may be needed to obtain enough material for
cytologic and molecular genetic analysis.
232,236,251,252
The yield from
FNABboth for cytologic confirmation of diagnosis and for molecular
analyses for prognosticationvaries across studies.
88,232,233,235,236,245,246,249-
258
Some studies suggest that transvitreal (vs. transscleral) approach is
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-13
associated with improved FNAB yield, but some studies found no
difference in yield.
232,233,235,256,258,259
Some studies have suggested that
FNAB yield is higher for larger tumors, and is particularly impacted by the
thickness of the tumor,
233,246,250,256-258
but a recent study showed that high
yields (>87%) can be obtained even in thin tumors (<3.5-mm thick), for
both the transscleral and transvitreal approach.
232
Studies of patients with
uncertain diagnoses based on standard noninvasive techniques have
shown that FNAB can help distinguish between uveal melanoma and
borderline or benign melanocytic nevus, even among small lesions,
88,257
and can clarify whether a lesion is a primary uveal melanoma versus
metastasis from another cancer, another type of primary ocular tumor, a
melanocytoma, or RPE proliferation.
251
For FNAB, the most common complication is vitreal hemorrhage, but its
rate varies widely across studies.
232,233,236,245,250,259
Some studies describe
methods to reduce or prevent this.
254
Most vitreal hemorrhages are
focal/localized and resolve without further intervention,
232,233,245,250,251,259
but
some are diffuse, more extensive, persist and/or impact VA, and require
secondary surgical intervention (eg, vitrectomy) for
management.
232,233,235,236
The risk of hemorrhage requiring secondary
surgical management varies widely, even across recent studies, ranging
from 1% to 15% of patients with FNAB.
232,233,235,236,250
Some studies have
suggested that vitreal hemorrhage is more likely with a transvitreal than
with a transscleral approach.
232,235
FNAB may also impact VA, improving
some cases but reducing VA in other cases.
233
Other complications that
have been reported, but are rare, include rhegmatogenous retinal
detachment and worsening exudative retinal
detachment.
233,235,236,245,246,251,259
Concerns about FNAB causing tumor seeding have been raised based on
findings of melanoma cells left in the needle track,
253,255,260-262
and some
have suggested procedure adjustments for reducing the likelihood of
tumor cell seeding FNAB.
248,253,261
A few case reports found local
recurrences at FNAB entry sites.
263-265
However, multiple follow-up studies
with large patient populations have revealed no local recurrences after
FNAB (and non-surgical treatment),
88,232,235,245,246,250,251,254,255,259
and one
case series found no increased risk of metastasis among untreated
patients who had post-biopsy evidence of melanoma cell dissemination
inside the eye.
260
FNAB is therefore generally thought to be safe and to
have low risk of seeding.
Other Biopsy Techniques
Other biopsy techniques that have been tested in large populations of
patients with uveal melanoma are those that use tools from vitrectomy
systems both to access tumor via a transvitreal/transretinal approach and
to extract tumor tissue using the vitreous cutter and aspiration through the
canula. These procedures do not necessarily include a vitrectomy. There
are a variety of procedures that fall into this category, and are described in
a series of publications based on clinical practices both in the United
States and Europe, including several reviews.
231,237,238,240,248,266,267
These
techniques have been used successfully on anterior, posterior, equatorial,
and peripapillary lesions.
237-240,268
They generally result in larger sample
sizes than FNAB, although like FNAB multiple passes may be
necessary.
237,238,266,267
For this reason it has been suggested that these
methods may be useful in patients with tumors that are too small or
inaccessible for FNAB, or for tumors where FNAB has failed. One study
reported that for choroidal melanomas with thickness of 2.0 mm or less,
sufficient sample for prognostic evaluation was obtained in 100% of
patients.
237
Reported yields from these procedures range from 89% to
99% for diagnosis of choroidal lesions, and 97% to 100% for prognostic
testing on uveal melanomas.
237-240,267,268
Studies have shown that these
biopsy techniques can be used to diagnose tumors that are unclassifiable
based on standard noninvasive diagnostic techniques, and can
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-14
differentiate uveal melanoma from benign nevus, metastases from other
cancers, vasoproliferative tumor, hemorrhage, gliosis, and scleritis.
240,268
These approaches are less broadly used than FNAB, are more expensive,
and require additional expertise.
231,248
Like FNAB, hemorrhage is the most
common complication with these biopsy procedures, is normally localized,
and usually resolves without intervention.
237,239,240
Because these
techniques are more invasive, however, retinal detachment and severe
hemorrhage are more common, and more patients require intervention for
management, either at the time of biopsy or as a later procedure.
234,238-
240,248
Decrease in VA due to these procedures is not uncommon.
238,239
Like FNAB, studies have published evidence that these procedures may
leave tumor cells along the access pathways, but most studies, including
those with large patient samples, have not observed local recurrence
during follow-up.
239,240
There are a few cases reported of local recurrences
at biopsy entry sites.
264,268-270
Incisional biopsy techniques are more
invasive than vitrectomy system-assisted biopsies, and likewise yield more
material, but also are more likely to lead to complications.
241-243
These
methods have been used for cases that are particularly hard to diagnose.
Excisional biopsy, using either transscleral resection or endoresection,
was also explored as an option for both biopsy and primary treatment,
271
but is not included in Table 2 because it is no longer used due to technical
challenges, risk of complications, and concerns about tumor
seeding.
170,272-282
NCCN Biopsy Recommendations
Biopsy may be considered if needed to confirm diagnosis or for prognostic
analysis for risk stratification. Biopsy is usually not necessary for initial
diagnosis of uveal melanoma and selection of first-line treatment, but may
be useful in cases of uncertainty regarding diagnosis, such as for
amelanotic tumors or retinal detachment. Biopsy of the primary tumor may
provide prognostic information that can help inform frequency of follow-up
and may be needed for clinical trial eligibility. If biopsy is performed,
molecular/chromosomal testing for prognostication is preferred over
cytology alone. The risks/benefits of biopsy for prognostic analysis should
be carefully considered and discussed.
Observation for Uncertain Diagnosis in Patients with Low
Risk
Findings from the clinical workup should be used to determine initial
management. Observation may be appropriate for patients with uncertain
diagnosis and/or fewer than three risk factors for lesion growth (Table 1).
Studies have found that for patients with small choroidal lesions presumed
to be nevi or indeterminate, deferring treatment until evidence of growth or
features of malignancy develop (eg, orange pigment, subretinal fluid,
symptoms) is associated with a very low risk of metastasis,
91,95
and even
lower risk of death from uveal melanoma.
84
For patients who meet the
criteria for observation (rather than immediate treatment), regular follow-up
is recommended to periodically re-evaluate for growth or features of
malignancy. Follow-up tests should include the same tests recommended
for initial workup and diagnosis that would help clarify if there is
progression and determine the natural history of the indeterminate lesion.
Initially (ie, upon first discovery of the lesion), these patients should be re-
evaluated every 2 to 4 months to determine rate of growth (if any) and to
monitor for other changes indicative of malignancy. Close follow-up for 5
years is recommended to firmly establish whether or not there is any
growth or progression; some lesions that initially seem stable may
suddenly begin to grow and transform. The frequency of re-evaluation
should depend on the index of suspicion, patient age, and medical frailty.
For example, the presence of one to two risk factors for growth (Table 1),
or evidence of changes would increase suspicion. If the size and features
of a lesion appear static after 5 years of follow-up, the patient can be
followed annually thereafter. Lesions that demonstrate growth or develop
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-15
additional risk factors for growth (>3 total) should be managed as uveal
melanoma, even if diagnosis is still uncertain.
Further Workup Prior to Treatment
Further workup prior to treatment may be needed in some cases to aid in
treatment selection and planning. For example, for tumors that are large,
close to the optic nerve, or have suspected/confirmed extraocular
extension, MRI should be performed, if not previously done, to determine
whether radiation (particle beam or SRS) is an option, or whether
enucleation is needed, and for radiation planning. MRI should be
performed with and without contrast unless contrast is contraindicated.
MRI is useful for detecting and characterizing extraocular extension and
for RT planning (particle beam or SRS).
45,80,170,194,227-229
Ciliary body
involvement or extraocular extension should be assessed and
documented, as these features may impact the feasibility, safety, and
efficacy of certain treatment options. If not already performed, biopsy of
the primary tumor should be considered for prognostic analysis, as risk
stratification should inform the frequency of follow-up treatment. There is
some evidence to suggest that radiation (all modalities) may alter
molecular genetic features of the tumors, reducing the accuracy of
prognostication based on samples taken after radiation treatment.
283
Baseline imaging to screen for systemic disease is also recommended
prior to treatment. Despite lack of treatment options for patients with
metastatic disease, NCCN favors staging before primary treatment. For
patients who have small, low-risk tumors, but are planning to receive
treatment (ie, those with a definite diagnosis of uveal melanoma, or
uncertain diagnosis but three or more risk factors for growth [Table 1]),
deferring extraocular baseline imaging until after primary treatment can be
considered. The most frequent sites of metastasis are liver, lungs,
skin/soft tissue, and bones.
21
At minimum, all patients receiving baseline
imaging should have contrast-enhanced MR or US of the liver, with
modality preference determined by expertise at the treating institution.
Additional imaging modalities may include chest/abdominal/pelvic CT with
contrast. However, screening should limit radiation exposure whenever
possible. Unless there is a specific contraindication to the administration of
IV contrast (ie, renal impairment or history of a severe allergy), all cross-
sectional imaging studies should be performed with and without IV
contrast.
If not already performed, biopsy of the primary tumor for prognostic
analysis should be considered prior to treatment.
Treatment of Localized Primary Uveal Melanoma
Most uveal melanomas are localized at first presentation, and only a small
percentage of cases present with metastases (<3%).
6,10-14
Local treatment
for primary uveal melanoma is effective in preventing local recurrence in
greater than 85% of cases,
272,284
yet the rate of metastasis within 20 years
after treatment is approximately 20% to 70% for patients who present with
localized uveal melanoma, depending on tumor stage/size at
diagnosis.
8,13,26,285
Whereas surgical approaches are the mainstay of
treatment for localized cutaneous melanoma, and historically most uveal
melanomas were treated with surgery, the field has moved away from
using surgery in all patients,
2,286
because different modalities, primarily
various forms of RT, have been found to be just as safe and effective for
those with limited disease, and can preserve the affected eye. Some
surgical approaches are still used in patients with extensive local disease,
but most patients with localized primary uveal melanoma are treated with
some form of RT.
287
There are a number of other ablative techniques that
are occasionally used for localized primary melanoma, including laser
therapy, cryotherapy, and photodynamic therapy. Each of these primary
treatment modalities is described in more detail below. Selection among
these techniques is guided by many case-specific factors, including the
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-16
size and location of the tumor, presence of extraocular extension, visual
potential, patient age, and preference.
Surgical Options
Prior to the development of effective RT options, surgery was used to treat
most uveal melanomas.
Local Resection
There are a variety of methods for local resection of uveal melanoma
aimed at conserving the eye and useful vision.
170,280,281
These include
transretinal (endoresection) and transscleral (exoresection) approaches.
170
These methods can be technically challenging,
41,170
with high rates of
immediate postoperative complications such as hemorrhage, retinal
detachment, ocular hypertension, and proliferative vitreoretinopathy, which
may require repeat surgery.
170,277,280-282
Endoresection is the less
technically challenging approach,
280,281
and based on retrospective
analyses is associated with low local failure rate (<6%).
272,288-293
Due to
lack of prospective data on this technique, it is unclear whether it provides
the same protection from recurrence and metastases as treatment options
that have been prospectively studied (ie, brachytherapy, enucleation).
Exoresection is the more technically challenging form of local resection,
particularly with large and posterior tumors, and is usually performed with
hypotensive anesthesia and other measures to control intraoperative
hemorrhage.
277,280,281
Most studies (all retrospective) have reported high
local failure rates (20%–24%) after trans-scleral resection,
272-275
which
tend to be higher than with enucleation or brachytherapy.
276-279
Local resection is not recommended in the NCCN Guidelines for
Melanoma: Uveal as a primary treatment option for choroidal or ciliary
body melanoma. For patients with primary tumors amenable to eye-
conserving approaches, RT-based approaches are preferred. For tumors
too large for brachytherapy, enucleation is preferred over local resection,
as the latter is technically difficult for large tumors.
Enucleation
Enucleation is a technically less challenging procedure than local
resection of uveal melanoma, and historically is the most widely used
treatment for uveal melanoma. Results from the Collaborative Ocular
Melanoma Study (COMS) prospective randomized trial suggest that
enucleation is associated with a very low risk of local recurrence
(~1%),
294,295
notably lower than the rate of local recurrence reported for
retrospective studies in patients treated with primary local resection.
272-
275,288-293
Enucleation procedures have been standardized; they involve
complete removal of the eye and in most cases include insertion of an
orbital implant.
170,281,296,297
Both porous and nonporous implants have been
shown to result in similar outcomes, although there may be a higher
incidence of ptosis with acrylic implants, and a greater need for ocularists’
treatment (eg, topical antibiotics, polishing or refitting of prosthesis) with
hydroxyapatite implants.
298
For enucleation, the complications reported in the COMS trial, during or
less than 24 hours following surgery, include pain, hemorrhage,
nausea/vomiting, cardiovascular or pulmonary problems, urinary retention,
fever, and local surgical problems.
294
Complications 1 to 6 weeks after
surgery included pain requiring longer hospital stay, pain requiring
medication, conjunctival wound dehiscence, infection, decreased facial
sensation, eyelid swelling, inflammation, implant displacement, loss of
hair, ptosis, conjunctival chemosis, ecchymosis, and orbital or conjunctival
hemorrhage.
294
Long-term follow-up has shown that other problems after
enucleation include poor motility of prosthesis, poor alignment of
prosthesis, severe ptosis, and displacement of implant.
294
Enucleation can
also result in phantom eye syndrome, including visual sensations, seeing,
and pain, which can be distressing to some patients.
299
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-17
Pre-enucleation RT is generally not used because results from the COMS
randomized trial in large tumors (height ≥2 mm and diameter ≥16 mm; or
height ≥10 mm and any diameter; or height ≥8, any diameter, if proximal
tumor border <2 mm to optic disc) showed that pre-enucleation RT had no
impact on survival (death from melanoma metastasis, all-cause death)
compared with enucleation alone,
294,300,301
confirming results of prior
retrospective studies.
302
In the COMS trial for large tumors, 5-year tumor-
related mortality was 28% for patients treated with enucleation.
300
In the COMS randomized trial in medium choroidal melanoma tumors
(height 2.510 mm, diameter ≤16 mm, and no extrascleral extension ≥2.0
mm thick), outcomes (ie, cumulative mortality, melanoma-specific
mortality) for enucleation were similar to those with iodine-125
brachytherapy.
303,304
For medium tumors, the 5-year disease-specific
survival rate was 11% for patients treated with enucleation (14% and 5%
for patients with tumor diameter >11 mm and ≤11 mm, respectively).
304
One prospective and several retrospective studies also found that survival
was similar after enucleation versus cobalt plaque brachytherapy,
305-309
or
a mix of brachytherapy plaque types,
310
or versus proton beam RT.
311
Retrospective studies suggest that outcomes (ie, overall survival [OS],
metastasis-free survival, melanoma-related mortality) are similar for
enucleation versus proton beam RT or versus SRS.
312-315
Despite the
negative aspects of enucleation (relative to RT therapy), including worse
effects on certain visual functions (eg, peripheral vision, night driving,
judging distances), greater decrease in role functioning, and larger
reductions in physical and functional well-being, some studies have found
that overall quality of life for patients undergoing enucleation appears to be
similar to that for those treated with RT.
316-321
Based on results of the prospective studies comparing enucleation with
brachytherapy, enucleation is generally only recommended for patients
with tumors that are unsuitable for brachytherapy treatment, such as those
that are too large to be effectively treated by commercially available
plaques, or that have optic nerve involvement. For such tumors,
enucleation is an option, but other types of RT (ie, particle beam,
stereotactic RT [SRT]) are also possibilities. Enucleation is sometimes
reserved for cases that would be difficult to treat using only RT, such as
those with neovascular glaucoma, tumor replacing greater than 50% of the
globe, blindness, painful eyes, or extensive extraocular extension. In
addition to use as a primary treatment, enucleation is also often used as a
secondary therapy for patients who develop local recurrence or
complications after eye-sparing primary treatment. Pathologic evaluation
should follow the uveal melanoma synoptic report recommendations by
the College of American Pathologists (available at:
http://documents.cap.org/protocols/cp-uveal-melanoma-17protocol-
4000.pdf).
322
Radiation Therapy
RT is the most commonly used first-line treatment for uveal melanoma,
2
as
several RT approaches have been shown to have similar efficacy as
enucleation for reducing the risk of metastasis and death from disease.
303-
315
Brachytherapy and charged particle RT are the RT modalities
considered appropriate for primary therapy for most cases of uveal
melanoma, whereas photon RT and SRT are less often used as primary
treatment for uveal melanoma. SRS is sometimes used for large primary
tumors, and photon RT is generally only used as an adjuvant to surgery.
Plaque Brachytherapy
Plaque brachytherapy is a commonly used form of definitive RT for the
primary tumor.
286,287,323,324
Brachytherapy is often used (for localized
primary uveal melanoma) based on results from a large prospective
randomized trial (COMS) showing that long-term outcomes were not
significantly different with plaque brachytherapy (n = 657) versus
enucleation (n = 660) for patients with small- to medium-sized choroidal
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-18
melanomas (2.510.0 mm in apical height [2.5-8.0 mm if peripapillary] and
≤16 mm in maximum basal diameter, no extrascleral extension ≥2.0 mm
thick).
303,304
In this study the 5-year risk of treatment failure after
brachytherapy was 10.3%.
187
Treatment failure was defined as tumor
expansion (≥15% increase in height or ≥250 µm in any tumor boundary) or
extrascleral extension (>2 mm). Risk factors for treatment failure were
older age, greater tumor thickness, and proximity of the tumor to the foveal
avascular zone. Other more recent studies have reported local failure
rates ranging from 0% to approximately 20% for patients treated with
iodine-125 plaques, and local failure rates were in this range for patients
treated with other types of brachytherapy plaques (ie, ruthenium-106,
palladium-103, cesium-131).
272,325-328
It is important to note that late
treatment failures (up to 12 years) after brachytherapy have been
observed.
327
In the COMS randomized trial in medium choroidal melanoma tumors
(height 2.510 mm, diameter ≤16 mm, no extrascleral extension ≥2.0 mm
thick), after a minimum of 5 years of follow-up (range, 515 years), there
were no treatment-dependent differences in all-cause mortality or death
with confirmed melanoma metastasis.
304
There was no difference across
arms in the rate of death with histologically confirmed metastasis
(enucleation vs. brachytherapy: 11 vs. 10% at 5 years; 17 vs. 18% at 10
years) or all-cause mortality (19% at 5 years and 35% at 10 years, for
each arm).
304
The only factors correlated with these outcomes were age
and maximum basal diameter, but even after adjustment for these
variables, there were no treatment-dependent differences in all-cause
mortality or mortality with confirmed melanoma metastasis at time of
death.
304
In the same trial, intraoperative/immediate postoperative complications
observed with similar frequency across brachytherapy and enucleation
arms included anesthetic complications, pain requiring medication, other
hemorrhage, cardiovascular or pulmonary problems, urinary problems,
and local surgical problems.
303
Immediate complications seen only in the
brachytherapy arm included intraocular hemorrhage, scleral perforation,
and vortex vein rupture.
303
In the brachytherapy arm, the most common
long-term complications were loss of VA and growth of tumor or other
indications that lead to enucleation.
303
After 3 years of follow-up,
approximately half of the patients (49%) treated with brachytherapy lost six
or more lines of VA (compared with before treatment), and of patients with
VA better than 20/200 before treatment, 43% had VA of 20/200 or
worse.
186
Factors associated with loss of VA included greater baseline
tumor apical height, shorter distance between the tumor and the foveal
avascular zone, presence of tumor-associated retinal detachment, non
dome-shaped tumor, and patient history of diabetes.
186
During the first 5
years of follow-up, cataracts developed in 68% of eyes treated with
brachytherapy, and 12% had undergone cataract surgery.
329
Cataract
surgery results in VA improving by 2 or more lines in 66%, and stabilizing
VA in 26%.
329
The 5-year cumulative rate of enucleation was
approximately 12%,
187,303
most often due to treatment failure during the
first 3 years after brachytherapy, and to eye pain beyond 3 years after
treatment.
187
When evaluating patients for brachytherapy, it is important to consider the
entry criteria and treatment parameters used in the COMS trial that
compared brachytherapy with enucleation. The COMS trial included only
patients with tumors that were choroidal;
303
those with tumors contiguous
with the optic disc were excluded, as were those with metastases from
melanoma or another cancer (except nonmelanoma skin cancers).
304
Only
16% of patients had tumors less than 2.0 mm from the optic disc.
303
Most
of the tumors included were dome-shaped on B-scan US (77%), and
about half had non-rhegmatogenous retinal detachment (54%55%); a
few (<1%) had rhegmatogenous retinal detachment.
303
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-19
Prospective studies in patients with small choroidal tumors also found that
iodine-125 brachytherapy resulted in tumor regression in most cases
(98%),
330
less than 3% recurred, and 98% achieved globe conservation.
331
Melanoma-specific mortality at 5 years was 3.9%.
331
Whereas the plaques used in the COMS trial were all iodine-125, one
prospective and several retrospective studies also found that survival was
similar after enucleation versus cobalt plaque,
305-309
or a mix of
brachytherapy plaque types.
310
A meta-analysis of studies testing Ru
plaques in patients with uveal melanoma reported a 5-year melanoma-
related mortality rate of 6% for small and medium tumors (T1/T2), and
26% for large tumors (T3).
332
Palladium-103 brachytherapy plaques also
appear to perform similarly to iodine-125 plaques.
333
Recent analyses, including a few prospective studies, have aimed at
identifying factors associated with loss of VA after brachytherapy, with
varying results. Factors identified in one or more studies include applicator
size, tumor basal diameter, juxtapapillary location, dose (close to foveola,
or retinal), increased tumor height, radiation maculopathy, and radiation
optic neuropathy.
184,334,335
Some studies have reported adjustments to
technique that may reduce the risk of vision decline.
328
One prospective
study of 650 patients with medium-sized choroidal melanoma found that
retinal hemorrhage in the macular and peripapillary zone, optical disc
hemorrhage, microaneurysms, and foveal RPE atrophy were more
prevalent and severe after brachytherapy than before treatment.
336
Macular angiographic leakage tended to worsen after brachytherapy, and
optic neuropathy was present in 27% of patients 5 years after treatment.
336
NCCN Recommendations for Brachytherapy
Plaque brachytherapy is appropriate as an upfront therapy after initial
diagnosis, or after local recurrence following a prior local therapy. Plaque
brachytherapy is appropriate for patients with tumors 19 mm or less in
largest base diameter, and 10 mm or less in thickness, based on the size
of the largest commercially available plaques. The plaque margin on the
tumor border should be 2 mm or greater when feasible (diameter of plaque
≥4 mm larger than largest base diameter of tumor; the plaque should
cover the tumor with a ≥2-mm circumferential margin). The exception is for
tumors near the optic nerve where it may be impossible to achieve
adequate coverage of the margins. The largest commercially available
brachytherapy plaque is 23 mm in diameter; thus, plaque brachytherapy is
recommended only for tumors with largest basal diameter 19 mm or less.
Round or custom plaques are most commonly used. Custom plaques (eg,
notched) are commonly used for tumors in specific locations (eg,
peripapillary). Preliminary data from a prospective study suggest that
slotted plaques provide local control of choroidal melanomas adjacent to
the optic nerve, but with a high risk of radiation optic neuropathy.
337
Plaque
brachytherapy should be performed by an experienced multidisciplinary
team including an ophthalmic oncologist, radiation oncologist, and
brachytherapy physicist.
185
Tumor localization for brachytherapy may be
performed using indirect ophthalmoscopy, transillumination, light pipe
diathermy, and/or US (intraoperative and/or preoperative).
338
MRI or CT
may be used for preoperative planning.
Dosing Recommendations for Brachytherapy
In patients receiving plaque brachytherapy for uveal melanoma, the
radiation dose at the base of the tumor (the surface of the tumor closest to
the plaque) will always be higher than the dose at the apex of the tumor
(the point of the tumor furthest from the plaque). Clinical practice varies
regarding whether the radiation dose prescribed is the dose at the base of
the tumor or the dose at the apex of the tumor.
Using iodine-125 COMS plaques, 85 Gy should be prescribed to the apex
of the tumor at low dose-rate (≥0.6 Gy/h), as this was the dose used in the
COMS study of medium-sized uveal melanomas, showing similar survival
with brachytherapy versus enucleation.
303,304
Dose adjustments may need
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-20
to be made for non-COMS plaques.
339
Other prospective and retrospective
studies using iodine-125 brachytherapy dosing similar to COMS have
reported similarly high rates of local control,
327,328,340-343
and a retrospective
study found that efficacy outcomes were similar to those in patients
treated with SRT.
344
As expected, studies using iodine-125 brachytherapy
doses higher than in COMS (eg, 100 Gy to tumor apex) also reported
recurrence rates that compared favorably with other treatment modalities
(eg, transpupillary thermotherapy [TTT], proton beam RT).
330,345
In
contrast, both retrospective and randomized prospective studies using
iodine-125 brachytherapy doses of less than or equal to 80 Gy reported
recurrence rates higher than with particle beam RT.
47,346-349
One
prospective and several retrospective studies have reported that a lower
iodine-125 brachytherapy dose was associated with local tumor
recurrence and decreased systemic control,
338,350-352
whereas other
retrospective studies have found no significant correlation between dose
and local recurrence, distant metastasis, or survival.
342,353,354
A meta-
analysis found that recurrence rates tended to decrease with increasing
iodine-125 brachytherapy dose to the tumor apex, but the effect was small
and not statistically significant.
325
Another issue of some debate is the relationship between brachytherapy
dose and complications, changes in vision after treatment, and eye
preservation. One prospective and multiple retrospective analyses have
found correlations between increasing iodine-125 dose and loss of VA,
risk of RT-related complications (eg, cataract, optic neuropathy,
glaucoma), and/or need for secondary enucleation.
334,342,351,353-358
Results
are mixed, however, and retrospective analyses did not always find
significant correlations between dose and these negative
outcomes.
356,359,360
It is important to note that for the few comparative
studies that used relatively high doses of iodine-125 brachytherapy (≥85
Gy to the tumor apex), rates of RT-related complications and vision loss
were still similar to or better than with SRT or particle-beam RT.
344,345
Prospective trials are needed to determine optimal iodine-125 dosing.
Studies using brachytherapy plaques made of other radioisotopes,
primarily ruthenium-106, but also palladium-103, strontium-90, and cobalt-
60, have tested a wide range of doses (60150 Gy to tumor apex), but
most reported mean/median dose to the apex between 80 and 130
Gy.
184,332,333,345,361-369
Only a few of these studies were prospective.
333,366
Although there is evidence to suggest that results differ between isotopes
(even when apex dose is similar),
345,362
the optimal dose has not been
determined for any of these isotopes. Results are inconsistent across
retrospective studies that attempted to evaluate the impact of dose on
local/distant failure rate,
361,367
or the impact of dose on RT-related
complications and VA loss.
184,365
Results from a few retrospective studies
using ruthenium-106 plaques suggest that while a target dose of 100 Gy to
the tumor apex may result in poorer local control than proton beam RT,
higher doses (mean 137 Gy at apex) may provide similar local control as
SRS.
345,368
Due to lack of data, the NCCN Panel recommendations are
fairly broad, suggesting that when using ruthenium-106, palladium-103,
strontium-90, cobalt-60, and cesium-131 plaques, 60 to 100 Gy may be
prescribed at low dose rate to the tumor apex; alternatively, a minimum
dose may be prescribed to the base of the tumor. The plaque margin on
the tumor border may vary for other (noniodine-125) radioisotopes.
Particle Beam Radiation Therapy
Particle beam RT includes radiation with protons, carbon ions, or helium
ions, and is a common form of definitive RT for the primary tumor.
323
Prospective studies and a systematic review found that disease-specific
survival in patients with uveal melanoma treated with particle beam RT
was similar or better than for plaque brachytherapy.
47,347,349
Compared with
brachytherapy, particle beam RT was associated with higher rates of local
control and similar or lower rates of enucleation during follow-up.
47,347,349
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-21
Across studies, local recurrence rates with charged particle therapy
ranged from 3% to 10%.
272,370
However, multivariate analysis of a real-
world database found that treatment with protons was associated with
poorer OS compared with brachytherapy treatment.
371
Decrease in VA and loss of vision can occur with particle beam RT.
370,372
Toxicities include vitreous hemorrhage, subretinal exudation in macula,
posterior subcapsular opacity, radiation keratopathy, rubeosis/neovascular
glaucoma, radiation maculopathy, and papillopathy.
370,373
NCCN Recommendations for Particle Beam Radiation Therapy
Particle beam RT should be performed by an experienced multidisciplinary
team including an ophthalmic oncologist, radiation oncologist, and particle
beam physicist.
374
In settings where the appropriate expertise is available,
particle beam therapy (proton, carbon ion, or helium ion) is appropriate as
upfront therapy after initial diagnosis, after margin-positive enucleation, or
for intraocular or orbital recurrence. It is important that the team have
experience treating uveal melanoma with the specific type of particle beam
used (proton, carbon ion, or helium ion). Particle beam RT is an option
regardless of the size of the primary lesion, and is the preferred method of
RT for tumors that are too large or too near the optic nerve to be
effectively treated with brachytherapy. Tumor localization for particle beam
RT may be performed using indirect ophthalmoscopy, transillumination,
and/or US (intraoperative and/or preoperative), MRI, and/or CT.
Dosing Recommendations for Particle Beam Radiation Therapy
Particle beam dosing for intraocular tumors is as follows: using protons,
5070 cobalt Gray equivalent (CGyE) in 4 to 5 fractions should be
prescribed to encompass the planning target volume (PTV) surrounding
the tumor;
370,374,375
using carbon ions, 60 to 85 CGyE in 5 fractions should
be prescribed to encompass the PTV surrounding the tumor.
373
Fiducial
markers (tantalum clips) are encouraged to permit eye and tumor position
verification for image-guided radiotherapy delivery. Volumetric planning in
three dimensions (with or without CT and/or MRI) is encouraged to
maximize radiation delivery to tumor and minimize radiation delivery to
organs and tissues at risk of injury from radiation.
Stereotactic Radiation
SRT includes both single-fraction and hypofractionated stereotactic
techniques, referred to collectively as SRS in these NCCN Guidelines.
Compared with brachytherapy and particle beam RT, there are fewer
prospective comparative study data on SRS for treatment of primary uveal
melanoma. Available data suggest that SRS may be as effective as other
RT modalities, but may also be associated with a higher risk of
complications. One series that compared SRS with iodine-125
brachytherapy found that rates of tumor recurrence, distant metastasis,
and secondary enucleation were similar across treatments.
344
Risk of
cataract appeared similar across treatments, but SRS appeared to be
associated with higher rates of certain complications, including
neovascular glaucoma, radiation retinopathy, and radiation
papillopathy.
344,376
Another study also reported similar rates of local control
with brachytherapy versus SRS.
368
A retrospective study comparing
ruthenium-106 brachytherapy versus SRS found a nonsignificant trend
toward increased secondary glaucoma after SRS.
364
A retrospective study
comparing SRS versus proton beam RT reported similar rates of local
control and eye retention across treatments, but higher rates of VA decline
with SRS.
377
Studies using SRS as primary treatment for uveal melanomas have
reported local failure rates ranging between 2% to 16%.
183,272,344,368,377-391
Rates of 5-year metastasis-free survival after SRS ranged from 69% to
84%.
344,379,381,392
OS 5 years after SRS has been reported to be 55% to
98%.
380,381,385,392,393
These large ranges likely reflect differences in the
populations studied; thus, in the absence of randomized trial data it is
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-22
difficult to know whether these outcomes are better or worse than those
reported for other treatment modalities.
In studies testing SRS as primary treatment for uveal melanoma, eye
retention rates ranged from 73% to 98%.
377,380,383-387,390,392-395
In addition to
causing a decrease in VA, complications associated with SRS include
cataracts, neovascular glaucoma, radiation retinopathy, radiation
papillopathy, radiation maculopathy, hemorrhage, macular edema, optic
neuropathy, and keratitis sicca (dry eye).
344,376,379,380,382,383,387-389,395-399
Some studies have linked the rate and/or severity of complications to the
radiation dose, tumor location, tumor size, and VA before
treatment.
382,384,387,397,400,401
NCCN Recommendations for Stereotactic Radiation
Due to the lack of randomized prospective data (compared with other RT
techniques described above), SRS is the least often used form of definitive
RT for the treatment of primary or recurrent intraocular tumors. Like
particle beam RT, SRS can be used to treat large choroidal melanomas.
The choice between these two options generally depends on the radiation
oncology facilities available. In rare cases when both particle beam RT
and SRS facilities are available, some NCCN Panel members prefer
particle beam RT because there are more supporting data for this
approach. Tumor localization, fiducial marker use, and planning for SRS
are generally consistent with particle beam RT approaches. Using
fractionated SRS, 45 to 70 Gy in 2 to 5 fractions should be prescribed.
Using single-fraction SRS, 18 to 45 Gy in 1 fraction should be prescribed.
RT Toxicity (Ocular)
In order to avoid secondary enucleation, a variety of methods for
preventing or managing RT-associated complications have been tested in
prospective studies. Toxicity management methods tested include
panretinal photocoagulation for ocular ischemia,
402
transscleral local
resection for exudative retinal detachment,
403
and intravitreal anti-vascular
endothelial growth factor (VEGF) (eg, bevacizumab, ranibizumab,
aflibercept) or intravitreal corticosteroids (eg, triamcinolone,
dexamethasone) for treating optic neuropathy, macular radiation
vasculopathy, or papillopathy or macular edema.
404-411
These intravitreal
therapies have also been tested for prophylaxis.
412-414
The NCCN
Guidelines for Melanoma: Uveal do not currently have recommendations
for management of RT side effects.
Other Ablative Techniques
Laser Therapy
Laser Photocoagulation
Laser photocoagulation has also been used for treatment of primary or
recurrent uveal melanomas, sometimes as monotherapy but more often as
an adjunct to RT or surgery.
45,46,171,277,281,415-430
The sparse data available
suggest that laser photocoagulation is associated with high rates of
recurrence if used as the sole primary therapy,
415,421
but that when used as
a supplement to brachytherapy can increase rate of tumor regression.
426
Transpupillary Thermotherapy
TTT, also called diode laser hyperthermia, is a technique that uses a
modified infrared diode laser to slowly heat up a specified area to
approximately 45 to 60°C. TTT can be used to treat a large spot and has
deep tissue penetration. TTT has been tested in prospective studies as
the sole primary treatment for uveal melanoma tumors, but local
recurrence rates varied widely across studies, with some studies reporting
high rates of recurrence (>50%), even for small tumors.
272,330,431-436
TTT
has also been tested as an adjunct therapy to radiation (brachytherapy,
proton beam RT, SRS or fractionated SRT) to reduce the risk of local
recurrence.
350,366,437-443
One prospective randomized trial found that TTT
administered at 1, 6, and 12 months after proton RT reduced the likelihood
of retinal detachment and lowered the secondary enucleation rate.
438
A
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-23
retrospective study (n = 133) on matched groups found that addition of
TTT to brachytherapy improved rate of tumor regression, 5-year tumor
recurrence rate, eye-globe preservation, and recurrence-free survival.
444
There was no impact on metastasis-free survival, OS, rate of
complications, or visual outcomes.
444
However, two other studies showed
that adding TTT to brachytherapy did not improve the rate of globe
conservation, and was associated with greater loss of VA.
366,445
The larger
of these studies (n = 449) also showed that rate of local failure, distant
metastasis, and cause-specific death were not improved by TTT.
366
Complications associated with TTT include retinal hemorrhage, vitreous
hemorrhage, retinal vascular occlusions, optic disc atrophy, macular
edema, retinal detachment, retinal traction, exudative serous
neurosensory detachment, vitritis, and postoperative pain.
433,446
Cryotherapy
Use of cryotherapy for treatment of primary or recurrent uveal melanomas,
either alone or in conjunction with other therapies, has been described in
case reports and case series in the literature,
435,447-453
and retrospective
reports and review articles suggest that this method is occasionally being
used in clinical practice.
26,310,454
Treatment for Extraocular/Extrascleral Extension
Extraocular/extrascleral extension has been reported to be present in
approximately 3% of patients at diagnosis of uveal melanoma,
166,173,200,455
is more common among tumors with higher T stage (12% of T4 tumors),
285
and is associated with poor prognosis.
18,26,284,456-458
Extrascleral/extraocular
extension can be detected by preoperative imaging, or found or confirmed
at the time of enucleation.
294,459
Sometimes the evidence of extraocular
extension is microscopically positive or close margins after enucleation,
without clinical, intraoperative, or radiographic evidence of gross residual
disease to the orbit. In other cases, extraocular tumor is visible
intraoperatively or intraoperative findings suggest that there may be gross
disease to the orbit. In the COMS trial of patients with large uveal
melanomas that tested enucleation with versus without pre-enucleation
RT, unexpected extrascleral extension was found in 2% of patients who
underwent enucleation, despite extensive clinical and imaging
workup.
294,301
Orbital Exenteration
Orbital exenteration is surgical removal of the globe and adjacent orbital
contents, for cases with extraocular extension and/or orbital
invasion.
41,185,196,422,425,460
Retrospective studies of large databases suggest
that exenteration is used in less than 1% of patients,
26
and among patients
undergoing enucleation, 2.5% need orbital exteneration.
461
The value of
orbital exenteration is disputed in the literature, largely based on low-
quality data such as case reports and retrospective studies. Some studies
support orbital exenteration because it provided superior outcomes
compared with other (nonsurgical) approaches,
462
whereas others report
poor outcomes after orbital exenteration, arguing that it may not be
justified.
463,464
Radiation Therapy for Extraocular/Extrascleral Extension
A retrospective study (n = 202) found that in patients treated with
enucleation, postoperative RT improved survival, particularly in young
patients (age <30 years) and those with choroidal tumor height greater
than 3 mm.
461
Another retrospective study (n = 17) reported a local
recurrence rate of 6% in patients with extrascleral extension who were
treated with enucleation followed by adjuvant external beam RT to the
orbit.
465
For patients with limited extraocular extension less than 3 mm
thick, brachytherapy may also be an option, as a retrospective study (n =
17) showed no intraocular or extraocular tumor relapse after a median
follow-up of 63 months (range, 23164 months).
466
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-24
Treatment of Localized Uveal Melanoma
Following workup and staging, patients with localized uveal melanoma
should be treated. Treatment options depend on the tumor size (diameter
and thickness) and proximity to the optic nerve.
Tumor Size: Largest Diameter 5–19 mm and Thickness <2.5 mm
For thin tumors (<2.5 mm) with largest diameter ranging from 5 to 19 mm,
the recommended primary treatment options are plaque brachytherapy or
particle beam RT. For highly select patients who are not good candidates
for brachytherapy or particle beam RT, other options to consider include
laser therapy or enucleation.
Tumor Size: Largest Diameter ≤19 mm and Thickness 2.510 mm
Brachytherapy and particle beam RT are also options for treating tumors
with largest diameter 19 mm or smaller and thickness 2.5 mm to 10 mm.
If there is concern that adequate response was not achieved from initial
RT, then further treatment should be considered. Recommended options
for further treatment include laser therapy or cryotherapy. In highly select
cases, resection is sometimes considered. Tumors in this size range may
also be treated with enucleation. Although there is a trend toward avoiding
enucleation, it is recommended for patients with neovascular glaucoma,
tumor replacing greater than 50% of globe, or blind, painful eyes.
Enucleation should also be considered in cases of extensive extraocular
extension.
Tumors Not Appropriate for Brachytherapy
Given the limitations in the size and RT penetrance of commercially
available brachytherapy plaques (diameter ≤23 mm), this method is not
appropriate (and not recommended) for tumors that are too large in
diameter (>19 mm; any thickness), too thick (>10 mm; any diameter), or
have optic nerve involvement and thickness (>8 mm; any diameter). RT
options for such tumors include particle beam RT and SRS. The choice
between these two RT modalities usually depends on which modality is
available at the treating institution. In the rare scenario that the institution
has both SRS and particle beam facilities, some practitioners would opt for
particle beam because there are more data supporting its efficacy.
Enucleation is also a recommended option, especially in cases with
extensive extraocular extension, neovascular glaucoma, tumor replacing
greater than 50% of globe, or blind, painful eyes.
Additional Treatment Considerations
An essential feature of high-quality care is that clinical decisions are
informed by a variety of case-specific factors (patient preferences and
characteristics such as age, status of the other eye, disease
characteristics, and medical history), such that for some patients the best
clinical approach may not be one of the listed options. The recommended
treatment options are largely based on data from choroidal melanomas.
For small ciliary body and iris tumors (<3 clock hours), surgical excision
may be considered.
Additional Primary Treatment for Extraocular Extension
For patients with limited extraocular extension, brachytherapy with scleral
patch graft should be considered. For patients treated with enucleation for
their primary tumor, additional treatment may be needed if extraocular
extension is present. For patients with microscopically positive or close
margins after enucleation, but no clinical, intraoperative, or radiographic
evidence of gross residual disease to the orbit, recommended options
include observation (no further treatment), mapping biopsy, and/or
consideration of RT to the orbit (using particle beam or photon beam
therapy). For patients with visible extraocular tumor or suspicion of gross
disease in the orbit at the time of enucleation, biopsy of the extraocular
tissue is recommended, if possible. Additional treatment options to
consider include one or more of the following: intraoperative cryotherapy,
orbital exenteration, and/or RT to orbit using particle beam or photon
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-25
beam RT. For photon or proton beam RT to the orbit (after enucleation), a
dose of 20 to 30 Gy in 5 fractions should be prescribed to the clinical
target volume at risk for recurrence
294,467
using intensity-modulated or
conformal techniques with image guidance.
Treatment of the Primary Tumor in Patients with Metastatic Disease
Palliative local therapy to the primary tumor may be considered in the
setting of metastatic disease. In general, if the metastatic disease is being
treated, the primary tumor should also be treated. Patients who present
with advanced metastatic disease and limited life expectancy may elect to
have no treatment to their primary tumor.
Follow-up
In order to make educated choices about monitoring and follow-up after
treatment of the primary lesion, it is important for patients and treating
clinicians to be aware of what is known about the typical trajectory of the
disease, including the typical characteristics and time frame until
presentation of treatment complications, recurrence, and metastasis.
Monitoring should also be informed by consideration of a patient’s risk for
each of these outcomes.
For patients whose primary uveal melanoma was treated with RT or
surgery, the subsequent disease-free interval is highly variable, ranging
from a few months to many years.
327,347,372,388,440,441,468
Uveal melanoma is
characterized by early micrometastasis (often before treatment) followed
by variable latency period before emergence of overt metastasis.
469
Local
recurrence is rare, occurring in less than 10% of patients after the primary
lesion is treated using one of the modalities recommended in these
guidelines.
47,187,272,294,295,326,327,330,331,333,337,338,347,366,439,441,468,470,471
Development of detectable distant metastatic disease is much more
common than local recurrence, and develops in up to 70% of patients,
depending on the stage and other risk factors at the time of
diagnosis.
8,13,26,285
For example, large retrospective studies (n > 7000)
found that after long-term follow-up the percent of patients who had
developed metastasis was 20% for those with stage I at diagnosis, but
~70% for those with stage III at diagnosis.
13,285
Patterns of Local Recurrence
Due to the rarity of local recurrence, data characterizing local recurrences
are somewhat limited. The likelihood, typical time frame for development,
location, and risk factors for local recurrence may depend on whether the
patient received primary treatment with enucleation or some form of
radiation (ie, brachytherapy, particle beam, SRT).
Local Recurrence After Enucleation
Local recurrence after enucleation for localized primary melanoma is
extremely rare, occurring in only ~1% of patients, and presents as tumor
growth in the orbit.
294,295,471
In the COMS randomized controlled trials (for
medium and large tumors), the few patients who developed local
recurrences after enucleation all had distant metastases before the local
recurrence developed.
294
Retrospective studies reported similar findings,
suggesting that development of distant metastasis is due to
micrometastasis that developed prior to enucleation.
471
The rarity of the
event precludes meaningful evaluation of risk factors for local recurrence
after enucleation.
Local Recurrence After Radiation (Brachytherapy, Particle Beam
RT, or Stereotactic RT)
In prospective trials reporting on patients who received brachytherapy as
primary treatment for localized uveal melanoma, tumor regression was
observed in most cases (97%–98%),
330,367
and occurred over the first 2
years after treatment.
367
Prospective studies have reported local failure
rates ranging from 0% to ~20% for patients with localized primary uveal
melanoma treated with brachytherapy with iodine-125 or other types of
plaques (ruthenium-106, palladium-103, cesium-
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-26
131).
47,187,326,327,330,331,333,337,338,347,366,439,441,468,470
Most of these studies
reported local failure rates less than 10%, and a systematic review that
included more than 3000 patients from 22 studies, including retrospective
studies, found that the weighted mean rate of local failure after
brachytherapy was 9.45%.
272
Prospective studies have reported that local
recurrence after brachytherapy, although rare, occurs over a wide time
range.
47,187,327,366,440,441,468
Whereas some studies report median time to
local recurrence between 2 and 4 years,
327,440,441,468
most also show curves
that never really plateau, due to late recurrences developing throughout
long-term follow-up, as late as 12 years or more after
treatment.
47,187,327,366,440,441,468
A variety of risk factors for local recurrence have been identified. Although
results vary across studies, multiple analyses have found that tumor size
and location are associated with risk of local recurrence after
brachytherapy.
47,187,347,366,441,468
Regarding primary tumor location as a risk
factor, prospective studies have found higher risk of local recurrence
associated with ciliary body involvement, epicenter location in the macula,
proximity to the foveal avascular zone, proximity to the optic nerve, and
extension under the foveola.
47,187,347,441,468
One large prospective study
reported that presence of visual symptoms at the time of uveal melanoma
diagnosis was correlated with higher risk of local recurrence after
brachytherapy.
327
Several studies have shown that local recurrence is at
least mildly associated with a higher risk of melanoma-related death.
187,327
For patients with primary uveal melanoma treated with particle beam RT,
local recurrence rates ranged from 3% to 10% across studies.
272,370
Tumor
regression following particle beam RT can begin within 6 months of
treatment, and tumor shrinkage may continue to occur throughout 5 years
of follow-up.
370,472
As with brachytherapy, local recurrence after particle
beam RT as treatment for primary localized uveal melanoma occurs over
a long time range, as early as 2 months after treatment and as late as 12
years.
372,416,473
Prospective studies have found that most local recurrences
occurred in the first 4 to 5 years, with median time to recurrence of less
than 2 years.
47,372,416,473
Primary tumor size and ciliary body involvement
were shown to be independent risk factors for local recurrence in a
multivariate analysis of results from a large prospective observational
study of patients who received particle beam RT as primary treatment for
uveal melanoma.
473
Local recurrence after particle beam RT has been
shown to be associated with an increased risk of metastasis and
increased risk of death from metastatic uveal melanoma.
372,473
Studies using SRT as primary treatment for uveal melanoma have
reported local failure rates ranging between 2% to 16%.
183,272,344,368,377-390
This large range likely reflects differences in the populations studied.
Following SRT of the primary tumor, prospective studies found that many
uveal melanomas showed a transient increase in tumor height, volume, or
both.
383
Responses to SRT first began to appear 6 months after treatment,
with progressive decreases in tumor height and volume continuing for at
least 3 years.
183,379,384,388,389
Prospective follow-up showed that the fraction
of patients with response or stable disease increased during the first year
after treatment.
183,389
Prospective studies have found that following SRT
there is a small percentage of patients with persistent tumor growth, which
can occur soon after treatment (failure to achieve local control) or after a
period of local control, and some studies reported recurrences many years
after treatment.
381,383,387-390,396
The few prospective studies that have
attempted to identify risk factors for tumor growth after SRT have not
found any correlation between RT dose or tumor diameter before
treatment.
379,388
There are some prospective data that suggest that local
recurrence after SRT may be correlated with poorer survival.
380
For all of the above RT modalities (brachytherapy, proton beam RT, and
SRT), prospective studies have reported that tumor growth after treatment
occurred at the margins of the treated area for some patients, but for other
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-27
patients appeared as tumor growth in all dimensions, including growth
within the treated volume.
347,370,373,383,389,416,468,473
Patterns of Treatment Complications
Complications After Enucleation
For patients treated with enucleation for primary uveal melanoma,
complications between 1 to 6 weeks after surgery included pain requiring
longer hospital stay, pain requiring medication, conjunctival wound
dehiscence, infection, decreased facial sensation, eyelid swelling,
inflammation, implant displacement, hair loss, ptosis, conjunctival
chemosis, ecchymosis, and orbital or conjunctival hemorrhage.
294
Long-
term follow-up showed that other problems include poor motility of
prosthesis, poor alignment of prosthesis, severe ptosis, and displacement
of implant.
294
Enucleation is also associated with loss of certain visual
functions, such as peripheral vision, night driving, and judging
distances.
316,317,319
Complications After Brachytherapy
Effects on Visual Acuity
Prospective studies of patients treated with brachytherapy for uveal
melanoma have shown that VA in the treated eye tends to steadily
decrease during at least the first 5 years of follow-up, both in terms of
severity and the percent of patients with poor vision.
186,331,334,335,347,366,468
Studies with long-term follow-up (>10 years) showed that the percent of
patients with poor VA increases more slowly between 10 and 15 years
after brachytherapy, and one study reported a median time to VA score
(VAS) less than or equal to 50 of 39 months.
334,468
Factors associated with
loss of VA or poor vision outcomes included greater baseline tumor apical
height, shorter distance between the tumor and the foveal avascular zone,
ciliary body involvement, presence of tumor-associated retinal
detachment, nondome-shaped tumor, and patient history of diabetes.
Results from multiple prospective studies suggest that risk of poor vision
outcomes depends on baseline tumor thickness and
location.
186,331,334,335,347,366,468
Locational elements suggested to be
associated with increased risk of poor vision include juxtapapillary
location, proximity to foveola, optic nerve or optic disc, ciliary involvement,
central tumor location, mid-choroid and macula location, and retinal
invasion. Some prospective studies have suggested additional risk factors
for poor vision outcomes, including baseline VA, patient age, diabetes,
baseline tumor shape, retinal detachment at baseline, and development of
radiation maculopathy or radiation optic neuropathy.
186,335,366,468
Cataracts
Prospective studies have reported that following treatment with
brachytherapy for uveal melanoma, the percent of patients with cataracts
in the treated eye steadily increases over the first 5 years of follow-up.
Cataracts may affect more than two thirds of patients by 5 years, although
a much smaller number of patients underwent cataract surgery.
329,468
In
the COMS trial, median time to development of cataract was 2.5 years,
and the median time to cataract surgery was 3.5 years.
329
Cataract
surgery resulted in VA improving by two or more lines in 66% of patients,
and stabilizing in 26%.
329
Older age, larger baseline tumor size, and higher
radiation dose may be risk factors for development of cataract after
brachytherapy.
329
Other Serious Complications
Treatment of uveal melanoma with brachytherapy can result in a variety of
radiation-related complications in the treated eye, or can worsen
conditions initially caused by the uveal tumor.
336,441,468
Examples of more
serious complications include radiation retinopathy, optic neuropathy,
papillopathy, maculopathy, neovascular glaucoma, retinal detachment,
and various types of hemorrhages and vascular abnormalities.
331,336,468
Prospective studies have found that the development or worsening of
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-28
complications typically occurs during the first 5 years after treatment,
although the time frame for development may differ slightly between
specific types of complications.
336,468
These complications occasionally
develop more than 5 years after treatment, especially in patients who had
large tumors prior to treatment.
468
Enucleation
Prospective studies of patients treated with brachytherapy for uveal
melanoma have found that up to 40% of patients needed enucleation
during follow-up, either due to treatment failure, loss of VA, or
complications.
47,186,187,303,366,468
The rate of enucleation after brachytherapy
was lower in some studies that included only patients with smaller tumors
at baseline.
187,366
In these studies the cumulative rate of enucleation
increased gradually over a long period of time, at least 15 years after
follow-up.
47,186,187,437,468
The need for enucleation at late time points reflects
that local recurrences can occur many years after
treatment,
47,187,327,366,440,441,468
that VA can persistently decline over many
years,
186,331,334,335,347,366,468
and that some complications have a long time to
onset and/or gradually worsen over many years.
329,336,468
Studies with
long-term follow-up reported that approximately half of the enucleations
were due to local treatment failure, and the other half were due to
complications or poor VA.
47,187,468
Complications that lead to secondary
enucleation included neovascular glaucoma, retinal detachment, vitreous
hemorrhage, and ocular pain.
187,468
Results from the COMS study in
medium-sized tumors suggest that enucleations at early time points were
more often caused by local treatment failure, whereas enucleations at later
time points were more often caused by complications or poor VA.
187
Factors that may be associated with increased risk of enucleation include
the following baseline features (prior to primary treatment): increased
primary tumor thickness, proximity to foveal avascular zone, anterior
location, and epithelioid cell type; poorer VA; and younger age.
187,366,468
Complications After Particle Beam RT or Stereotactic RT
Effects on Visual Acuity
Prospective studies of patients with uveal melanoma have reported
steadily declining VA in the treated eye following proton RT or SRT, both
in terms of the percent of patients with poor VA and in terms of the mean
VA across the whole patient population.
183,347,379-381,383,384,387,388,473
This
decline was observed throughout the duration of follow-up, which was up
to 5 years in these studies. Some of these studies suggest increased risk
of declining VA after proton RT or SRT for patients with low baseline VA,
increased radiation dose to optic nerve, and posterior tumor location.
379,380
Time to loss of VA may be longer for patients treated with lower doses.
388
Other Complications
Following particle beam RT for primary uveal melanoma, toxicities
reported in prospective studies include vitreous hemorrhage, subretinal
exudation in macula, posterior subcapsular opacity, radiation keratopathy,
rubeosis/neovascular glaucoma, retinal detachment, radiation
maculopathy, and papillopathy.
370,373,438
Eyelash loss and low-grade
dermatitis are common short-term toxicities that develop soon after
treatment.
347,474,475
Whereas development of retinal detachment or
neovascular glaucoma rarely occurs beyond 2 to 4 years after
treatment,
373,438
development of RT maculopathy and RT papillopathy may
occur beyond 5 years of follow-up.
473
Results from prospective studies
suggest that risk of RT maculopathy may depend on the distance between
the tumor and the macula, whereas RT papillopathy and neovascular
glaucoma may be related to proximity to the optic disc.
373,473
Increased
tumor size and larger irradiated volumes of critical normal eye structures
may also be associated with increased risk of neovascular glaucoma.
373
Prospective studies have reported a wide variety of complications
associated with SRT treatment of primary uveal melanoma. Alopecia and
eyelash loss typically develop within 1 year of treatment, and have been
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-29
reported to occur in less than 10% of patients, although much higher rates
were observed in patients with large primary tumors.
183,384,387,399
Doses
high enough to cause alopecia can also cause dermatitis, another early,
transient, mild toxicity associated with SRT.
387,399
Fatigue, pain, and dry
eye may also occur after SRT.
183,379,380,387
Fatigue and pain typically occur
soon after treatment, and then abate.
183
Dry eye also typically occurs soon
after treatment (median time to onset 6 months), but can occur much later
(range 360 months), and can be severe and/or persistent.
183,379,387
Baseline tumor size (base diameter and height) and higher dose to the
lacrimal gland may increase risk of dry eye.
379,387
Uveitis has been
reported to develop between 6 to 52 months after SRT, occurring in less
than 15% of patients, although larger tumor volume may increase the risk
of uveitis.
379,384,387,399
Diseases of the lacrimal drainage system tend to
have late onset, occurring between 1 to 4 years after treatment, and
usually occur in less than 10% of patients.
387,399
Mild iritis has been
reported after SRT for tumors with ciliary body involvement.
383
Conjunctival problems (such as hyperemia, irritation, tears, and chemosis)
are typically mild, mostly occur within 6 months of treatment, and usually
resolve.
183
Corneal epithelial defect and corneal ulcer have been reported in up to
30% and up to 10% of patients, respectively, and can develop between 6
months and 5 years after SRT treatment for uveal melanoma.
384,387,389,399
These corneal complications can be serious, and some studies suggest
that risk of corneal damage may increase with increasing dose and
increasing tumor diameter.
382,384,399
SRT can cause or worsen exudative retinal detachment, but some cases
of retinal detachment prior to treatment were unchanged or resolved after
treatment.
183,379,383,384,387
One study reported resolution of over half of the
cases with retinal detachment at baseline, with a median time to resolution
of 15.7 months.
387
Increased patient age and tumor size may be
associated with increased risk of retinal detachment.
379,384
Various types of
hemorrhage, including vitreous hemorrhage, subretinal bleeding, retinal
hemorrhage, cutaneous bleeding, and subconjunctival hemorrhage, have
been reported following SRT.
183,379,380,384
Most of these bleeding events are
mild and resolve.
380,384
Vitreous hemorrhage has been reported in 10% or
less of patients, can be more severe, and may develop between 3 to 48
months of follow-up, with a median time to development of 15
months.
379,380,384
SRT can also cause or worsen cataracts, which can be severe and may
require surgery.
183,379,380,382-384,387,389,399
Cataract development after
treatment occurs over a wide time range (3100 months), with one study
reporting a mean time to development of 19.6 months, and another
reporting a median time to development of 12 months.
183,379,387,389,399
Prospective studies suggest that risk factors for cataract development
after SRT may include advanced age, ciliary body involvement, larger
tumor size, larger PTV, larger RT dose, and larger dose to the lens and
ciliary body.
379,382,384,399
Secondary glaucoma, including neovascular glaucoma, has been
observed in multiple prospective studies of patients treated with
SRT.
183,379,380,382,384,387,389,396,399
Neovascular glaucoma may be mild or
severe, and may effect more than 25% of patients.
379,380,384,387,399
Neovascular glaucoma typically has late onset, developing between 5
months and 10 years after treatment, with one study reporting median time
to development of 30 months.
379,380,387,389,396,399
Results from prospective
studies agree that greater tumor height (at baseline) is a risk factor for
neovascular glaucoma, but differ regarding whether greater RT dose is
associated with greater risk.
379,382,384,387,399
Retinopathy has been reported to occur in up to two thirds of patients
treated with SRT, and may be mild or severe.
183,379,380,383,384,387,389,399
Retinopathy typically has delayed onset, and has been reported to
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-30
develop between 5 to 110 months, with one prospective study reporting a
mean time to development of 15 months, and another reporting a median
time to development of 24 months.
183,379,380,383,387,389
Development of optic
neuropathy after SRT has been observed in multiple prospective studies,
may occur in up to 40% of patients, and can be
severe.
183,379,380,382,384,387,389,399
Optic neuropathy typically has delayed
onset, and has been reported to develop between 4 months and 9 years
after treatment, with a mean time to development of 21
months.
183,380,384,387,389,399
Prospective studies have suggested a variety of
risk factors for optic neuropathy, including larger tumor size, tumor stance
to the optic disc, higher total RT dose, higher dose to the optic nerve, the
ratio of gross tumor volume (GTV) to PTV, or the distance from PTV to the
optic nerve.
379,384,387,399
SRT can also cause optic disc edema, which is
often severe, develops between 1 to 2 years after treatment, and has a
median time to development of 18 months.
379
Enucleation
Prospective studies have found that secondary enucleation was
sometimes needed due to local recurrence or complications in patients
with primary uveal melanoma treated with proton RT or
SRT.
47,373,379,380,384,387,388,396
Among cases of enucleation due to
complications, neovascular glaucoma was the most common cause;
others included angle closure glaucoma, tumor necrosis syndrome,
corneal ulcer, retinal detachment, and pain, sometimes due to elevated
intraocular pressure.
47,373,380,384,387,388,396
These secondary enucleations
occurred over a wide time range, from 2 months to greater than 10 years
after treatment.
47,373,384,389,438,476
Although most enucleations occurred
within 5 years of treatment, the risk of enucleation between 5 to 10 years
after treatment is non-negligible.
47,388,389,396,438,476
Larger tumor size,
proximity to the optic disc, high intraocular pressure, or retinal detachment
before treatment may increase the risk of secondary enucleation after
proton RT.
476
Surveillance Methods for Local Recurrence or Complications
To monitor for local recurrence and possible complications after treatment
of the primary tumor in patients with uveal melanoma, prospective studies
have followed patients with regular clinical and ophthalmologic exams.
For patients treated with enucleation in the COMS trial, there were follow-
up exams at 1 to 2 weeks after surgery to assess healing status, and at 6
months, 12 months, and annually thereafter, in which the eye socket and
eyelids were examined for possible recurrence or complications, and the
fit of prosthesis checked.
301,303,477
For patients treated with RT (brachytherapy, particle beam RT, or SRT) in
prospective studies, follow-up exams typically included complete
ophthalmologic exam of the treated eye, with indirect ophthalmoscopy, slit-
lamp exam, tonometry, color fundus photography, A-scan and B-scan US,
and measurement of VA and visual
field.
47,183,186,187,327,330,331,333,335,347,366,367,370,380,383,384,387-
389,396,437,439,440,468,472,473,477-479
Many prospective studies also included
fluorescein angiography in follow-up exams, either at regular intervals or
as needed.
47,187,330,333,335,336,347,366,384,388,389,439,478
A few prospective studies
used gonioscopy,
187,388
OCT,
335,366,437
or MRI.
383,396
For most prospective
studies following patients with primary uveal melanoma treated with RT,
data taken at regular intervals included VA, intraocular pressure, tumor
dimensions and shape, development of new extrascleral growth, orbital or
ciliary body mass, and retinal invasion, pathologic changes, changes in
tumor appearance, and patient symptoms (ie, ocular pain, vision
problems).
183,186,187,303,331,333,335,367,370,373,380,384,387-389,440,472,479
Some studies
monitored for changes in tumor reflectivity or tumor vascularity as signs of
regrowth/recurrence.
366,367,396,472
The frequency of follow-up exams after RT (brachytherapy, particle beam
RT, or SRT) varied across studies. For patients who were treated with
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-31
brachytherapy in the COMS study, follow-up included an exam within 6
weeks of surgery, at 6 months after treatment, then every 6 months for 5
years, and every 12 months thereafter.
303
Patients with suspected tumor
growth had more frequent follow-up exams. Whereas some prospective
studies in patients treated with brachytherapy had follow-up protocols
similar to COMS,
327,331,367
others used more frequent follow-up, with more
than 2 exams per year during the first year or first few years after
treatment, and exams every 6 to 12 months during later
years.
47,330,333,334,347,439-441,468,479
Prospective studies following patients after
particle beam RT or SRT for primary uveal melanoma sometimes had
frequent follow-up during the first 6 months after treatment (eg, 35 exams
in the first 6 months),
183,375,379,380,383,384
and most had follow-up exams at
least every 3 to 4 months during the first year or two.
47,183,347,373,379,384,387-389
A few studies switched to the 6-month follow-up interval starting early (<1
year) from treatment.
370,375,380,396,472,478
Long-term follow-up intervals
ranged between 4 to 12 months.
47,183,347,370,373,375,379,380,383,384,387,388,396
Because some patients treated with RT may have an increase in tumor
size before regression, studies have defined local recurrence in terms of
specific thresholds for growth. For example, the COMS trial defined local
treatment failure (after brachytherapy) as one or more of the following:
increase in height of ≥15% by echography or a ≥250-um expansion of any
tumor boundary by photographs or clinical examination followed by an
additional ≥15% increase in elevation or a further ≥250-um expansion of
any tumor boundary observed and confirmed on subsequent examination;
extrascleral extension based on clinical or echographic findings; or
development of orbital mass, ciliary body mass, or retinal invasion.
187
Similarly, prospective studies following patients after SRT have used
definitions of tumor recurrence that include tumor growth rate above a
specific threshold that is confirmed over two exam intervals, usually based
on US measurements.
183,373,387,388,396
Follow-up for the Treated Eye
At NCCN Member Institutions, standard follow-up in the affected eye
includes imaging with color fundus photography and ultrasonography
every 3 to 6 months for 3 to 5 years, then every 6 to 12 months thereafter,
if stable. The frequency of follow-up should depend on the size and
location (eg, juxtapapillary location, ciliary body involvement) of the tumor
at presentation, as these factors impact the risk for recurrence.
Risk in Contralateral (Fellow) Eye
In patients who have received treatment for primary unilateral uveal
melanoma, disease can develop in the contralateral eye, but the incidence
is very low. In the COMS trials of patients with medium or large primary
uveal melanoma tumors, prospective monitoring showed that less than 1%
of patients developed disease in the contralateral eye during follow-up
after primary treatment.
295,480,481
Moreover, for the majority of patients who
did not develop disease in the contralateral eye, results from regular
ophthalmologic exams showed that good VA was retained in their fellow
eye throughout the 10 years of follow-up, regardless of the modality used
to treat the primary lesion (brachytherapy, enucleation, or RT followed by
enucleation).
482
Analysis of 8165 patients with ocular melanoma in the
SEER database found bilateral involvement in 0.1% of patients.
10
One
retrospective study of 52 cases of bilateral uveal melanoma suggests
prognosis in these cases is similar to that of unilateral cases.
483
Additional
cases of uveal melanoma metastasizing to the contralateral eye are
described in case studies.
484-492
NCCN Recommendations for Follow-up for the Contralateral Eye
All patients should receive follow-up for the affected eye. In patients with
uveal melanoma, the contralateral eye is not at increased risk of uveal
melanoma,
10,295,480,481
and can be followed with routine ophthalmologic
care.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-32
Patterns of Metastases
Most uveal melanomas are localized at first presentation, and only a small
percentage of cases (<3%) have detectable metastatic disease at the time
of diagnosis.
6,10-14,166,493
Development of distant metastatic disease is much
more common, occurring in ~20% to 70% of patients within 20 years after
treatment for primary uveal melanoma, depending on stage/size and
genetic characteristics of the tumor at diagnosis.
8,13,26,48,67,285
Prospective
studies with long-term follow-up (≥5 years) after treatment of primary uveal
melanoma have shown that the cumulative rate of development of distant
metastatic disease steadily increases over many years of follow-
up.
47,331,347,366,372,379,381,387,388,440,441,468,473,480,481
For example, in the COMS
studies in which patients with medium tumors were treated with
brachytherapy or enucleation, and patients with large tumors were treated
with enucleation with or without RT prior to surgery, the Kaplan-Meier
estimates of 2-, 5-, and 10-year metastasis rates were 10%, 25%, and
34%.
481
Although incidence rates declined after the 2-year examination,
and most cases of distant metastatic disease developed within 90 months
of treatment, new cases of distant metastatic disease were detected at
every 6-month interval over the 12-year follow-up period.
480
Although the
proportion of patients who developed distant metastasis differed
depending on the primary tumor size prior to treatment, the cumulative
incidence curves (proportion of patients with metastasis) for both medium
and large tumors did not appear to be plateauing even after 10 years of
follow-up.
480,481
Similar findings have been reported for other prospective
studies monitoring for development of distant metastatic disease after
primary treatment with brachytherapy,
331,366,440,441,468
particle beam
RT,
47,347,372
or SRT.
384,387,388
These studies found that the proportion of
patients with metastasis increased between all consecutive time points
(eg, Kaplan-Meier estimates at 1, 2, 3, 5, 7, 8, 10, and 15
years).
47,331,372,384,387,388,440,441,468
In these studies the development of
metastatic disease occurred as early as 4 months and as late as 14 years
after treatment of primary uveal melanoma,
347,372,381,468
developing
continuously throughout the follow-up period such that cumulative
incidence curves and disease-free survival curves did not appear to be
plateauing even after 6 to 20 years of follow-up.
47,366,388,440,468
Although
difficult to calculate due to ongoing development of distant metastatic
disease, several studies reported mean and median times to first distant
metastasis (for those who developed distant metastasis), which ranged
from 39 to 45 months, and from 35 to 37 months, respectively.
347,372,388,468
Sites of Metastasis
Uveal melanoma most often metastasizes to the liver.
19,21,22,166,295,480,481,494-
501
Of those with distant metastasis, greater than 90% have liver
metastases.
295,480,481
For patients with only one metastasis at the time that
distant metastasis is first detected, most have liver metastasis.
19,21,22,295,497
Other common sites of metastasis, listed in order of decreasing
prevalence, are lung, bone, skin/soft tissue, and lymph
nodes.
11,21,166,295,373,379,480,481
In large prospective studies following patients
after treatment for primary uveal melanoma, metastasis to the lung was
observed in 20% to 30% of patients who developed distant metastasis, to
the bone in 16% to 18%, to skin/soft tissue in 11% to 12%, and to lymph
nodes in 10% to 11%.
295,480
Retrospective studies show similar trends.
21,501
A few studies reported brain metastases in 4% to 5% of patients who
developed distant metastases.
21,295,480
Most patients who develop
metastasis ultimately have multiple sites involved.
295,480
Risk Factors for Metastasis
Numerous studies have evaluated factors prognostic for development of
distant metastasis or for shorter time to development of distant metastasis.
Although the AJCC staging system is based on survival data from large
epidemiologic studies,
12,13,18
characteristics used for AJCC staging have
also been shown to be prognostic for development of distant metastasis.
In the AJCC staging system for melanomas arising in the choroid or ciliary
body, T stage is based on the largest basal diameter and thickness of the
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-33
primary tumor, as well as the presence or absence of ciliary body
involvement and presence and size of extraocular extension.
31
Multiple
prospective studies and several large retrospective studies (N > 1000)
have shown by multivariable analysis that primary tumor diameter and/or
thickness is associated with risk of metastasis after primary
treatment.
91,324,347,372,456,468,502
Several retrospective studies on large patient
populations have found that risk of metastasis is correlated with AJCC T
stage and with AJCC staging.
8,13,284,501,503-507
Primary uveal melanomas can contain spindle cells, which have ovoid
nuclei and tend to grow in a compact fashion, and epithelial cells, which
are larger, more irregularly contoured, pleomorphic, and contain abundant
cytoplasm, larger nuclei, and nucleoli.
31
The cell types present in the
primary lesion have been linked to risk of metastasis. One prospective and
several retrospective analyses have found that the histologic cell type(s) in
the primary tumor can be prognostic for metastasis, in that patients whose
primary tumor contains epithelioid cells (either entirely or mixed with
spindle cells) are more likely to develop metastases than those who have
only spindle cell type.
67,257,501,502,506,508-510
The presence of spindle versus
epithelioid cells in the primary tumor had been used as a prognostic factor
to inform frequency of follow-up after primary treatment, but this prognostic
feature is now considered less important than other tumor features and
molecular markers. Lack of concordance among pathologists makes
implementation of risk stratification by histopathology difficult.
In addition to AJCC T stage and tumor histology, multiple tumor molecular
markers have been shown to be associated with increased risk and/or
shorter time to development of distant metastases. Chromosomal changes
were among the first molecular markers to be found to be associated with
risk of distant metastasis in patients with uveal melanoma.
508
Multiple
studies have found that monosomy 3 and gain of chromosome 8q,
especially when numerous copies are found, in the primary uveal
melanoma is associated with increased risk of metastasis.
24,36,48,67,324,509-525
Risk of metastasis is even higher when both of these abnormalities are
present.
24,67,513,516,518,522
Some studies have identified additional
chromosomal abnormalities associated with increased risk of metastasis,
such as loss of 8p,
24,513,515,516,518
loss of 1p,
518
loss of 16q,
513
and loss of
6q.
24
Gain of 6q may be protective against metastasis,
518
at least in the
context of monosomy 3 and gain of 8q.
24
A method of using gene expression profiling (GEP) has been developed
as a prognostic tool for uveal melanoma.
526-528
These methods have been
used to sort tumors into two classes, showing that class 2 was associated
with higher risk of metastasis than class 1.
48,252,502,503,512,521,529-535
Multivariate analyses have found that class 2 is associated with a 5-fold to
20-fold higher risk of metastasis than class 1.
48,502,531,534,535
Mutation and expression of certain specific genes have also been
associated with risk of metastasis in patients with uveal melanoma.
Multiple studies have found that BAP1 mutation/deletion (observed in
approximately half of uveal melanomas) and loss of BAP expression in the
primary tumor is associated with increased risk of
metastasis.
36,72,504,506,521,523,532,536,537
One study showed that risk of
metastasis is highest with BAP1 somatic mutation, although also
somewhat elevated in patients with BAP1 germline mutation (compared
with wild-type).
519
Other studies found that BAP1 mutation was associated
with early metastasis (after treatment of primary uveal melanoma).
538,539
Mutation in EIF1AX, found in up to 20% of uveal melanomas,
36,510,532,540
has been associated with lower risk of distant metastasis in patients with
uveal melanoma.
506,538,539
Some studies have found that SF3B1 mutation,
which is present in approximately 20% of uveal melanomas, was
associated with lower risk of metastasis,
36,73
while others found that
patients with this mutation developed late metastases.
506,538,539
PRAME
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-34
expression, present in about a third of uveal melanomas,
503,541
has also
been associated with increased risk of metastasis.
503,533
Surveillance Methods for Distant Metastatic Disease
There are very little data to inform the frequency and modality of follow-up
screening for development of metastatic disease. Prospective studies
following patients after treatment for localized primary melanoma have
typically monitored for development of distant metastasis using regular
follow-up visits including complete cancer-oriented physical exams and
one or more of the following: chest x-ray, liver US, and serum liver function
tests (LFTs).
47,301,327,330,347,366,367,380,384,388,478,480
Liver US and LFTs were
often included as part of routine follow-up because the liver is the most
likely site of distant metastasis.
19,21,22,166,295,480,481,497
Most of these
prospective studies followed patients with physical exams, imaging, and
blood tests every 6 months for at least the first 5
years.
47,327,330,347,366,367,380,384,388,478,480
In real-world clinical practice, there
are regional differences in the preferred methods and frequency of follow-
up screening for metastatic disease in patients with uveal melanoma.
542
For most of the prospective studies that monitored for development of
distant metastatic disease after treatment of primary uveal melanoma, the
total number of patients who developed distant metastases was too small
to produce meaningful results regarding the sensitivity and specificity of
different surveillance modalities. The COMS trials for patients with medium
and large tumors were the largest of these prospective studies, and
included cancer-oriented physical exams, chest x-ray, and LFTs as part of
routine follow-up.
480
Elevated LFTs (aspartate aminotransferase [AST] >2x
the upper limit of normal [ULN]; alanine transaminase [ALT] >2x ULN;
alkaline phosphatase [APH] >1.5 xULN, bilirubin [BIL] ≥2.0 mg/100 mL)
were confirmed by repeat LFT testing, and further diagnostic testing, such
as biopsy and CT/MRI/US of the liver were used to confirm or rule out
distant recurrence.
480,481
These trials measured LFTs at 6, 12, and 18
months after treatment, and then annually thereafter.
477
Using this
approach, the likelihood of an abnormal LFT was low (<1%).
480
Based on
all patients with reported metastasis, the sensitivity, specificity, positive
predictive value, and negative predictive value associated with at least one
abnormal LFT before first diagnosis of metastasis at any site was 14.7%,
92.3%, 45.7%, and 71.0%, respectively. Of the LFTs, APH had the highest
diagnostic attributes. Other diagnostic tests appeared to have higher
sensitivity and specificity because they were often triggered by abnormal
LFTs. The results suggest that use of LFT results followed by other
diagnostic tests has high specificity and predictive values, but low
sensitivity.
480
Whereas 739 patients had distant metastases detected
during follow-up, 13 did not have their metastasis discovered until time of
death.
481
The utility of LFTs for early detection of liver metastases is an
issue of ongoing debate, with sensitivity, specificity, and positive and
negative predictive values varying across specific LFT test types and
differing between studies.
496,497,499,543-547
Whereas some analyses have
concluded that LFTs are among the most useful methods for
screening,
496,543-545,548,549
others argue that the specificity and sensitivity of
these tests is too low to warrant routine use.
499,500,547
The optimal strategy for imaging surveillance is also an issue of debate,
because for each of the methods commonly used (ie, chest x-ray, CT, US,
MRI, PET/CT) results vary, and for all of these options there is at least one
study that reported poor performance on at least one metric.
496-500,544,546,550-
560
Due to the low probability of metastasis at any specific time point, the
yield of all these tests is low, and there is concern regarding cumulative
radiation exposure due to the long-term follow-up needed.
561
Therefore,
imaging is usually focused on the liver, as it is the most likely site of distant
metastasis, and liver US and MRI are favored over CT or PET/CT. Some
studies have found MRI to be moderately better than CT or PET/CT for
detection of liver metastases from uveal melanoma,
550,554,555
and
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-35
prospective studies in high-risk patients showed promising results for
using liver MRI for early detection of liver metastases.
500,556
The value of surveillance imaging and tests is debated because it is
unclear whether early detection of distant metastases improves outcomes,
especially given that most treatments for distant metastatic disease are
relatively ineffective. Some retrospective studies found that survival was
better for patients whose distant metastases were asymptomatic at the
time of detection,
495,562
whereas other studies observed no such
correlation,
21,563
or reported that the difference was transient.
564
Risk of Developing Secondary Cancers During Follow-up
Due to the long-term surveillance needed for detection of distant
metastatic disease in patients with uveal melanoma, it is not uncommon
for screens to identify other primary cancers.
11,159,160,162,559,565-568
The two
COMS randomized trials in patients with medium to large primary uveal
melanomas showed that the proportion of patients with secondary cancers
increased steadily over the entire duration of follow-up (median 10 years,
range 516 years).
566
Various types of secondary malignancies were
observed.
159,566
Uveal melanoma may increase the risk of developing other
cancers,
160
especially in patients with familial uveal melanoma or other
familial cancers.
113,114
Follow-up for Distant Metastasis
Given the lack of high-quality data to inform the frequency or modality of
follow-up screening for distant metastatic disease, the NCCN
recommendations are based on clinical practice at NCCN Member
Institutions. Patients with no evidence of disease (NED) after treatment for
uveal melanoma should be followed for signs of metastatic disease. The
most frequent sites of metastasis are liver, lungs, skin/soft tissue, and
bones. Recommended follow-up for distant metastatic disease includes
imaging to evaluate signs or symptoms of distant metastasis, and may
include regular surveillance imaging. LFTs may be considered as a
component of follow-up visits, although some studies showed poor
sensitivity for early detection of liver metastases.
Recognizing that there are limited options for systemic recurrence and that
regular imaging may cause patient anxiety, patients should discuss with
their treating physician the potential benefits and risks of surveillance
imaging, and some patients may elect to forgo surveillance imaging.
Providers may also want to discuss mental health resources with patients.
See the NCCN Guidelines for Survivorship (www.NCCN.org
). Participation
in a clinical trial is strongly encouraged.
For patients who elect to have surveillance imaging to screen for distant
metastatic disease, options include contrast-enhanced MR or US of the
liver, with modality preference determined by expertise at the treating
institution. Additional imaging modalities may include
chest/abdominal/pelvic CT with contrast, or dual-energy subtraction chest
x-ray. However, screening should limit radiation exposure whenever
possible. Scans should be performed with IV contrast unless
contraindicated. Recommendations for imaging modality are based on
clinical practice at NCCN Member Institutions, as there are very few data
to inform selection of modality.
For those choosing to have regular surveillance (imaging with or without
blood tests), the recommended frequency is based on the risk of distant
metastasis. The NCCN Guidelines recommend risk stratifying patients into
low, medium, and high risk of distant metastasis based on the highest risk
factor present. For patients with high risk of distant metastasis who opt to
have surveillance imaging, the recommended frequency is every 3 to 6
months for 5 years, then every 6 to 12 months for years 6 through 10, then
as clinically indicated. For patients with medium risk of distant metastasis
who opt to have surveillance imaging, the recommended frequency is
every 6 to 12 months for 10 years, then as clinically indicated. For patients
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-36
with low risk of metastasis who opt to have surveillance imaging, consider
imaging every 12 months. Adjusting follow-up frequency based on risk of
metastasis is based on clinical practice at NCCN Member Institutions, and
this approach has not been prospectively tested to determine whether it
results in better yield from imaging or better outcomes. There are very little
data to inform the recommended frequency of imaging follow-up.
Risk factors for metastasis include a variety of genetic markers as well as
tumor size at presentation. The NCCN Guidelines list specific risk factors
to be used for risk stratification to determine the frequency of surveillance
imaging during follow-up. The NCCN Guidelines recommend using AJCC
T stage for risk stratifying according to primary tumor size. T1 is
considered low risk, T2 and T3 medium risk, and T4 high risk. For patients
who had a biopsy of their primary tumor, both cell histology and certain
molecular features have been shown to be prognostic for risk of distant
spread, and should be used for risk stratification. GEP as described by
Onken et al
48
is recommended to determine whether the tumor is Class 1A
(low risk), Class 1B (medium risk), or Class 2 (high risk) to inform
frequency of follow-up. The following chromosomal abnormalities are also
considered risk factors that should inform frequency of follow-up: disomy 3
(low risk), gain of chromosome 6p (low risk), monosomy 3 (high risk), and
gain of chromosome 8q (high risk). Mutations in several genes have also
been shown to be prognostic for distant metastasis, and should be used
for risk stratification: EIF1AX (low risk), SF3B1 (medium risk), and BAP1
(high risk). PRAME expression is also an indicator of high risk to be used
to inform frequency of follow-up. If biopsy not performed, then follow
medium- or high-risk pathways, depending on whether any high-risk
features are present.
Management of Recurrence
Workup for Recurrence
If a recurrence is detected, workup should include history and physical to
identify any signs or symptoms associated with recurrence or metastasis.
Biopsy may also be appropriate. Whereas intraocular recurrence can often
be diagnosed and managed without a biopsy, additional prognostic FNA
biopsy may be valuable to determine whether the tumor has developed
any high-risk features that warrant more frequent surveillance. Extraocular
recurrence or metastasis should be confirmed histologically whenever
possible or if clinically indicated. Appropriate biopsy techniques in this
setting may include FNA or core biopsy. For patients with metastasis who
are considering treatment with targeted therapy, tissue should be obtained
for genetic analysis (screening for mutations that may be potential targets
for treatment or to determine eligibility for a clinical trial) from either biopsy
of the metastasis (preferred) or archival material. Broader genomic
profiling may be considered if the results could inform future treatment
decisions or eligibility for clinical trials.
Patients with local recurrence should have ocular orbital imaging (if not
recently previously done) to evaluate the extent of local recurrence.
Patients who develop distant metastatic disease after treatment of primary
uveal melanoma should have ocular orbital imaging as part of workup to
check for local recurrence, since asymptomatic local recurrences may be
present at the time distant metastasis is discovered. Workup for patients
with recurrence should include broader imaging to investigate specific
signs or symptoms, and/or for baseline staging. Because the most
frequent sites of metastasis are liver, lungs, skin/soft tissue, and bones,
imaging options for baseline staging in patients with recurrence or
metastasis include contrast-enhanced MR or US of the liver, with modality
preference determined by expertise at the treating institution. Additional
imaging may include chest/abdominal/pelvic CT with contrast and/or
whole-body FDG PET/CT. Brain MRI with IV contrast may be performed if
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-37
neurologic symptoms are present, but routine CNS imaging is not
recommended. All scans should be performed with IV contrast unless
contraindicated. As described below, for patients with distant metastasis, a
thorough evaluation of the size and location of all metastases can help in
assessment of prognosis and evaluation of treatment options.
For patients with distant metastases, consider measuring LFTs, including
LDH, as part of workup. However, their role in risk stratification of
metastatic uveal melanoma is unknown. As described below, elevation of
LFTs has been associated with poorer OS in patients with metastatic
uveal melanoma.
19,20,22,497,569-574
Treatment for Local Recurrence
Given the rarity of local recurrence after treatment of primary uveal
melanoma, data on treatment of local recurrences are scant, and it is
unclear which approaches result in the best outcomes. Most studies had
fewer than 10 patients with local recurrence, and many studies either
managed all local recurrences with enucleation or did not report on
retreatment approaches. In the few studies that reported outcomes after
treatment of local recurrence (n 10),
348,388,416,423,443,575-579
reasonably high
rates of local control were achieved with the following globe-conserving
modalities: laser photocoagulation,
348,416
TTT,
443,578
proton-beam
RT,
416,423,576-578
and plaque brachytherapy RT.
388,443,578,579
Similar to the
primary treatment setting, some patients treated with globe-conserving
therapy for local recurrence subsequently underwent enucleation due to
(suspected or confirmed) tumor regrowth or complications such as pain
and neovascular glaucoma.
348,416,423,575,576,578,579
There is very little
evidence to inform selection of treatment for recurrence. Results from one
retrospective study of 73 patients with local recurrence suggest that
treating recurrence with proton-beam RT (n = 31) versus enucleation (n =
42) may result in similar metastasis-free survival and OS.
577
Another
retrospective analysis of 51 patients with local recurrence found that local
control after treatment of recurrence was more likely in those with longer
times between primary treatment and development of recurrence, and risk
of metastasis was higher in patients whose local recurrence was
characterized by vertical/diffuse growth versus horizontal/marginal
growth.
348
NCCN Recommendations for Treatment of Local Recurrence
The recommended treatment options for local recurrence depend on the
extent of disease. For intraocular recurrence (limited to the eye, without
orbital involvement), the recommended options include radiation, either by
plaque brachytherapy or particle beam, enucleation, or laser ablation. For
small recurrences in patients who cannot undergo RT or surgery, trans
TTT is recommended. TTT is usually reserved for small recurrences,
particularly when recurrence is likely due to incomplete plaque coverage
during primary brachytherapy; it is generally not appropriate for
recurrences occurring within the RT field or recurrences that may be too
thick (>3 mm) for laser treatment to reach the base. It is important to have
an in-depth discussion with patients about their treatment options for local
recurrence. If there is extraocular involvement, surgical resection is
needed, but can be coupled with RT to the orbit (particle beam or photon
beam) and/or cryotherapy to the orbital tumor. If there is orbital
involvement and the patient has had prior enucleation, options include
surgical resection or cryotherapy to the orbital tumor, and/or RT to the
orbit (particle beam or photon beam). Recommendations for administration
of different RT modalities are in the section entitled Radiation Therapy
(above).
Treatment for Metastatic Disease
Survival after detection of distant metastatic disease varies widely across
studies, with median OS ranging from 3 to 30 months.
569,580
Survival
outcomes vary widely even between studies testing the same or similar
treatments,
569,580,581
and even when only considering studies with large
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-38
patient sample sizes (n > 100; median OS 320 months).
21-23,481,495,570,580
Part of the variation could be due to differences in patient selection, as
there are many factors that have been shown to be associated with
survival. Some meta-analyses suggest that survival after metastasis is
shorter for studies of unselected patients compared with studies of
selected patients, but those with selected patients were also more likely to
be testing an active treatment and those of unselected patients were more
likely to include cases managed with best supportive care or palliative
approaches.
569
Several studies have found that a small percentage of
patients with uveal melanoma metastasis experience long-term survival
(≥5 years) after development of distant metastatic disease.
19,21,481
Several
long-term studies have shown a bimodal distribution suggesting a
population with short-term survival (median OS <1 year) and a separate
population with long-term survival (median OS >2 years).
19,21
Studies have reported a wide range of factors associated with OS after
detection of metastasis, and results vary across studies. Patient
characteristics reported by multiple studies, including multivariable
analyses, to be associated with poorer OS after metastasis include older
age, male sex, and poorer performance status, although there are
opposing data for each of these factors.
19-22,481,497,501,563,569-572,574
Multiple
studies have also found that poor OS after metastasis is associated with
symptoms at the time of metastasis (compared with asymptomatic
metastasis detected by surveillance), shorter disease-free interval before
metastasis, higher number of anatomic sites involved, involvement of liver,
and greater disease volume (based on various metrics, such as percent of
liver involvement, volume of liver metastases, total number of metastases,
number of liver metastases, size of largest metastasis or largest liver
metastasis, M-stage).
19-22,497,501,546,562-564,569-574,582-585
In addition, elevated
liver enzymes at the time of diagnosis of metastasis, particularly LDH and
APH, have been associated with shorter OS.
19,20,22,497,569-574
Whereas in recent years the options for treating metastatic cutaneous
melanoma have dramatically improved, treatment of distant metastases
from uveal melanoma still presents a major clinical challenge. Several
retrospective studies suggest that treatment for distant metastases (from
uveal melanoma) improves survival, although it is unclear whether these
results are influenced by selection bias.
21,23,501,574,584
Other studies did not
find that treatment improved survival,
481,572
reporting that patients treated
with supportive care only had median OS ranging from 1.7 to 4.9 months,
although those opting for supportive care are more likely to have risk
factors for poor survival.
23,501,584
For treatment of distant metastasis from
uveal melanoma, a wide variety of approaches have been tested,
including surgery, RT, ablative approaches, vaccines, various systemic
therapies (chemotherapies, immunotherapies, targeted therapies, and
various combinations), and localized
chemotherapy/immunotherapy.
22,569,580,581,585-587
Systematic reviews, meta-
analyses, and retrospective studies that included patients treated with a
variety of therapies suggest that the best outcomes are seen in patients
who received liver-directed treatments, particularly those treated with
surgery, especially if complete resection was
achieved.
19,22,500,562,563,569,571,574,580,582,585,588,589
It is unclear whether these
effects are due to other factors, such as the lower volume of metastatic
disease, which would make a patient eligible for surgery and/or liver-
directed treatments, and more likely to have complete resection. One
phase III randomized trial (EORTC 10821) found no difference in OS in
171 patients with liver metastasis from uveal melanoma treated with IV
fotemustine versus hepatic arterial infusion (HAI) with fotemustine, despite
the HAI approach showing higher response rates and progression-free
survival (PFS).
590
A few studies suggest that better survival was seen in
those who had a response to therapy for metastatic disease,
21,569
but
some did not find an association.
19
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-39
Resection of Metastases
As mentioned above, multiple prospective and retrospective studies and
meta-analyses have found that among patients with metastatic uveal
melanoma, those who can be treated with surgery have the best
outcomes, especially if complete resection is achieved.
21,562,563,569,580-
582,585,589,591-595
Multiple studies have reported median OS greater than 20
months after resection of uveal melanoma
metastases.
21,562,563,581,585,589,591,592,594-599
For liver metastases, rates of
complete resection ranged from 27% to 88%.
21,563,582,585,591,592,595,597
Multiple studies reported on combination therapy with resection and
hepatic arterial infusion, radiofrequency ablation (RFA), transarterial
chemoembolization (TACE), or adjuvant systemic
therapy.
562,563,581,585,591,596,600-602
Liver-Directed Therapy
Due to the tendency of uveal melanoma to metastasize to the liver, a wide
range of liver-directed therapies have been tested in patients with liver
metastases from uveal melanoma. These include regional isolation
perfusion of the liver, various methods of embolization (eg, chemotherapy,
radiation, immunotherapy), ablative procedures (eg, RFA), and
resection.
22,569,580,581,589,603-605
Whereas there are a large number of
prospective pilot, phase I, and phase II studies that reported outcomes for
liver-directed approaches, it is unclear which liver-directed approaches are
the best because of the lack of randomized comparative studies in
patients with liver metastases from uveal melanoma.
Regional Isolation Perfusion
Several techniques have been developed for localized delivery of
pharmaceutical therapy to the liver for treatment of hepatic metastases.
The idea behind these techniques is that higher doses can be
administered locally than would be feasible systemically due to toxicity.
Methods include isolated hepatic infusion (IHP), percutaneous hepatic
perfusion (PHP), HAI, and embolization techniques, which are described
in the next section.
Liver metastases derive most of their blood supply from the hepatic artery,
whereas the blood source for benign hepatocytes is primarily the portal
vein.
606
IHP and HAI both deliver therapy via the hepatic artery to
maximize drug delivery to liver metastases while limiting exposure to
healthy parenchyma.
607
For IHP, higher temperatures are often used to
further increase the effective concentration of therapeutic agent while
limiting systemic exposure.
607
Whereas IHP is done during surgery, has
risk of morbidity, and usually can only be done once, HAI and PHP are
less invasive techniques, with lower risk of morbidity, and have the
potential to be performed multiple times to increase depth of
response.
581,589,605,607
IHP is an open surgical procedure involving vascular isolation of the liver,
allowing high doses of heated chemotherapy to be directly delivered to the
organ through an arterial catheter.
581,589,603,605,607
Alkylating agents are
generally preferred because they can be effective even with short
exposure time, and the dose-response curve is steep.
589
Several small
prospective studies (n < 40)
608-612
and a few retrospective studies (n = 19
to 68)
613-616
have tested hyperthermic IHP in patients with liver metastasis
from uveal melanoma. The agent most commonly used in these studies
was melphalan, with or without tumor necrosis factor alpha (TNF-alpha) or
cisplatin.
608-612
Good response rates to IHP have been reported in some of
these prospective studies (overall response rate [ORR], 50%–68%),
608-611
and retrospective studies (ORR, 67%–83%).
613-615
However, the IHP
procedure is often lengthy (>8 hours),
581,589,603,608,609
involves significant
blood loss (median 23.5 L),
581,608,609
involves long hospital stays (710
days),
589,603,608,609,615,616
can result in significant complications/morbidities
(eg, portal vein thrombosis, transient grade 3/4 hepatic toxicity),
589,603,608-
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-40
610,613-616
and mortalities from the procedure were also
observed.
603,608,613,616
For these reasons IHP is not widely used.
HAI is a technique using catheters in the hepatic artery to deliver
chemotherapy directly to the liver.
589
The number of times HAI can be
repeated depends on the type of catheter used.
589
Efficacy of HAI for liver
metastases from uveal melanoma has been reported in several small
prospective studies (n = 8–31)
617-621
and several retrospective studies (n =
10100).
622-626
Most of these studies used fotemustine,
618,619,622,623,625
but
other agents tested include carboplatin;
617,625
nab-paclitaxel;
620
decitabine;
621
a combination of cisplatin, vinblastine and dacarbazine;
624
and melphalan.
626
Response rates to HAI were lower and more variable
(than for IHP) in both prospective (ORR, 0%–44%)
617-621
and retrospective
studies (16%–36%).
622,625
One phase III randomized trial (EORTC 10821)
in patients with liver metastasis from uveal melanoma found no difference
in OS for IV fotemustine versus HAI fotemustine (median OS, 13.8 vs.
14.6 months), despite the HAI approach showing higher response rates
(2.4 vs. 10.5%) and PFS (median 3.5 vs. 4.5 months; HR, 0.62; 95% CI,
0.450.84, P = .002).
590
Complication rates varied widely across studies,
but in general appear to be lower than those reported in studies testing
IHP, and are manageable.
590,617-619,622,623,625,626
Grade 3/4 adverse events
(AEs) were mostly hematologic (eg, anemia, leukopenia,
thrombocytopenia, neutropenia).
590,617,621-623
Grade 34 catheter-related
complications occurred in a minority of patients (≤12%), as did liver toxicity
(<10%).
590,622
Deaths from toxicity were reported in one study,
590
but not in
others.
PHP is a simpler, less invasive alternative to IHP that can be
repeated.
589,605
It uses a double-balloon catheter inserted into the inferior
vena cava to isolate hepatic venous blood that is then filtered
extracorporeally.
589,605
Several prospective studies tested PHP in patients
with liver metastases from uveal melanoma, but efficacy results are
lacking.
627-629
Retrospective studies suggest good response rates (>40%),
especially if multiple rounds are used.
630-632
In all these studies melphalan
was the agent used.
627-632
PHP appears to be somewhat better tolerated
than IHP, with no treatment-related fatalities, but many patients still
experienced hematologic grade 34 events, some had non-hematologic
grade 34 AEs (eg, bleeds, thromboembolism), and some had extended
hospital stays (45 days) or had to be readmitted.
629-631
Hepatic Embolization
Hepatic arterial embolization is a method for delivering to the liver
chemotherapy, immunotherapy, or radioactive agents, while increasing
dwell time and providing selective ischemia.
589,603
Hepatic Chemoembolization
There are no standard protocols for hepatic chemoembolization, also
called hepatic TACE or hepatic arterial chemoembolization (HACE). In
general, two approaches have been used for treatment of uveal melanoma
metastases to the liver. One involves HAI with the active agent, followed
by addition of either a transient or permanent embolic agent.
589,603
The
active agent is usually mixed with ethiodized oil (to increase dwell time),
and embolization agents include absorbable gelatin sponge or polyvinyl
alcohol particles.
589,603
The other approach uses drug-eluting beads
produced from a polyvinyl alcohol hydrogel that has been modified with
sulfonate groups for the controlled loading and delivery of chemotherapy
agents.
603
These beads serve for localized drug delivery and as an
embolic agent to render tumors ischemic.
603
Multiple retrospective
633-640
as well as a few prospective studies
641-644
have
evaluated chemoembolization for treatment of hepatic metastasis in
patients with uveal melanoma. Using traditional methods (active agent
infusion + addition of embolic agent), response rates varied widely, but
could be as high as 57%.
633,634,636,637,639-645
Chemotherapies used included
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-41
BCNU, mitomycin C, fotemustine, cisplatin, carboplatin, doxorubicin, and
1,3-bis (2-chloroethyl)-1-nitrosourea; embolic agents used included gelatin
sponge, polyvinyl sponge, resorbable microspheres, and polyvinyl alcohol
particles.
633,635,636,638-645
Only a few studies reported outcomes for patients with liver metastases
from uveal melanoma that were treated with trans-arterial chemotherapy-
eluting beads.
646-648
One phase II study reported 100% ORR in the 10
patients treated with irinotecan-loaded polyvinyl alcohol microspheres.
646
In contrast, a retrospective study of irinotecan-loaded beads in 28 patients
with uveal melanoma liver metastases reported much lower response
rates.
648
A retrospective study including 58 patients treated with TACE
using irinotecan-charged microbeads reported an ORR of 27.5% (all
partial responses).
649
Another prospective noncontrolled study testing
beads loaded with doxorubicin in patients with unresectable liver
metastases from ocular melanomas only reported toxicity and quality of
life.
647
Comparisons of hepatic chemoembolization with other treatments for
uveal melanoma liver metastasis are limited. One retrospective study
compared cisplatin-based chemoembolization (TACE) versus HAI and
versus systemic therapy, and found that chemoembolization was
associated with the best response rate, although OS did not differ between
the groups.
634
However, those who responded to TACE had better OS
than those who did not respond and better than those treated with HAI or
systemic therapy.
634
For patients with uveal melanoma liver metastases
treated with chemoembolization, several studies found that OS was better
in responders versus non-responders,
634,637,638,640,641,643
whereas others did
not find a significant association.
642,649
Chemoembolization is well tolerated in patients with liver metastases from
uveal melanoma, with few or no treatment-related deaths.
636,642,644,645,647-650
AEs reported in more than one study include abdominal pain, fever,
nausea, vomiting, liver dysfunction, and
thryombocytopenia.
633,636,639,640,644,646,648,649
Some studies recommend
supportive treatment with antibiotic and antiemetic prophylaxis, IV
hydration, and major analgesic before and after the procedure.
644,646
Hepatic Immunoembolization
Hepatic immunoembolization involves infusion of an immunologic
stimulant into the hepatic artery, followed by addition of an embolizing
agent.
603
The rationale is that the ischemia will start the destruction of the
tumor, releasing antigens so that local simulation of the immune system
may result in systemic immune response to prevent tumor growth.
603
Several studies in patients with liver metastases from uveal melanoma
have tested immunoembolization using granulocyte-macrophage colony-
stimulating factor (GM-CSF) ethiodized oil plus a gelatin sponge.
651,652
GM-CSF is a glycoprotein secreted by immune cells such as activated T
cells that increases myeloid cell production, stimulates macrophages,
increases cytotoxicity of monocytes toward tumor cell lines, and promotes
maturation of dendritic cells.
603
A phase I trial with 34 uveal melanoma
patients and unresectable liver metastases occupying less than 50% of
total liver volume reported an ORR in the liver of 32%, and response
correlated with better OS.
651
A phase II randomized trial with 52 patients
with uveal melanoma and hepatic metastases only (also <50% liver
volume) reported an ORR of 21.2% for the 25 patients treated with
immunoembolization versus 16.7% for the 27 who received bland
embolization (GM-CSF replaced with saline).
652
In this study hepatic
response was not correlated with OS, but was associated with better PFS.
AEs associated with immunoembolization in these studies included
abdominal pain, fever, nausea, and transient increases in hepatic
enzymes.
651,652
One patient had acute respiratory failure but recovered;
there were no treatment-related deaths.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-42
Hepatic Radioembolization
Hepatic radioembolization, also called hepatic transarterial
radioembolization (TARE) or selective internal RT (SIRT), is a procedure
in which glass or resin yttrium-90 microspheres are introduced to the
hepatic artery, both as a mechanism for radiation delivery and for
embolization.
603
The microspheres must be of sufficient size to have
embolic effect.
603
Response rates from retrospective studies varied widely
(6%–100%), but disease control rate was consistently greater than
50%.
653-658
A phase II study reported ORR of 39% in the 23 patients who
received radioembolization as first-line treatment for liver metastasis, and
ORR of 33% in the 24 patients who received radioembolization after
progression on immunoembolization, with disease control rate of 87% and
58%, respectively.
659
Across studies, radioembolization was well tolerated,
with most toxicities being grade 12 and self-limiting, and no treatment-
related deaths.
653-657
AEs included abdominal pain/discomfort, nausea and
vomiting, LFT elevation (sometimes due to progression), transient
lymphopenia, and gastric ulcer.
654-657,659
Ablative Procedures for Liver Metastases
Although ablative procedures such as cryotherapy and thermal ablation
have been used with some success for liver metastases from other kinds
of cancer, there are very little reported clinical data on the efficacy of these
techniques for liver metastases from uveal melanoma. The supposed
advantage of ablative techniques is that compared to surgery these
techniques are tissue-sparing, less invasive, cost-saving, and have lower
rates of complications, while still being potentially curative.
660
Ablation may
be feasible for tumors that are difficult to resect (due to location), although
for some tumors ablative options may be limited due to tumor visibility and
correct interpretation of tumor extent and stage.
660
The most commonly
used methods of thermal ablation for liver tumors include RFA and
microwave ablation (MWA).
660-662
Both methods use heat induction and
destroy tissue through thermally induced coagulative necrosis. The
difference is that RFA uses alternating electric current at frequencies from
375 to 500 kHz, and MWA uses an electromagnetic field at frequencies
greater than 900 kHz.
660,662,663
RFA uses an electrode to deliver alternating
current that heats tissue to 50 to 100°C near the electrode, causing almost
instant coagulation necrosis.
660,662,663
For RFA, ablation volume is limited
by tissue boiling and charring that then insulate the effect through
increased impedance, and limited by the heat-sink effect caused by blood
flow dispersing the thermal energy.
661-663
The resulting ablation area may
have unpredictable size and shape, and multiple sessions or multiple
electrodes may be needed to fully ablate the target area.
661,663
MWA uses
an antenna probe to generate an electromagnetic field that rapidly heats
surrounding tissue to greater than 150°C, causing necrosis.
660-663
The
heat-sink effect is more limited than in RFA, and the resulting ablation
zone is larger and more homogeneous than with RFA.
660-664
However, the
ability to rapidly ablate a larger area can result in larger areas of healthy
tissue damage.
663,665
Based on data from other (non-uveal) types of liver
tumors, safety of MFA and RFA appears similar.
662,663,665-667
Both methods
rarely lead to major complications (<3%), such as hemorrhage, infection,
organ injury, liver failure, pneumothorax, pleural effusions, ascites, fever,
and portal vein thrombosis.
662,663,667
Due to the need for grounding pads to
complete the circuit, RFA can also cause skin burns.
663
Cryoablation uses rapid gas expansion at a probe tip to quickly cool
surrounding tissues to as low as -140°C.
660,661,668
This creates an ice ball
that dehydrates tissues, and causes irreversible cell damage and cell
death.
660,661,668,669
Vascular injury also causes cell death by ischemic
hypoxia.
668,669
Repeated freeze-thaw cycles are often used to maximize
tissue cell death throughout the target area.
660,661,668,670
Cryotherapy can
be performed percutaneously, laparoscopically, or during open surgery.
670
The result of the procedure is a zone of central necrosis surrounded by
tissue in which cells are not fully damaged.
670
Damage to the
microvasculature can cause edema, inflammation, and thrombosis.
670
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-43
Complications may include pain, infection (eg, wound infection,
pneumonia), hemorrhage, biliary injury, thrombocytopenia, pleural
effusion, renal impairment, and in older literature, occasionally a fatal
complication called “cryoshock.”
661,668,670,671
Whereas some studies found
that cryoablation was more likely to cause complications compared with
RFA, other analyses suggest that complication rates are similar to RFA,
especially in more recent studies.
660,661,668,669,671-673
There are very few studies reporting outcomes for patients with liver
metastases from uveal melanoma treated with ablative therapy.
596,601,674-677
Methods tested include RFA
596,601,675,677
and laser-induced interstitial
thermotherapy (LITT).
674
The efficacy and safety of ablative techniques is
difficult to ascertain, because most of these studies contained fewer than
20 patients with uveal melanoma,
601,674-677
included both patients with
uveal melanoma and other types of melanoma (and did not report results
separately),
675,676
or combined the ablative therapy with other therapies (ie,
surgery, TACE, systemic therapy).
596,601,674,677
Nonetheless, it is notable
that several retrospective studies reported that relatively long median OS
was greater than 20 months in cohorts of uveal melanoma patients with
liver metastases treated with LITT (±TACE),
674
RFA,
675
or percutaneous
thermal ablation (± systemic therapy).
676
In a retrospective study of uveal
melanoma patients with liver metastases treated with surgery alone (n =
57) or a combined approach in which some metastases were resected and
others treated with RFA (n = 13), those treated with the combination
approach had similar disease-free survival as those treated with surgery
alone (median 7 vs. 10 months) and OS (median 28 vs. 27 months).
596
Moreover, there were no recurrences at the 22 sites treated with RFA after
a median follow-up of 63 months (range 783 months).
596
These results
suggest that RFA may be as effective as surgery, and could be used in
lieu of surgery for metastases that are difficult to resect.
External Beam Radiation for Uveal Melanoma Metastases
Published data on external beam RT for uveal melanoma metastases are
extremely scant with no study reporting outcomes or palliative effects.
Systemic Therapy for Distant Metastatic Disease
Many systemic therapies have been tested in prospective trials as
treatment for metastatic uveal melanoma, including chemotherapies
590,678-
688
targeted therapies,
685-687,689-699
and immunotherapies.
700-713
Many
systemic therapy combinations have also been tested in prospective trials,
including chemotherapy combinations,
681,714-723
biochemotherapy
(chemotherapy + immunotherapy),
724-727
and a variety of other systemic
therapy combinations.
684,728-732
For treatment of metastatic uveal
melanoma, systemic therapies have largely been tested in small phase II
studies, and most have shown little activity (response rate <10%),
587,589,733-
735
especially compared with the efficacy of checkpoint immunotherapies
and BRAF/MEK inhibitor combinations in metastatic cutaneous
melanoma.
736-744
The few larger randomized phase II/III trials comparing
systemic therapies for metastatic uveal melanoma have failed to identify
any systemic therapies that are consistently more effective than
chemotherapy,
681,684-687,693,695,728
although there are some recent promising
results from noncomparative studies that warrant further investigation. As
noted above, meta-analyses suggest that for metastatic uveal melanoma,
systemic therapy appears to result in worse outcomes than localized
treatment (surgery or liver-directed therapies), although differing patient
selection criteria across studies may be a confounding factor in these
analyses.
22,580,581,589
One randomized trial showed that (IV) systemic
chemotherapy results in lower response rate and shorter PFS than HAI
chemotherapy in patients with liver metastases, although OS was similar
across arms.
590
For patients who are not appropriate for localized therapy,
selection of systemic therapy is very challenging, necessitating further
study into better options.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-44
Cytotoxic Regimens
A wide variety of chemotherapies have been tested in prospective trials
(ie, pilot, phase I, phase II) in patients with metastatic uveal melanoma,
and outcomes have been reported for the following single-agent
chemotherapies: dacarbazine,
684-687
paclitaxel,
680
DHA-paclitaxel,
679
temozolomide,
678,685,687
fotemustine,
590
bendamustine,
682,683
treosulfan,
681
liposomal vincristine,
683
arsenic trioxide,
688
and lenalidomide.
745
Most
studies evaluated patients for response to treatment, but responses to
these therapies were rarely observed.
590,678-688,745
In these prospective
trials the few responses observed were in patients treated with
fotemustine (2/83),
590
liposomal vincristine (1/4),
683
dacarbazine (3/36),
686
and DHA-paclitaxel (1/22).
679
For the studies that measured PFS and OS
with these single-agent chemotherapy regimens, median PFS was always
less than 4 months,
590,678,679,681,684-687
and median OS was nearly always
greater than 10 months.
590,678,679,685-687
EORTC 10821, a phase III
randomized trial, reported median OS of 13.8 months for patients treated
with IV fotemustine, but this longer OS is likely due to the trial including
only patients with liver metastases (no extrahepatic metastases).
590
It
remains unclear whether any single-agent chemotherapy improves
survival relative to best supportive care.
Combination chemotherapies that have been tested in prospective trials
for metastatic uveal melanoma include gemcitabine/treosulfan,
681,714-718
dacarbazine/treosulfan,
720
cisplatin/gemcitabine/treosulfan,
719,746
cisplatin/dacarbazine/vinblastine,
721
docetaxel/carboplatin,
722
and
tirapazamine/cisplatin.
723
All of these were tested in pilot, phase I, or
phase II studies, most of which reported response rates of less than 5%.
Due to an early pilot study that reported a response rate of nearly 29% for
patients with metastatic uveal melanoma (n = 14),
718
gemcitabine/treosulfan was tested in multiple studies, but in the five
subsequent studies the response rate was much lower, ranging from 0%
to 4.2%.
681,714-717
Four of these studies (including the pilot) reported PFS
and OS data, with median PFS ranging from 2.5 to 6.7 months, and
median OS ranging from 7.5 to 14.2 months.
681,714,715,717,718
Results from
one phase II study suggested that higher treosulfan doses (≥3500 mg/m
2
)
provided better outcomes with this combination. Nonetheless the response
rate at these higher doses was only 5.2%.
714
A randomized phase II study
found that gemcitabine/treosulfan combination, using the higher treosulfan
dose, did provide better PFS and a trend for better response compared
with treosulfan alone, but the overall response was still relatively low
(4.2% in the combination arm), and the PFS relatively short (mean 3
months in the combination arm).
681
Addition of cisplatin to
gemcitabine/treosulfan did not improve resultsthere were no responses
in both of the studies that tested this triplet.
719,746
There were also no
responses seen in the phase II trials that tested dacarbazine/treosulfan,
720
docetaxel/carboplatin,
722
and tirapazamine/cisplatin.
723
The one phase II
study testing cisplatin/dacarbazine/vinblastine reported a surprisingly high
ORR of 20%, with median PFS of 5.5 months and median OS of 13.0
months.
721
This result needs to be repeated, and may be a product of
patient selection.
Meta-analyses combining results across studies that tested chemotherapy
in patients with metastatic uveal melanoma report ORRs ~4%,
587
median
PFS of 2.6 months,
22
and median OS ranging from 9 to 11 months.
22,580
Targeted Therapy
Targeted therapies that have been tested in prospective studies as single-
agent therapy for patients with metastatic uveal melanoma include the
tyrosine kinase inhibitors (TKIs) imatinib
689,690,747
and sunitinib,
686,692
the
mitogen-activated protein kinase (MEK) inhibitors trametinib
693,694
and
selumetinib,
685
the multi-kinase inhibitor sorafenib,
691
the receptor TKI
cabozantinib,
687,695
the VEGF inhibitor aflibercept,
698
the HSP90 inhibitor
ganetespib,
696
and the topoisomerase inhibitor 9-nitro-camptothecin.
699
Targeted therapy combinations that have been tested in prospective trials
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-45
include trametinib plus uprosertib, an inhibitor of protein kinase B (AKT);
693
binimetinib (MEK inhibitor) plus sotrastaurin (PKC inhibitor);
735
and
everolimus (mTOR inhibitor) plus pasireotide (IG1FR inhibitor).
697
Prospective studies in patients with metastatic uveal melanoma have also
tested combinations of chemotherapy plus targeted therapy, including
carboplatin/paclitaxel/sorafenib,
731
fotemustine/sorafenib,
748
dacarbazine/selumetinib,
684
temozolomide/bevacizumab,
732
and
carboplatin/paclitaxel/bevacizumab ± everolimus.
728
These prospective
studies were pilot studies, phase I trials, and phase II trials, and in most of
them the overall response was less than 10%.
685,689-691,693-699,747
Although uveal melanomas often express KIT, they rarely harbor the c-KIT
mutations associated with response to imatinib in other cancers.
59,689,747,749
It is thus perhaps not surprising that response rates to imatinib were low in
patients with metastatic uveal melanoma.
689,690,747
Two studies reported no
responses,
689,690
and 8% response was observed in a third study that
selected patients with high KIT expression.
747
These responses were seen
in patients who did not have activating mutations of c-KIT in exons 11, 13,
or 17.
747
Given the unimpressive median PFS (2.8 months) and OS (6.9
months) for the 25 patients in this study, imatinib is not considered a good
option for patients with metastatic uveal melanoma.
747
Results were similar
for the other TKI, sunitinib. A pilot study in 20 patients with metastatic
uveal melanoma expressing KIT reported an ORR of 5% for patients
treated with sunitinib,
692
but a subsequent larger phase II randomized
study reported no responses in the 38 patients treated with sunitinib (vs.
ORR of 8% with dacarbazine), and PFS and OS with sunitinib were no
better than the comparator dacarbazine.
686
Although BRAF mutations are rare in uveal melanoma,
34,51,53,54,60,62,63
most
uveal melanomas carry mutations in GNAQ or GNA11 that result in
constitutive activation of the RAS/RAF/MEK/ERK
pathway.
34,35,51,64,66,69,70,750
Prospective studies of MEK inhibitors have
yielded mixed results in patients with metastatic uveal melanoma.
685,693,694
A large phase II randomized trial in 101 patients with metastatic uveal
melanoma reported responses in 14% of the 50 patients treated with
selumetinib, and no responses in the 51 patients in the comparator arm
(chemotherapy with temozolomide or dacarbazine).
685
Selumetinib
modestly improved PFS compared with chemotherapy (median 3.7 vs. 1.6
months; P < .001), although the effect on OS was not significant (median
11.8 vs. 9.1 months; P = .09).
685
However, in a phase III randomized trial
in 129 patients with metastatic uveal melanoma that compared selumetinib
plus dacarbazine versus placebo plus dacarbazine, selumetinib did not
improve response (3% vs. 0%) or PFS (median 2.8 vs. 1.8 months).
684
Results from other MEK inhibitors were not impressive. No responses to
trametinib were observed in the 16 patients with metastatic uveal
melanoma in a phase I trial, and the median PFS was unremarkable (1.8
months).
694
In a subsequent phase II study, 1 of 18 (5.6%) patients with
metastatic uveal melanoma responded to single-agent trametinib, and
PFS was slightly better (median 3.6 months).
693
Addition of uprosertib, an
AKT inhibitor, yielded very similar results (ORR 4.8%, PFS median 3.6
months).
693
A phase Ib/II trial of combination binimetinib (MEK inhibitor)
and sotrastaurin (PKC inhibitor) yielded no responses in patients with
metastatic uveal melanoma.
735
Across studies it appeared that GNAQ and
GNA11 mutation status did not impact response rate or outcomes in
patients treated with MEK inhibitors.
684,685,694
Taken together, these data
show that some patients with uveal melanoma may respond to the MEK
inhibitor selumetinib, and there are limited data suggesting that trametinib
may also be marginally effective. Although the data are strongest for
selumetinib, it was not FDA approved for use in humans at the time of the
most recent Guidelines update, so it could not be included as a
recommended option. Trametinib is included as a recommended option
based on the positive data for selumetinib and the general lack of systemic
therapy options for patients with uveal melanoma. Further study of
trametinib for uveal melanoma is needed. Low-grade AEs are common in
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-46
patients treated with MEK inhibitors, with the most common being rash or
dermatitis acneiform, diarrhea, nausea, fatigue, hypertension, peripheral
edema, elevated AST/ALT, creatinine kinase elevation, and blurred vision
or other visual changes.
684,685,693,694,751
Grade 34 events were observed in
20% to 40% of patients, and dose reductions were often needed to
manage toxicities.
684,685,693,694,751
The multikinase inhibitor sorafenib did not result in any responses in the
32 patients with metastatic uveal melanoma in a phase II trial.
691
A phase
II trial combining sorafenib with carboplatin/paclitaxel also showed no
responses in 24 patients with metastatic uveal melanoma.
731
Of the other single-agent targeted therapies tested in prospective trials in
patients with metastatic uveal melanoma (aflibercept,
698
cabozantinib,
687,695
ganetespib,
696
and 9-nitro-camptothecin
699
), responses
were reported only for the HSP90 inhibitor ganetespib, with one response
in the 17 patients tested in the phase II study (ORR of 5.9%).
696
Across
these studies median PFS was less than 6 months for all these
agents.
687,695-699
The combination of everolimus and pasireotide resulted in no responses in
the 13 patients in a phase II trial with metastatic uveal melanoma.
693
Combining bevacizumab with chemotherapy resulted in response rates
between 0% and 6%, with median PFS less than 6 months, and adding
everolimus did not help.
728,732
Combining bevacizumab and interferon
(IFN) alpha yielded similar results.
729
Taken together, most meta-analyses
concluded that targeted therapy did not improve outcomes relative to
conventional chemotherapy.
22,580,587,735
Immunotherapy
Given that immunotherapies have dramatically improved treatment
landscapes for other difficult-to-treat cancers, there is some hope that the
same will be true for uveal melanoma, and a wide variety of
immunotherapies have been tested for treatment of metastatic disease,
including checkpoint immunotherapies, IFN, interleukin-2 (IL-2), vaccines,
and tumor-infiltrating lymphocytes (TILs).
Biochemotherapy
Interferon alpha-2b (IFN alpha-2b) and IL-2 were among the first
immunotherapies tested in prospective trials in metastatic uveal
melanoma, mostly in combination with chemotherapy as part of
biochemotherapy regimens. Two prospective trials of
bleomycin/vincristine/lomustine/dacarbazine (BOLD) chemotherapy in
combination with IFN-alpha-2b yielded different results, with one reporting
no response in 24 patients with uveal melanoma metastasis,
724
and
another reporting four responses among 23 patients (ORR 20%).
727
A third
prospective trial tested BOLD in combination with human leukocyte IFN-
alpha, and reported three responses in the 20 evaluable patients with
metastatic uveal melanoma (ORR 15%).
725
A fourth prospective trial tested
BOLD + INF alpha-2b + IL-2, and reported two responses among 25
patients with metastatic ocular melanoma (ORR 8%), including one
complete response.
726
Results from these studies suggest that
biochemotherapy may provide slightly better response rates than
conventional chemotherapy for patients with metastatic uveal melanoma,
but it is not clear that these regimens improve PFS or OS.
580,587
Checkpoint Immunotherapy
Checkpoint immunotherapies tested in prospective studies for metastatic
uveal melanoma include anti-CTLA-4 agents ipilimumab and
tremelimumab,
704,705,712
anti-PD-1 agents nivolumab and
pembrolizumab,
707-710
and ipilimumab/nivolumab combination
therapy.
711,713
In prospective trials,
704,705,712
expanded access programs (EAPs),
752-757
and
a named patient program,
758
anti-CTLA-4 agents resulted in response
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-47
rates ranging between 0% to 6.5%, median PFS between 2 to 4 months,
and median OS of less than 13 months. Meta-analyses have concluded
that anti-CTLA-4 systemic therapy results in outcomes that are similar or
worse than conventional systemic chemotherapy.
580,587,734
Results for other immune checkpoint inhibitor regimens in patients with
metastatic uveal melanoma look somewhat more promising. Although one
prospective study reported no responses in 17 patients treated with anti-
PD-1 systemic therapy,
708
three other prospective trials and one EAP
reported ORRs between 6% to 38%, with eight responses among a total of
81 patients treated (8%).
707,709,710,759
Median PFS ranged from 2.3 to 11
months.
707-709,759
Two ongoing phase II trials (NCT02626962 and
NCT01585194) testing ipilimumab/nivolumab in patients with metastatic
uveal melanoma have reported ORRs of 15.8% and 17.0%, median PFS
of 5.0 and 6.1 months, and median OS of 19.4 months in one study and
not reached in the other due to insufficient follow-up.
711,713
Comparative
studies are needed to determine whether combination anti-CTLA-4/anti-
PD-1 consistently improves outcomes in patients with metastatic uveal
melanoma.
Other Immunotherapy
Other immunotherapies for which there are efficacy data from prospective
studies in metastatic uveal melanoma include tebentafusp (formerly
IMCgp100),
700,760
dendritic cell vaccination,
702
and adoptive transfer of
TILs.
701
Of these, responses were only seen with TILs adoptive transfer,
with responses in 7 of 20 evaluable patients (ORR 35%).
701
These results
need further investigation.
NCCN Recommendations for Treatment of Distant Metastatic
Disease
Given that there are no treatments for metastatic uveal melanoma that
have clearly and consistently been shown to improve outcomes, it is
important to consider all clinical trial options carefully, and when available
and clinically appropriate, enrollment in a clinical trial is recommended.
For those who are not appropriate for treatment in the context of a clinical
trial, the recommended options are largely based on clinical practice at
NCCN Member Institutions. It is important to be aware that even among
therapies often used at NCCN Member Institutions, efficacy is limited, and
it is not clear which approaches are most effective. Therefore, the
guidelines indicate that a combination of approaches may be needed, and
it is important to consider each patient’s prognosis and treatment goals to
determine whether palliative care is the most appropriate option.
Selection of treatment should depend on the location and extent of
disease. For patients with metastasis to the liver, regionally hepatic-
directed therapies should be considered. Options include: hepatic isolation
perfusion, embolization (ie, chemoembolization, radioembolization,
immunoembolization), and ablation procedures (ie, thermal ablation,
cryotherapy). For patients with extrahepatic disease or hepatic disease
that is not amenable to liver-directed therapy, systemic therapy can be
considered, although there are no systemic therapies that have reliably
improved OS in patients with metastatic uveal melanoma. See below for
recommended systemic therapy options.
For both hepatic and extrahepatic metastases, patients with limited or
symptomatic disease should consider resection and/or RT by photon
beam or SRS. Recommendations for treating uveal melanoma metastases
with RT can be found in the section (above) describing Radiation Therapy
recommended for uveal melanoma, and in the Principles of Radiation for
Metastatic Disease in the NCCN Guidelines for Melanoma: Cutaneous
(available at www.NCCN.org
). Photon beam radiotherapy can be used for
treatment of distant metastases at risk for causing symptoms or for
palliation of symptomatic distant metastases. Dosing for distant
metastases: Doses of 8 to 30 Gy in 1 to 10 fractions should be prescribed
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-48
to the appropriate target volume
761
using appropriate 3-D or intensity-
modulated RT (IMRT) techniques with or without image guidance.
NCCN Recommendations for Systemic Therapy for Metastatic Disease
Given the lack of positive phase III studies, when available and clinically
appropriate, enrollment in a clinical trial is recommended as the preferred
option for systemic therapy. The literature is not directive regarding the
specific systemic agent(s) offering superior outcomes, but does provide
evidence that uveal melanoma is sensitive to some of the same systemic
therapies used to treat cutaneous melanoma. In general, uveal
melanomas have lower response rates than cutaneous melanoma and no
systemic therapies have reliably improved the OS of metastatic uveal
melanoma patients. However, individual patients may derive benefit on
occasion.
Options to consider (other recommended regimens) include select
checkpoint immunotherapies, cytotoxic regimens, and targeted therapy.
Recommended checkpoint immunotherapy options for uveal melanoma
include anti-PD-1 monotherapy with pembrolizumab or nivolumab,
ipilimumab (monotherapy), and combination therapy with nivolumab and
ipilimumab. Treatment-related AEs occur in a high percentage of patients
treated with anti-CTLA-4 or anti-PD-1 agents, and grade 34 related AEs
occur in as many as 20% of patients receiving single-agent therapy and in
~50% receiving ipilimumab monotherapy or nivolumab/ipilimumab
combination therapy. Careful selection of patients and AE monitoring and
management are therefore critical to the safe administration of these
agents. See the NCCN Guidelines for Management of Immunotherapy-
Related Toxicities (available at www.NCCN.org
). Recommended cytotoxic
regimens include dacarbazine, paclitaxel, albumin-bound paclitaxel, or
carboplatin/paclitaxel. The only recommended targeted therapy is
trametinib. For patients being treated with trametinib, see Management of
Toxicities Associated with Targeted Therapy in the NCCN Guidelines for
Melanoma: Cutaneous (available at www.NCCN.org).
NCCN Recommendations for Follow-up and Subsequent Therapy
Following treatment for metastatic disease, patients should receive
imaging to assess response or progression. The recommended cross-
sectional imaging modalities are the same as those recommended for
workup. At minimum, all patients should have contrast-enhanced MR or
US of the liver, with modality preference determined by expertise at the
treating institution. Additional imaging may include chest/abdominal/pelvic
CT with contrast and/or whole-body FDG PET/CT; however, screening
should limit radiation exposure whenever possible. Brain MRI with IV
contrast may be performed if neurologic symptoms are present, but
routine CNS imaging is not recommended. Scans should be performed
with IV contrast unless contraindicated. Those with NED after treatment
for metastases may be eligible for clinical trials testing adjuvant therapies.
If they opt to forgo adjuvant treatment, then the recommended follow-up
surveillance is similar to the follow-up for patients with NED after treatment
of localized disease. See recommendations in the Follow-up section. If
post-treatment imaging shows residual or progressive disease, the NCCN
Panel recommends trying other options for treatment of distant metastatic
disease.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-49
Table 1. Risk Factors for Growth in Small Intraocular Melanocytic Lesions
Risk Factor
Studies Demonstrating Correlation with Tumor Growth
Detection Method(s)
Symptoms Augsburger, 1989
90
; Butler, 1994
95
; Shields, 1995
91
;
Shields, 2000
92
; Singh, 2006
93
; Shields, 2009
94
; Lane, 2010
84
History and physical
Lesion thickness
(>2 mm)
Augsburger, 1989
90
; Butler, 1994
95
; Shields, 1995
91
;
COMS 1997
83
; Shields, 2000
92
; Singh, 2006
93
; Lane, 2010
84
;
Shields, 2009
94
; Dalvin, 2019
96
Ultrasound
Lesion diameter
(>5 mm)
COMS 1997
83
; Dalvin, 2019
96
Comprehensive eye exam, color fundus photography,
ultrasound
Subretinal fluid Shields, 1995
91
; Shields, 2000
92
; Shields, 2009
94
Color fundus photography, comprehensive eye exam
Ancillary option: OCT
Orange pigment
(lipofuscin)
Augsburger, 1989
90
; Butler, 1994
95
; Shields, 1995
91
;
COMS 1997
83
; Shields, 2000
92
; Singh, 2006
93
;
Shields, 2009
94
; Dalvin, 2019
96
Color fundus photography, comprehensive eye exam
Ancillary options: ocular fundus autofluorescence, OCT
Proximity to optic disc
(tumor margin <3 mm)
Augsburger, 1989
90
; Shields, 1995
91
; Shields, 2000
92
;
Shields, 2009
94
Comprehensive eye exam, color fundus photography
Ultrasound hollowness
Shields, 2009
94
; Dalvin, 2019
96
Ultrasound (A- and B-scan)
Absence of halo
Shields, 2009
94
Comprehensive eye exam, color fundus photography
OCT, optical coherence tomography
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-50
Table 2. Biopsy Options for Choroidal or Ciliary Body Tumors
a,231,248
Biopsy Type
a
Surgical
Approach
b
Tumor
c
Sample Analysis
d
Studies Describing Technique, Reporting Yield or Safety
e
(number of patients biopsied)
Fine-needle
aspiration biopsy
(FNAB)
Transscleral Anterior Cytopathology Glasgow, 1988
261
(n=6)
Eide, 1999
251
(n=50)
Shields, 2007
259
(n=73)
Shields, 2007
258
(n=24; 3 mm thick)
McCannel, 2012
246
(n=170)
Chang, 2014
256
(n=38)
Grixti, 2014
234
(n=291)
Sellam, 2016
235
(n=185)
Singh, 2016
233
(n=71)
Kim, 2018
232
(n=11)
Matet, 2019
249
(n=24)
Transvitreal Posterior Cytopathology Glasgow, 1988
261
(n=15)
Eide, 1999
251
(n=14)
Cohen, 2001
250
(n=83)
Augsburger, 2002
88
(n=34; diameter
≤10 mm; thickness, ≥1.5 mm but ≤3 mm)
Shields, 2007
259
(n=67)
Shields, 2007
258
(n=32; 3 mm thick)
Correa, 2014
252
(n=159)
Chang, 2014
256
(n=38)
f
Augsburger, 2015
236
(n=80)
Singh, 2016
233
(n=64)
Sellam, 2016
235
(n=32)
Kim 2017
255
(n=10)
Kim, 2018
232
(n=33)
Reddy 2017
254
(n=57)
f
Singh, 2017
253
(n=20)
Vitrectomy system
(vitreous-cutter)-
assisted biopsy
g
Transvitreal/
transretinal
equatorial
Cytopathology or
Histopathology
(depends on
exact technique
used)
Jensen, 1997
240
(n=92)
Bechrakis, 2002
268
(n=23)
Sen, 2006
267
(n=14)
Bagger, 2013
239
(n=123)
Grixti, 2014
234
(n=448)
Bagger, 2015
266
(n=39)
Nagiel 2017
237
(n=17; 2.0 mm thick)
Grewal, 2017
238
(n=18)
Incisional biopsy
h
with Essen forceps
Transvitreal Anterior Histopathology Akgul, 2011
242
(n=20)
Incisional biopsy
h
(standard forceps)
Transvitreal Posterior Histopathology Kvanta, 2005
243
(n=10)
Seregard, 2013
241
(n=46)
a
Although review articles on techniques for biopsying intraocular lesions include excisional biopsy,
231,248
and historical literature includes reports of transscleral
resection or endoresection being used for uveal melanoma biopsy, these methods are not included in this table because they are no longer commonly used for uveal
melanoma due to technical challenges, risk of complications, and concerns about tumor seeding.
170,272-282
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-51
b
Surgical Approach: Table lists typical surgical approach used for each biopsy method. In procedures using a transscleral (direct) approach, where the tumor is
approached from the outside, with the needle first puncturing the sclera over the tumor, then the tumor itself, leaving the retina intact.
231
Procedures using a
transvitreal (indirect) approach involve anterior entry through the pars plana opposite the tumor, going through the vitreous body and retina to reach the tumor.
231
c
Tumor Location: Although each biopsy method may be used successfully in a range of locations, the feasibility and success rate for each method varies based on
tumor location. The table lists the tumor location(s) for which the biopsy method was developed and/or is most often used.
d
Sample Analysis: Some methods provide cellular aspirate that can be used for cytopathology; others provide tissue samples that can be sectioned for histopathology.
The table reflects the type of sample usually obtained by each of the biopsy methods listed. Both types of samples can be used for molecular analyses for
prognostication.
e
For each biopsy method, the table lists representative studies including at least 10 cases that reported at least one of the following: detailed description of biopsy
technique used, analysis of yield (percent of biopsies providing sufficient material for diagnostic or prognostic analyses), analysis of safety (rates of intraoperative or
postoperative procedure-related complication), or analysis to assess risk of seeding (histologic analyses to detect tracts of tumor cells or follow-up for local
recurrence). Note that inclusion criteria varied across studies listed in the table. Whereas some studies included only patients with suspected or confirmed uveal
melanoma, others included patients with other intraocular conditions.
f
In these studies, biopsy procedure removal of vitreous body (vitrectomy) to reduce the risk of vitreous hemorrhage from transvitreal FNAB.
g
Vitrectomy system (vitreous-cutter)-assisted biopsy: Includes a variety of methods that use vitrectomy tools both to access tumor via a transvitreal/transretinal
approach and to extract tumor tissue using the vitreous cutter and aspiration through the canula. These procedures do not necessarily include a vitrectomy.
h
Incisional biopsy techniques described in the literature sometimes included use of a vitreous cutter, vitrectomy, and other procedures to access the biopsy site,
wherein the tumor tissue was incised with a diamond knife and removed with forceps.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-52
References
1. McLaughlin CC, Wu XC, Jemal A, et al. Incidence of noncutaneous
melanomas in the U.S. Cancer 2005;103:1000-1007. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15651058
.
2. Singh AD, Turell ME, Topham AK. Uveal melanoma: trends in
incidence, treatment, and survival. Ophthalmology 2011;118:1881-1885.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/21704381
.
3. Vajdic CM, Kricker A, Giblin M, et al. Incidence of ocular melanoma in
Australia from 1990 to 1998. Int J Cancer 2003;105:117-122. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/12672041
.
4. Swanson MW, Cloud G. A retrospective analysis of primary eye
cancer at the University of Alabama at Birmingham 1958-1988. Part I:
Eye and orbital cancer. J Am Optom Assoc 1991;62:815-819. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/1813507
.
5. Aronow ME, Topham AK, Singh AD. Uveal Melanoma: 5-Year Update
on Incidence, Treatment, and Survival (SEER 1973-2013). Ocul Oncol
Pathol 2018;4:145-151. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29765944
.
6. Mahendraraj K, Lau CS, Lee I, Chamberlain RS. Trends in incidence,
survival, and management of uveal melanoma: a population-based study
of 7,516 patients from the Surveillance, Epidemiology, and End Results
database (1973-2012). Clin Ophthalmol 2016;10:2113-2119. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/27822007
.
7. Virgili G, Gatta G, Ciccolallo L, et al. Incidence of uveal melanoma in
Europe. Ophthalmology 2007;114:2309-2315. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17498805
.
8. Force AOOT. International Validation of the American Joint Committee
on Cancer's 7th Edition Classification of Uveal Melanoma. JAMA
Ophthalmol 2015;133:376-383. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25555246
.
9. Damato EM, Damato BE. Detection and time to treatment of uveal
melanoma in the United Kingdom: an evaluation of 2,384 patients.
Ophthalmology 2012;119:1582-1589. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22503229
.
10. Mahendraraj K, Shrestha S, Lau CS, Chamberlain RS. Ocular
melanoma-when you have seen one, you have not seen them all: a
clinical outcome study from the Surveillance, Epidemiology and End
Results (SEER) database (1973-2012). Clin Ophthalmol 2017;11:153-
160. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28115829
.
11. Freton A, Chin KJ, Raut R, et al. Initial PET/CT staging for choroidal
melanoma: AJCC correlation and second nonocular primaries in 333
patients. Eur J Ophthalmol 2012;22:236-243. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21959680
.
12. Bagger M, Andersen MT, Andersen KK, et al. The prognostic effect
of American Joint Committee on Cancer staging and genetic status in
patients with choroidal and ciliary body melanoma. Invest Ophthalmol Vis
Sci 2014;56:438-444. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25537201
.
13. Shields CL, Kaliki S, Furuta M, et al. American Joint Committee on
Cancer Classification of Uveal Melanoma (Anatomic Stage) Predicts
Prognosis in 7,731 Patients: The 2013 Zimmerman Lecture.
Ophthalmology 2015;122:1180-1186. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25813452
.
14. The Collaborative Ocular Melanoma Study (COMS) randomized trial
of pre-enucleation radiation of large choroidal melanoma I:
characteristics of patients enrolled and not enrolled. COMS report no. 9.
Am J Ophthalmol 1998;125:767-778. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9645715
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-53
15. Chang AE, Karnell LH, Menck HR. The National Cancer Data Base
report on cutaneous and noncutaneous melanoma: a summary of 84,836
cases from the past decade. The American College of Surgeons
Commission on Cancer and the American Cancer Society. Cancer
1998;83:1664-1678. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9781962
.
16. Polednak AP, Flannery JT. Brain, other central nervous system, and
eye cancer. Cancer 1995;75:330-337. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8001004
.
17. Bishop KD, Olszewski AJ. Epidemiology and survival outcomes of
ocular and mucosal melanomas: a population-based analysis. Int J
Cancer 2014;134:2961-2971. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24272143
.
18. Kujala E, Damato B, Coupland SE, et al. Staging of ciliary body and
choroidal melanomas based on anatomic extent. J Clin Oncol
2013;31:2825-2831. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23816968
.
19. Lorenzo D, Ochoa M, Piulats JM, et al. Prognostic Factors and
Decision Tree for Long-Term Survival in Metastatic Uveal Melanoma.
Cancer Res Treat 2018;50:1130-1139. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29198096
.
20. Kolandjian NA, Wei C, Patel SP, et al. Delayed systemic recurrence
of uveal melanoma. Am J Clin Oncol 2013;36:443-449. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22706174
.
21. Rietschel P, Panageas KS, Hanlon C, et al. Variates of survival in
metastatic uveal melanoma. J Clin Oncol 2005;23:8076-8080. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/16258106
.
22. Khoja L, Atenafu EG, Suciu S, et al. Meta-analysis in metastatic
uveal melanoma to determine progression free and overall survival
benchmarks: an international rare cancers initiative (IRCI) ocular
melanoma study. Ann Oncol 2019;30:1370-1380. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31150059
.
23. Lane AM, Kim IK, Gragoudas ES. Survival Rates in Patients After
Treatment for Metastasis From Uveal Melanoma. JAMA Ophthalmol
2018;136:981-986. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29955797
.
24. Shields CL, Say EAT, Hasanreisoglu M, et al. Personalized
Prognosis of Uveal Melanoma Based on Cytogenetic Profile in 1059
Patients over an 8-Year Period: The 2017 Harry S. Gradle Lecture.
Ophthalmology 2017;124:1523-1531. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28495150
.
25. Isager P, Engholm G, Overgaard J, Storm H. Uveal and conjunctival
malignant melanoma in denmark 1943-97: observed and relative survival
of patients followed through 2002. Ophthalmic Epidemiol 2006;13:85-96.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16581612
.
26. Shields CL, Furuta M, Thangappan A, et al. Metastasis of uveal
melanoma millimeter-by-millimeter in 8033 consecutive eyes. Arch
Ophthalmol 2009;127:989-998. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19667335
.
27. Shields CL, Shields JA, Materin M, et al. Iris melanoma: risk factors
for metastasis in 169 consecutive patients. Ophthalmology
2001;108:172-178. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11150284
.
28. Geisse LJ, Robertson DM. Iris melanomas. Am J Ophthalmol
1985;99:638-648. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/4014387
.
29. Khan S, Finger PT, Yu GP, et al. Clinical and pathologic
characteristics of biopsy-proven iris melanoma: a multicenter
international study. Arch Ophthalmol 2012;130:57-64. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21911649
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-54
30. Shields CL, Di Nicola M, Bekerman VP, et al. Iris Melanoma
Outcomes Based on the American Joint Committee on Cancer
Classification (Eighth Edition) in 432 Patients. Ophthalmology
2018;125:913-923. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29342436
.
31. Amin MB, Edge S, Greene F, et al., eds. AJCC Cancer Staging
Manual (ed 8th). New York: Springer International Publishing; 2017.
32. Henriquez F, Janssen C, Kemp EG, Roberts F. The T1799A BRAF
mutation is present in iris melanoma. Invest Ophthalmol Vis Sci
2007;48:4897-4900. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17962436
.
33. van Poppelen NM, Vaarwater J, Mudhar HS, et al. Genetic
Background of Iris Melanomas and Iris Melanocytic Tumors of Uncertain
Malignant Potential. Ophthalmology 2018;125:904-912. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29371009
.
34. de Lange MJ, Razzaq L, Versluis M, et al. Distribution of GNAQ and
GNA11 Mutation Signatures in Uveal Melanoma Points to a Light
Dependent Mutation Mechanism. PLoS One 2015;10:e0138002.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/26368812
.
35. Onken MD, Worley LA, Long MD, et al. Oncogenic mutations in
GNAQ occur early in uveal melanoma. Invest Ophthalmol Vis Sci
2008;49:5230-5234. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18719078
.
36. Ewens KG, Kanetsky PA, Richards-Yutz J, et al. Chromosome 3
status combined with BAP1 and EIF1AX mutation profiles are associated
with metastasis in uveal melanoma. Invest Ophthalmol Vis Sci
2014;55:5160-5167. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24970262
.
37. Singh AD. Small incision guarded hydroaspiration of iris lesions. Br J
Ophthalmol 2017;101:1570-1575. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28292778
.
38. Harbour JW, Augsburger JJ, Eagle RC, Jr. Initial management and
follow-up of melanocytic iris tumors. Ophthalmology 1995;102:1987-
1993. Available at: https://www.ncbi.nlm.nih.gov/pubmed/9098306
.
39. Conway RM, Chua WC, Qureshi C, Billson FA. Primary iris
melanoma: diagnostic features and outcome of conservative surgical
treatment. Br J Ophthalmol 2001;85:848-854. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11423461
.
40. Klauber S, Jensen PK, Prause JU, Kessing SV. Surgical treatment of
iris and ciliary body melanoma: follow-up of a 25-year series of patients.
Acta Ophthalmol 2012;90:122-126. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20456258
.
41. Kaliki S, Shields CL. Uveal melanoma: relatively rare but deadly
cancer. Eye (Lond) 2017;31:241-257. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27911450
.
42. Riechardt AI, Karle B, Cordini D, et al. Proton therapy of iris
melanoma with 50 CGE : Influence of target volume on clinical outcome.
Strahlenther Onkol 2017;193:943-950. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28631017
.
43. Boyd SR, Gittos A, Richter M, et al. Proton beam therapy and iris
neovascularisation in uveal melanoma. Eye (Lond) 2006;20:832-836.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16167079
.
44. Detorakis ET, Engstrom RE, Jr., Wallace R, Straatsma BR. Iris and
anterior chamber angle neovascularization after iodine 125
brachytherapy for uveal melanoma. Ophthalmology 2005;112:505-510.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/15745782
.
45. Tarlan B, Kiratli H. Uveal Melanoma: Current Trends in Diagnosis
and Management. Turk J Ophthalmol 2016;46:123-137. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27800275
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-55
46. Jovanovic P, Mihajlovic M, Djordjevic-Jocic J, et al. Ocular
melanoma: an overview of the current status. Int J Clin Exp Pathol
2013;6:1230-1244. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23826405
.
47. Mishra KK, Quivey JM, Daftari IK, et al. Long-term Results of the
UCSF-LBNL Randomized Trial: Charged Particle With Helium Ion Versus
Iodine-125 Plaque Therapy for Choroidal and Ciliary Body Melanoma. Int
J Radiat Oncol Biol Phys 2015;92:376-383. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25841624
.
48. Onken MD, Worley LA, Char DH, et al. Collaborative Ocular
Oncology Group report number 1: prospective validation of a multi-gene
prognostic assay in uveal melanoma. Ophthalmology 2012;119:1596-
1603. Available at: https://www.ncbi.nlm.nih.gov/pubmed/22521086
.
49. Yousef YA, Finger PT. Predictive value of the seventh edition
American Joint Committee on Cancer staging system for conjunctival
melanoma. Arch Ophthalmol 2012;130:599-606. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22652846
.
50. Shields CL, Kaliki S, Al-Dahmash SA, et al. American Joint
Committee on Cancer (AJCC) clinical classification predicts conjunctival
melanoma outcomes. Ophthalmic Plast Reconstr Surg 2012;28:313-323.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/22965011
.
51. Van Raamsdonk CD, Bezrookove V, Green G, et al. Frequent
somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature
2009;457:599-602. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19078957
.
52. Soparker CN, O'Brien JM, Albert DM. Investigation of the role of the
ras protooncogene point mutation in human uveal melanomas. Invest
Ophthalmol Vis Sci 1993;34:2203-2209. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8505202
.
53. Zuidervaart W, van Nieuwpoort F, Stark M, et al. Activation of the
MAPK pathway is a common event in uveal melanomas although it rarely
occurs through mutation of BRAF or RAS. Br J Cancer 2005;92:2032-
2038. Available at: https://www.ncbi.nlm.nih.gov/pubmed/15928660
.
54. Rimoldi D, Salvi S, Lienard D, et al. Lack of BRAF mutations in uveal
melanoma. Cancer Res 2003;63:5712-5715. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14522889
.
55. Gear H, Williams H, Kemp EG, Roberts F. BRAF mutations in
conjunctival melanoma. Invest Ophthalmol Vis Sci 2004;45:2484-2488.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/15277467
.
56. Lake SL, Jmor F, Dopierala J, et al. Multiplex ligation-dependent
probe amplification of conjunctival melanoma reveals common BRAF
V600E gene mutation and gene copy number changes. Invest
Ophthalmol Vis Sci 2011;52:5598-5604. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21693616
.
57. Griewank KG, Westekemper H, Murali R, et al. Conjunctival
melanomas harbor BRAF and NRAS mutations and copy number
changes similar to cutaneous and mucosal melanomas. Clin Cancer Res
2013;19:3143-3152. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23633454
.
58. Larsen AC, Dahmcke CM, Dahl C, et al. A Retrospective Review of
Conjunctival Melanoma Presentation, Treatment, and Outcome and an
Investigation of Features Associated With BRAF Mutations. JAMA
Ophthalmol 2015;133:1295-1303. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26425792
.
59. Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations
and copy number in melanoma subtypes. Clin Cancer Res
2008;14:6821-6828. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18980976
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-56
60. Spendlove HE, Damato BE, Humphreys J, et al. BRAF mutations are
detectable in conjunctival but not uveal melanomas. Melanoma Res
2004;14:449-452. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15577314
.
61. Goldenberg-Cohen N, Cohen Y, Rosenbaum E, et al. T1799A BRAF
mutations in conjunctival melanocytic lesions. Invest Ophthalmol Vis Sci
2005;46:3027-3030. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16123397
.
62. Edmunds SC, Cree IA, Di Nicolantonio F, et al. Absence of BRAF
gene mutations in uveal melanomas in contrast to cutaneous
melanomas. Br J Cancer 2003;88:1403-1405. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12778069
.
63. Horkovicova K, Markus J, Krcova I, et al. [the Braf Mutation and the
Possibilities of Uveal Melanoma Metastasing Prognostic Markers
Identification]. Cesk Slov Oftalmol;72:149-156. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27860480
.
64. Bauer J, Kilic E, Vaarwater J, et al. Oncogenic GNAQ mutations are
not correlated with disease-free survival in uveal melanoma. Br J Cancer
2009;101:813-815. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19654573
.
65. Damato B, Dopierala JA, Coupland SE. Genotypic profiling of 452
choroidal melanomas with multiplex ligation-dependent probe
amplification. Clin Cancer Res 2010;16:6083-6092. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20975103
.
66. Koopmans AE, Vaarwater J, Paridaens D, et al. Patient survival in
uveal melanoma is not affected by oncogenic mutations in GNAQ and
GNA11. Br J Cancer 2013;109:493-496. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23778528
.
67. Cassoux N, Rodrigues MJ, Plancher C, et al. Genome-wide profiling
is a clinically relevant and affordable prognostic test in posterior uveal
melanoma. Br J Ophthalmol 2014;98:769-774. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24169649
.
68. Caines R, Eleuteri A, Kalirai H, et al. Cluster analysis of multiplex
ligation-dependent probe amplification data in choroidal melanoma. Mol
Vis 2015;21:1-11. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25684974
.
69. Van Raamsdonk CD, Griewank KG, Crosby MB, et al. Mutations in
GNA11 in uveal melanoma. N Engl J Med 2010;363:2191-2199.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/21083380
.
70. Daniels AB, Lee JE, MacConaill LE, et al. High throughput mass
spectrometry-based mutation profiling of primary uveal melanoma. Invest
Ophthalmol Vis Sci 2012;53:6991-6996. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22977135
.
71. Harbour JW, Onken MD, Roberson ED, et al. Frequent mutation of
BAP1 in metastasizing uveal melanomas. Science 2010;330:1410-1413.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/21051595
.
72. Gupta MP, Lane AM, DeAngelis MM, et al. Clinical Characteristics of
Uveal Melanoma in Patients With Germline BAP1 Mutations. JAMA
Ophthalmol 2015;133:881-887. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25974357
.
73. Harbour JW, Roberson ED, Anbunathan H, et al. Recurrent
mutations at codon 625 of the splicing factor SF3B1 in uveal melanoma.
Nat Genet 2013;45:133-135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23313955
.
74. Martin M, Masshofer L, Temming P, et al. Exome sequencing
identifies recurrent somatic mutations in EIF1AX and SF3B1 in uveal
melanoma with disomy 3. Nat Genet 2013;45:933-936. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23793026
.
75. Qiu M, Shields CL. Choroidal Nevus in the United States Adult
Population: Racial Disparities and Associated Factors in the National
Health and Nutrition Examination Survey. Ophthalmology
2015;122:2071-2083. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26255109
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-57
76. Sumich P, Mitchell P, Wang JJ. Choroidal nevi in a white population:
the Blue Mountains Eye Study. Arch Ophthalmol 1998;116:645-650.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/9596501
.
77. Smith RE, Ganley JP. Ophthalmic survey of a community. 1.
Abnormalities of the ocular fundus. Am J Ophthalmol 1972;74:1126-
1130. Available at: https://www.ncbi.nlm.nih.gov/pubmed/4646717
.
78. Eskelin S, Kivela T. Mode of presentation and time to treatment of
uveal melanoma in Finland. Br J Ophthalmol 2002;86:333-338. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/11864894
.
79. Shields CL, Pefkianaki M, Mashayekhi A, et al. Cytogenetic results of
choroidal nevus growth into melanoma in 55 consecutive cases. Saudi J
Ophthalmol 2018;32:28-32. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29755268
.
80. Kivela T. Diagnosis of uveal melanoma. Dev Ophthalmol 2012;49:1-
15. Available at: https://www.ncbi.nlm.nih.gov/pubmed/22042009
.
81. Singh AD, Kalyani P, Topham A. Estimating the risk of malignant
transformation of a choroidal nevus. Ophthalmology 2005;112:1784-
1789. Available at: https://www.ncbi.nlm.nih.gov/pubmed/16154197
.
82. Ganley JP, Comstock GW. Benign nevi and malignant melanomas of
the choroid. Am J Ophthalmol 1973;76:19-25. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/4717339
.
83. Factors predictive of growth and treatment of small choroidal
melanoma: COMS Report No. 5. The Collaborative Ocular Melanoma
Study Group. Arch Ophthalmol 1997;115:1537-1544. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9400787
.
84. Lane AM, Egan KM, Kim IK, Gragoudas ES. Mortality after diagnosis
of small melanocytic lesions of the choroid. Arch Ophthalmol
2010;128:996-1000. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20696999
.
85. Li HK, Shields CL, Mashayekhi A, et al. Giant choroidal nevus clinical
features and natural course in 322 cases. Ophthalmology 2010;117:324-
333. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19969359
.
86. Shields CL, Dalvin LA, Yu MD, et al. Choroidal nevus transformation
into melanoma per millimeter increment in thickness using multimodal
imaging in 2355 cases: The 2019 Wendell L. Hughes Lecture. Retina
2019;39:1852-1860. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30883534
.
87. Shields CL, Dalvin LA, Ancona-Lezama D, et al. Choroidal nevus
imaging features in 3,806 cases and risk factors for transformation in
melanoma in 2,355 cases: The 2020 Taylor R. Smith and Victor T. Curtin
Lecture. Retina 2019;39:1840-1851. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30608349
.
88. Augsburger JJ, Correa ZM, Schneider S, et al. Diagnostic transvitreal
fine-needle aspiration biopsy of small melanocytic choroidal tumors in
nevus versus melanoma category. Trans Am Ophthalmol Soc
2002;100:225-232; discussion 232-224. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12545696
.
89. Augsburger JJ, Correa ZM, Trichopoulos N, Shaikh A. Size overlap
between benign melanocytic choroidal nevi and choroidal malignant
melanomas. Invest Ophthalmol Vis Sci 2008;49:2823-2828. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18408179
.
90. Augsburger JJ, Schroeder RP, Territo C, et al. Clinical parameters
predictive of enlargement of melanocytic choroidal lesions. Br J
Ophthalmol 1989;73:911-917. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2605146
.
91. Shields CL, Shields JA, Kiratli H, et al. Risk factors for growth and
metastasis of small choroidal melanocytic lesions. Ophthalmology
1995;102:1351-1361. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9097773
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-58
92. Shields CL, Cater J, Shields JA, et al. Combination of clinical factors
predictive of growth of small choroidal melanocytic tumors. Arch
Ophthalmol 2000;118:360-364. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10721958
.
93. Singh AD, Mokashi AA, Bena JF, et al. Small choroidal melanocytic
lesions: features predictive of growth. Ophthalmology 2006;113:1032-
1039. Available at: https://www.ncbi.nlm.nih.gov/pubmed/16650475
.
94. Shields CL, Furuta M, Berman EL, et al. Choroidal nevus
transformation into melanoma: analysis of 2514 consecutive cases. Arch
Ophthalmol 2009;127:981-987. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19667334
.
95. Butler P, Char DH, Zarbin M, Kroll S. Natural history of indeterminate
pigmented choroidal tumors. Ophthalmology 1994;101:710-716;
discussion 717. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8152767
.
96. Dalvin LA, Shields CL, Ancona-Lezama DA, et al. Combination of
multimodal imaging features predictive of choroidal nevus transformation
into melanoma. Br J Ophthalmol 2019;103:1441-1447. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30523045
.
97. Mims JL, 3rd, Shields JA. Follow-up studies of suspicious choroidal
nevi. Ophthalmology 1978;85:929-943. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/733186
.
98. MacIlwaine WAt, Anderson B, Jr., Klintworth GK. Enlargement of a
histologically documented choroidal nevus. Am J Ophthalmol
1979;87:480-486. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/443311
.
99. Mashayekhi A, Siu S, Shields CL, Shields JA. Slow enlargement of
choroidal nevi: a long-term follow-up study. Ophthalmology
2011;118:382-388. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20801518
.
100. Dalvin LA, Shields CL, Pulido JS, et al. Uveal Melanoma Associated
With Myotonic Dystrophy: A Report of 6 Cases. JAMA Ophthalmol
2018;136:543-547. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29596556
.
101. Velazquez-Martin JP, Pavlin CJ, Simpson ER. Association between
uveal melanoma and myotonic dystrophy: a series of 3 cases. JAMA
Ophthalmol 2013;131:246-249. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23411896
.
102. Win AK, Perattur PG, Pulido JS, et al. Increased cancer risks in
myotonic dystrophy. Mayo Clin Proc 2012;87:130-135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22237010
.
103. Fontcuberta IC, Salomao DR, Quiram PA, Pulido JS. Choroidal
melanoma and lid fibrofoliculomas in Birt-Hogg-Dube syndrome.
Ophthalmic Genet 2011;32:143-146. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21401403
.
104. Marous CL, Marous MR, Welch RJ, et al. Choroidal Melanoma,
Sector Melanocytosis, and Retinal Pigment Epithelial Microdetachments
in Birt-Hogg-Dube Syndrome. Retin Cases Brief Rep 2019;13:202-206.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28541267
.
105. Spees EK, Jr., Kramer KK, Light JA, et al. Reactivation of ocular
malignant melanoma after renal transplantation. Transplantation
1980;29:421-424. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/6990569
.
106. Shields JA, Narsipur SS, Shields CL, Sperber DE. Choroidal
melanoma in an immunosuppressed child with minimal change nephrotic
syndrome. Retina 2004;24:454-455. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15187671
.
107. Singh AD, De Potter P, Fijal BA, et al. Lifetime prevalence of uveal
melanoma in white patients with oculo(dermal) melanocytosis.
Ophthalmology 1998;105:195-198. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9442799
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-59
108. Gonder JR, Shields JA, Albert DM, et al. Uveal malignant
melanoma associated with ocular and oculodermal melanocytosis.
Ophthalmology 1982;89:953-960. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7133641
.
109. Singh AD, Shields CL, Shields JA, Sato T. Uveal melanoma in
young patients. Arch Ophthalmol 2000;118:918-923. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10900104
.
110. Velazquez N, Jones IS. Ocular and oculodermal melanocytosis
associated with uveal melanoma. Ophthalmology 1983;90:1472-1476.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/6677847
.
111. Gonder JR, Ezell PC, Shields JA, Augsburger JJ. Ocular
melanocytosis. A study to determine the prevalence rate of ocular
melanocytosis. Ophthalmology 1982;89:950-952. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7133640
.
112. Shields CL, Kaliki S, Livesey M, et al. Association of ocular and
oculodermal melanocytosis with the rate of uveal melanoma metastasis:
analysis of 7872 consecutive eyes. JAMA Ophthalmol 2013;131:993-
1003. Available at: https://www.ncbi.nlm.nih.gov/pubmed/23681424
.
113. Singh AD, Shields CL, De Potter P, et al. Familial uveal melanoma.
Clinical observations on 56 patients. Arch Ophthalmol 1996;114:392-
399. Available at: https://www.ncbi.nlm.nih.gov/pubmed/8602775
.
114. Abdel-Rahman MH, Pilarski R, Ezzat S, et al. Cancer family history
characterization in an unselected cohort of 121 patients with uveal
melanoma. Fam Cancer 2010;9:431-438. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20157784
.
115. Singh AD, Wang MX, Donoso LA, et al. Familial uveal melanoma,
III. Is the occurrence of familial uveal melanoma coincidental? Arch
Ophthalmol 1996;114:1101-1104. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8790096
.
116. Masoomian B, Shields JA, Shields CL. Overview of BAP1 cancer
predisposition syndrome and the relationship to uveal melanoma. J Curr
Ophthalmol 2018;30:102-109. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29988936
.
117. Helgadottir H, Hoiom V. The genetics of uveal melanoma: current
insights. Appl Clin Genet 2016;9:147-155. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27660484
.
118. Wang A, Papneja A, Hyrcza M, et al. Gene of the month: BAP1. J
Clin Pathol 2016;69:750-753. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27235536
.
119. Harbour JW. The genetics of uveal melanoma: an emerging
framework for targeted therapy. Pigment Cell Melanoma Res
2012;25:171-181. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22268848
.
120. Carbone M, Ferris LK, Baumann F, et al. BAP1 cancer syndrome:
malignant mesothelioma, uveal and cutaneous melanoma, and MBAITs.
J Transl Med 2012;10:179. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22935333
.
121. Foretova L, Navratilova M, Svoboda M, et al. BAP1 Syndrome -
Predisposition to Malignant Mesothelioma, Skin and Uveal Melanoma,
Renal and Other Cancers. Klin Onkol 2019;32:118-122. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31409087
.
122. Boru G, Grosel TW, Pilarski R, et al. Germline large deletion of
BAP1 and decreased expression in non-tumor choroid in uveal
melanoma patients with high risk for inherited cancer. Genes
Chromosomes Cancer 2019;58:650-656. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30883995
.
123. Haugh AM, Njauw CN, Bubley JA, et al. Genotypic and Phenotypic
Features of BAP1 Cancer Syndrome: A Report of 8 New Families and
Review of Cases in the Literature. JAMA Dermatol 2017;153:999-1006.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28793149
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-60
124. Pilarski R, Cebulla CM, Massengill JB, et al. Expanding the clinical
phenotype of hereditary BAP1 cancer predisposition syndrome, reporting
three new cases. Genes Chromosomes Cancer 2014;53:177-182.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/24243779
.
125. Walpole S, Pritchard AL, Cebulla CM, et al. Comprehensive Study
of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families
Worldwide. J Natl Cancer Inst 2018;110:1328-1341. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30517737
.
126. Pilarski R, Carlo M, Cebulla C, Abdel-Rahman M. BAP1 Tumor
Predisposition Syndrome. In: Adam MP, Ardinger HH, Pagon RA, et al.,
eds. GeneReviews((R)). Seattle (WA); 1993.
127. Wiesner T, Obenauf AC, Murali R, et al. Germline mutations in
BAP1 predispose to melanocytic tumors. Nat Genet 2011;43:1018-1021.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/21874003
.
128. O'Shea SJ, Robles-Espinoza CD, McLellan L, et al. A population-
based analysis of germline BAP1 mutations in melanoma. Hum Mol
Genet 2017;26:717-728. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28062663
.
129. Rai K, Pilarski R, Cebulla CM, Abdel-Rahman MH. Comprehensive
review of BAP1 tumor predisposition syndrome with report of two new
cases. Clin Genet 2016;89:285-294. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26096145
.
130. Carbone M, Flores EG, Emi M, et al. Combined Genetic and
Genealogic Studies Uncover a Large BAP1 Cancer Syndrome Kindred
Tracing Back Nine Generations to a Common Ancestor from the 1700s.
PLoS Genet 2015;11:e1005633. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26683624
.
131. Njauw CN, Kim I, Piris A, et al. Germline BAP1 inactivation is
preferentially associated with metastatic ocular melanoma and
cutaneous-ocular melanoma families. PLoS One 2012;7:e35295.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/22545102
.
132. Testa JR, Cheung M, Pei J, et al. Germline BAP1 mutations
predispose to malignant mesothelioma. Nat Genet 2011;43:1022-1025.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/21874000
.
133. Abdel-Rahman MH, Sample KM, Pilarski R, et al. Whole Exome
Sequencing Identifies Candidate Genes Associated with Hereditary
Predisposition to Uveal Melanoma. Ophthalmology 2020;127:668-678.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/32081490
.
134. Derrien AC, Rodrigues M, Eeckhoutte A, et al. Germline MBD4
Mutations and Predisposition to Uveal Melanoma. J Natl Cancer Inst
2021;113:80-87. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32239153
.
135. Rodrigues M, Mobuchon L, Houy A, et al. Evolutionary Routes in
Metastatic Uveal Melanomas Depend on MBD4 Alterations. Clin Cancer
Res 2019;25:5513-5524. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31227496
.
136. Repo P, Jantti JE, Jarvinen RS, et al. Germline loss-of-function
variants in MBD4 are rare in Finnish patients with uveal melanoma.
Pigment Cell Melanoma Res 2020;33:756-762. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32421892
.
137. Rodrigues M, Mobuchon L, Houy A, et al. Outlier response to anti-
PD1 in uveal melanoma reveals germline MBD4 mutations in
hypermutated tumors. Nat Commun 2018;9:1866. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29760383
.
138. Mataftsi A, Zografos L, Chamot L, Schalenbourg A. [Choroidal
melanoma in neurofibromatosis type 2: description of a case]. J Fr
Ophtalmol 2003;26:477-480. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12819605
.
139. Friedman SM, Margo CE. Choroidal melanoma and
neurofibromatosis type 1. Arch Ophthalmol 1998;116:694-695. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/9596516
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-61
140. Antle CM, Damji KF, White VA, Rootman J. Uveal malignant
melanoma and optic nerve glioma in von Recklinghausen's
neurofibromatosis. Br J Ophthalmol 1990;74:502-504. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2117971
.
141. Specht CS, Smith TW. Uveal malignant melanoma and von
Recklinghausen's neurofibromatosis. Cancer 1988;62:812-817. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/3135111
.
142. Croxatto JO, Charles DE, Malbran ES. Neurofibromatosis
associated with nevus of Ota and choroidal melanoma. Am J Ophthalmol
1981;92:578-580. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/6794373
.
143. Wiznia RA, Freedman JK, Mancini AD, Shields JA. Malignant
melanoma of the choroid in neurofibromatosis. Am J Ophthalmol
1978;86:684-687. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/102202
.
144. Nordmann J, Brini A. Von Recklinghausen's disease and melanoma
of the uvea. Br J Ophthalmol 1970;54:641-648. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/4990591
.
145. Yanoff M, Zimmerman LE. Histogenesis of malignant melanomas of
the uvea. 3. The relationship of congenital ocular melanocytosis and
neurofibromatosis in uveal melanomas. Arch Ophthalmol 1967;77:331-
336. Available at: https://www.ncbi.nlm.nih.gov/pubmed/6019552
.
146. Honavar SG, Singh AD, Shields CL, et al. Iris melanoma in a patient
with neurofibromatosis. Surv Ophthalmol 2000;45:231-236. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11094247
.
147. Nayman T, Bostan C, Logan P, Burnier MN, Jr. Uveal Melanoma
Risk Factors: A Systematic Review of Meta-Analyses. Curr Eye Res
2017;42:1085-1093. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28494168
.
148. Weis E, Shah CP, Lajous M, et al. The association of cutaneous
and iris nevi with uveal melanoma: a meta-analysis. Ophthalmology
2009;116:536-543 e532. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19167086
.
149. Smith JH, Padnick-Silver L, Newlin A, et al. Genetic study of familial
uveal melanoma: association of uveal and cutaneous melanoma with
cutaneous and ocular nevi. Ophthalmology 2007;114:774-779. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/17207529
.
150. van Hees CL, de Boer A, Jager MJ, et al. Are atypical nevi a risk
factor for uveal melanoma? A case-control study. J Invest Dermatol
1994;103:202-205. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8040610
.
151. Richtig E, Langmann G, Mullner K, Smolle J. Ocular melanoma:
epidemiology, clinical presentation and relationship with dysplastic nevi.
Ophthalmologica 2004;218:111-114. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15004500
.
152. Seddon JM, Gragoudas ES, Glynn RJ, et al. Host factors, UV
radiation, and risk of uveal melanoma. A case-control study. Arch
Ophthalmol 1990;108:1274-1280. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2400347
.
153. Bataille V, Pinney E, Hungerford JL, et al. Five cases of coexistent
primary ocular and cutaneous melanoma. Arch Dermatol 1993;129:198-
201. Available at: https://www.ncbi.nlm.nih.gov/pubmed/8434978
.
154. Bataille V, Sasieni P, Cuzick J, et al. Risk of ocular melanoma in
relation to cutaneous and iris naevi. Int J Cancer 1995;60:622-626.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/7860135
.
155. van Hees CL, Jager MJ, Bleeker JC, et al. Occurrence of cutaneous
and uveal melanoma in patients with uveal melanoma and their first
degree relatives. Melanoma Res 1998;8:175-180. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9610873
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-62
156. Lischko AM, Seddon JM, Gragoudas ES, et al. Evaluation of prior
primary malignancy as a determinant of uveal melanoma. A case-control
study. Ophthalmology 1989;96:1716-1721. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2622616
.
157. Shors AR, Iwamoto S, Doody DR, Weiss NS. Relationship of uveal
and cutaneous malignant melanoma in persons with multiple primary
tumors. Int J Cancer 2002;102:266-268. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12397648
.
158. Hemminki K, Zhang H, Czene K. Association of first ocular
melanoma with subsequent cutaneous melanoma: results from the
Swedish Family-Cancer Database. Int J Cancer 2003;104:257-258.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/12569585
.
159. Scelo G, Boffetta P, Autier P, et al. Associations between ocular
melanoma and other primary cancers: an international population-based
study. Int J Cancer 2007;120:152-159. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17036322
.
160. Bergman L, Nilsson B, Ragnarsson-Olding B, Seregard S. Uveal
melanoma: a study on incidence of additional cancers in the Swedish
population. Invest Ophthalmol Vis Sci 2006;47:72-77. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16384946
.
161. Spanogle JP, Clarke CA, Aroner S, Swetter SM. Risk of second
primary malignancies following cutaneous melanoma diagnosis: a
population-based study. J Am Acad Dermatol 2010;62:757-767.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/20223559
.
162. Lains I, Bartosch C, Mondim V, et al. Second Primary Neoplasms in
Patients With Uveal Melanoma: A SEER Database Analysis. Am J
Ophthalmol 2016;165:54-64. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26940164
.
163. Swerdlow AJ, Storm HH, Sasieni PD. Risks of second primary
malignancy in patients with cutaneous and ocular melanoma in Denmark,
1943-1989. Int J Cancer 1995;61:773-779. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7790110
.
164. Damato B. Detection of uveal melanoma by optometrists in the
United Kingdom. Ophthalmic Physiol Opt 2001;21:268-271. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11430620
.
165. Ah-Fat FG, Damato BE. Delays in the diagnosis of uveal melanoma
and effect on treatment. Eye (Lond) 1998;12 ( Pt 5):781-782. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/10070508
.
166. Vidal JL, Bacin F, Albuisson E, et al. ["Melanoma 92".
epidemiological study of uveal melanoma in France]. J Fr Ophtalmol
1995;18:520-528. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7560797
.
167. Accuracy of diagnosis of choroidal melanomas in the Collaborative
Ocular Melanoma Study. COMS report no. 1. Arch Ophthalmol
1990;108:1268-1273. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2205183
.
168. Collaborative Ocular Melanoma Study G, Boldt HC, Byrne SF, et al.
Baseline echographic characteristics of tumors in eyes of patients
enrolled in the Collaborative Ocular Melanoma Study: COMS report no.
29. Ophthalmology 2008;115:1390-1397, 1397 e1391-1392. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18267342
.
169. Bove R, Char DH. Nondiagnosed uveal melanomas. Ophthalmology
2004;111:554-557. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15019335
.
170. Grisanti S, Tura A. Uveal Melanoma. In: Scott JF, Gerstenblith MR,
eds. Noncutaneous Melanoma. Brisbane (AU): Codon Publications;
2018.
171. Damato B. Progress in the management of patients with uveal
melanoma. The 2012 Ashton Lecture. Eye (Lond) 2012;26:1157-1172.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/22744385
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-63
172. Demirci H, Shields CL, Shields JA, et al. Ring melanoma of the
ciliary body: report on twenty-three patients. Retina 2002;22:698-706;
quiz 852-693. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12476094
.
173. Shields CL, Kaliki S, Furuta M, et al. Clinical spectrum and
prognosis of uveal melanoma based on age at presentation in 8,033
cases. Retina 2012;32:1363-1372. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22466491
.
174. Shields JA, Augsburger JJ, Brown GC, Stephens RF. The
differential diagnosis of posterior uveal melanoma. Ophthalmology
1980;87:518-522. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7413140
.
175. Shields JA, Mashayekhi A, Ra S, Shields CL. Pseudomelanomas of
the posterior uveal tract: the 2006 Taylor R. Smith Lecture. Retina
2005;25:767-771. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16141866
.
176. Ghassemi F, Bazvand F, Hosseini SS. Pseudomelanoma at a
referral center in iran. J Ophthalmic Vis Res 2014;9:50-53. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24982732
.
177. Saari JM, Kivela T, Summanen P, et al. Digital imaging in
differential diagnosis of small choroidal melanoma. Graefes Arch Clin
Exp Ophthalmol 2006;244:1581-1590. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16636839
.
178. Freedman MI, Folk JC. Metastatic tumors to the eye and orbit.
Patient survival and clinical characteristics. Arch Ophthalmol
1987;105:1215-1219. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3307718
.
179. Shields CL, Shields JA, Gross NE, et al. Survey of 520 eyes with
uveal metastases. Ophthalmology 1997;104:1265-1276. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9261313
.
180. Shields CL, Welch RJ, Malik K, et al. Uveal Metastasis: Clinical
Features and Survival Outcome of 2214 Tumors in 1111 Patients Based
on Primary Tumor Origin. Middle East Afr J Ophthalmol 2018;25:81-90.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/30122853
.
181. Nathan P, Cohen V, Coupland S, et al. Uveal Melanoma UK
National Guidelines. Eur J Cancer 2015;51:2404-2412. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26278648
.
182. Golebiewska J, Kecik D, Turczynska M, et al. Optical coherence
tomography in diagnosing, differentiating and monitoring of choroidal
nevi - 1 year observational study. Neuro Endocrinol Lett 2013;34:539-
542. Available at: https://www.ncbi.nlm.nih.gov/pubmed/24378457
.
183. Muller K, Nowak PJ, de Pan C, et al. Effectiveness of fractionated
stereotactic radiotherapy for uveal melanoma. Int J Radiat Oncol Biol
Phys 2005;63:116-122. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16111579
.
184. Heilemann G, Fetty L, Blaickner M, et al. Retina dose as a predictor
for visual acuity loss in (106)Ru eye plaque brachytherapy of uveal
melanomas. Radiother Oncol 2018;127:379-384. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29452902
.
185. American Brachytherapy Society - Ophthalmic Oncology Task
Force. Electronic address pec, Committee AO. The American
Brachytherapy Society consensus guidelines for plaque brachytherapy of
uveal melanoma and retinoblastoma. Brachytherapy 2014;13:1-14.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/24373763
.
186. Melia BM, Abramson DH, Albert DM, et al. Collaborative ocular
melanoma study (COMS) randomized trial of I-125 brachytherapy for
medium choroidal melanoma. I. Visual acuity after 3 years COMS report
no. 16. Ophthalmology 2001;108:348-366. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11158813
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-64
187. Jampol LM, Moy CS, Murray TG, et al. The COMS randomized trial
of iodine 125 brachytherapy for choroidal melanoma: IV. Local treatment
failure and enucleation in the first 5 years after brachytherapy. COMS
report no. 19. Ophthalmology 2002;109:2197-2206. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12466159
.
188. Dieckmann K, Georg D, Zehetmayer M, et al. Stereotactic photon
beam irradiation of uveal melanoma: indications and experience at the
University of Vienna since 1997. Strahlenther Onkol 2007;183 Spec No
2:11-13. Available at: https://www.ncbi.nlm.nih.gov/pubmed/18166998
.
189. Muscat S, Parks S, Kemp E, Keating D. Secondary retinal changes
associated with choroidal naevi and melanomas documented by optical
coherence tomography. Br J Ophthalmol 2004;88:120-124. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14693788
.
190. Muftuoglu IK, Gaber R, Bartsch DU, et al. Comparison of
conventional color fundus photography and multicolor imaging in
choroidal or retinal lesions. Graefes Arch Clin Exp Ophthalmol
2018;256:643-649. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29492687
.
191. Albertus DL, Schachar IH, Zahid S, et al. Autofluorescence
quantification of benign and malignant choroidal nevomelanocytic
tumors. JAMA Ophthalmol 2013;131:1004-1008. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23787920
.
192. Garcia MD, Salomao DR, Marmorstein AD, Pulido JS.
Histopathologic Findings in the Areas of Orange Pigment Overlying
Choroidal Melanomas. Transl Vis Sci Technol 2016;5:4. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27190699
.
193. Meyer K, Augsburger JJ. Independent diagnostic value of
fluorescein angiography in the evaluation of intraocular tumors. Graefes
Arch Clin Exp Ophthalmol 1999;237:489-494. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10379610
.
194. Skalicky SE, Holt PE, Giblin M, et al. Australian Cancer Network
clinical practice guidelines for the management of ocular and periocular
melanoma: an evidence-based literature analysis. Clin Exp Ophthalmol
2008;36:646-658. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18983550
.
195. Yonekawa Y, Kim IK. Epidemiology and management of uveal
melanoma. Hematol Oncol Clin North Am 2012;26:1169-1184. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/23116575
.
196. Chattopadhyay C, Kim DW, Gombos DS, et al. Uveal melanoma:
From diagnosis to treatment and the science in between. Cancer
2016;122:2299-2312. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26991400
.
197. Fuller DG, Snyder WB, Hutton WL, Vaiser A. Ultrasonographic
features of choroidal malignant melanomas. Arch Ophthalmol
1979;97:1465-1472. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/464871
.
198. Bedi DG, Gombos DS, Ng CS, Singh S. Sonography of the eye.
AJR Am J Roentgenol 2006;187:1061-1072. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16985158
.
199. Delgado S, Rodriguez Reyes A, Mora Rios L, et al. Ultrasound,
histopathological, and genetic features of uveal melanoma in a Mexican-
Mestizo population. Arch Soc Esp Oftalmol (Engl Ed) 2018;93:15-21.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28728954
.
200. Collaborative Ocular Melanoma Study G. Comparison of clinical,
echographic, and histopathological measurements from eyes with
medium-sized choroidal melanoma in the collaborative ocular melanoma
study: COMS report no. 21. Arch Ophthalmol 2003;121:1163-1171.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/12912695
.
201. Ossoinig KC. Standardized echography: basic principles, clinical
applications, and results. Int Ophthalmol Clin 1979;19:127-210. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/395120
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-65
202. Sobottka B, Schlote T, Krumpaszky HG, Kreissig I. Choroidal
metastases and choroidal melanomas: comparison of ultrasonographic
findings. Br J Ophthalmol 1998;82:159-161. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9613382
.
203. Materin MA, Raducu R, Bianciotto C, Shields CL. Fundus
autofluorescence and optical coherence tomography findings in choroidal
melanocytic lesions. Middle East Afr J Ophthalmol 2010;17:201-206.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/20844674
.
204. Muskens RP, Van Best JA, Bleeker JC, Keunen JE. Corneal
autofluorescence in choroidal melanoma or in choroidal naevus. Br J
Ophthalmol 2001;85:662-665. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11371483
.
205. Shields CL, Bianciotto C, Pirondini C, et al. Autofluorescence of
choroidal melanoma in 51 cases. Br J Ophthalmol 2008;92:617-622.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/18441171
.
206. Cennamo G, Romano MR, Velotti N, et al. Fundus autofluorescence
of choroidal nevi and melanoma. Acta Ophthalmol 2018;96:e102-e104.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28205424
.
207. Gunduz K, Pulido JS, Ezzat K, et al. Review of fundus
autofluorescence in choroidal melanocytic lesions. Eye (Lond)
2009;23:497-503. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18670456
.
208. Lavinsky D, Belfort RN, Navajas E, et al. Fundus autofluorescence
of choroidal nevus and melanoma. Br J Ophthalmol 2007;91:1299-1302.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/17431017
.
209. Gunduz K, Pulido JS, Bakri SJ, Petit-Fond E. Fundus
autofluorescence in choroidal melanocytic lesions. Retina 2007;27:681-
687. Available at: https://www.ncbi.nlm.nih.gov/pubmed/17621175
.
210. Shields CL, Pirondini C, Bianciotto C, et al. Autofluorescence of
choroidal nevus in 64 cases. Retina 2008;28:1035-1043. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18779708
.
211. Salvanos P, Utheim TP, Moe MC, et al. Autofluorescence imaging
in the differential diagnosis of optic disc melanocytoma. Acta Ophthalmol
2015;93:476-480. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25923254
.
212. Singh AD, Belfort RN, Sayanagi K, Kaiser PK. Fourier domain
optical coherence tomographic and auto-fluorescence findings in
indeterminate choroidal melanocytic lesions. Br J Ophthalmol
2010;94:474-478. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19822920
.
213. Krema H, Habal S, Gonzalez JE, Pavlin CJ. Role of optical
coherence tomography in verifying the specificity of ultrasonography in
detecting subtle subretinal fluid associated with small choroidal
melanocytic tumors. Retina 2014;34:360-365. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23807190
.
214. Shah SU, Kaliki S, Shields CL, et al. Enhanced depth imaging
optical coherence tomography of choroidal nevus in 104 cases.
Ophthalmology 2012;119:1066-1072. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22297027
.
215. Shields CL, Kaliki S, Rojanaporn D, et al. Enhanced depth imaging
optical coherence tomography of small choroidal melanoma: comparison
with choroidal nevus. Arch Ophthalmol 2012;130:850-856. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22776921
.
216. Filloy A, Caminal JM, Arias L, et al. Swept source optical coherence
tomography imaging of a series of choroidal tumours. Can J Ophthalmol
2015;50:242-248. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26040226
.
217. Espinoza G, Rosenblatt B, Harbour JW. Optical coherence
tomography in the evaluation of retinal changes associated with
suspicious choroidal melanocytic tumors. Am J Ophthalmol 2004;137:90-
95. Available at: https://www.ncbi.nlm.nih.gov/pubmed/14700649
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-66
218. Sayanagi K, Pelayes DE, Kaiser PK, Singh AD. 3D Spectral domain
optical coherence tomography findings in choroidal tumors. Eur J
Ophthalmol 2011;21:271-275. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21038307
.
219. Leshno A, Vishnevskia-Dai V, Barak A, et al. Optical Density Ratio
of the Subretinal Fluid in Choroidal Melanoma and Metastasis. Retina
2019;39:685-691. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29252972
.
220. Toledo JJ, Asencio M, Garcia JR, et al. OCT Angiography: Imaging
of Choroidal and Retinal Tumors. Ophthalmol Retina 2018;2:613-622.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/31047615
.
221. Garcia-Arumi Fuste C, Peralta Iturburu F, Garcia-Arumi J. Is optical
coherence tomography angiography helpful in the differential diagnosis
of choroidal nevus versus melanoma? Eur J Ophthalmol 2020;30:723-
729. Available at: https://www.ncbi.nlm.nih.gov/pubmed/31210061
.
222. Bianciotto C, Shields CL, Guzman JM, et al. Assessment of anterior
segment tumors with ultrasound biomicroscopy versus anterior segment
optical coherence tomography in 200 cases. Ophthalmology
2011;118:1297-1302. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21377736
.
223. Augsburger JJ, Golden MI, Shields JA. Fluorescein angiography of
choroidal malignant melanomas with retinal invasion. Retina 1984;4:232-
241. Available at: https://www.ncbi.nlm.nih.gov/pubmed/6531518
.
224. Sallet G, Amoaku WM, Lafaut BA, et al. Indocyanine green
angiography of choroidal tumors. Graefes Arch Clin Exp Ophthalmol
1995;233:677-689. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8566823
.
225. Shields CL, Pellegrini M, Kligman BE, et al. Ciliary body and
choroidal pseudomelanoma from ultrasonographic imaging of
hypermature cataract in 20 cases. Ophthalmology 2013;120:2546-2551.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/23916484
.
226. Polivogianis L, Seddon JM, Glynn RJ, et al. Comparison of
transillumination and histologic slide measurements of tumor diameter in
uveal melanoma. Ophthalmology 1988;95:1576-1582. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3062526
.
227. Recsan Z, Karlinger K, Fodor M, et al. MRI for the evaluation of
scleral invasion and extrascleral extension of uveal melanomas. Clin
Radiol 2002;57:371-376. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12014934
.
228. Wang Y, Li YY, Su F, Xiao LH. [Clinical observation of extraocular
extension of choroidal melanoma]. Zhonghua Yan Ke Za Zhi
2011;47:242-247. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21609626
.
229. Nguyen HG, Sznitman R, Maeder P, et al. Personalized Anatomic
Eye Model From T1-Weighted Volume Interpolated Gradient Echo
Magnetic Resonance Imaging of Patients With Uveal Melanoma. Int J
Radiat Oncol Biol Phys 2018;102:813-820. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29970318
.
230. Zoberi JE, Garcia-Ramirez J, Hedrick S, et al. MRI-based treatment
planning and dose delivery verification for intraocular melanoma
brachytherapy. Brachytherapy 2018;17:31-39. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28818442
.
231. Bagger MM. Intraocular biopsy of uveal melanoma Risk assessment
and identification of genetic prognostic markers. Acta Ophthalmol
2018;96 Suppl A112:1-28. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30133961
.
232. Kim RS, Chevez-Barrios P, Divatia M, et al. Yield, Techniques, and
Complications of Transvitreal and Transscleral Biopsies in Small Uveal
Melanoma. JAMA Ophthalmol 2018;136:482-488. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29596552
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-67
233. Singh AD, Medina CA, Singh N, et al. Fine-needle aspiration biopsy
of uveal melanoma: outcomes and complications. Br J Ophthalmol
2016;100:456-462. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26231747
.
234. Grixti A, Angi M, Damato BE, et al. Vitreoretinal surgery for
complications of choroidal tumor biopsy. Ophthalmology 2014;121:2482-
2488. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25085629
.
235. Sellam A, Desjardins L, Barnhill R, et al. Fine Needle Aspiration
Biopsy in Uveal Melanoma: Technique, Complications, and Outcomes.
Am J Ophthalmol 2016;162:28-34 e21. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26556006
.
236. Augsburger JJ, Correa ZM, Augsburger BD. Frequency and
implications of discordant gene expression profile class in posterior uveal
melanomas sampled by fine needle aspiration biopsy. Am J Ophthalmol
2015;159:248-256. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25448994
.
237. Nagiel A, McCannel CA, Moreno C, McCannel TA. Vitrectomy-
Assisted Biopsy for Molecular Prognostication of Choroidal Melanoma 2
Mm or Less in Thickness with a 27-Gauge Cutter. Retina 2017;37:1377-
1382. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28486310
.
238. Grewal DS, Cummings TJ, Mruthyunjaya P. Outcomes of 27-Gauge
Vitrectomy-Assisted Choroidal and Subretinal Biopsy. Ophthalmic Surg
Lasers Imaging Retina 2017;48:406-415. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28499052
.
239. Bagger M, Tebering JF, Kiilgaard JF. The ocular consequences and
applicability of minimally invasive 25-gauge transvitreal retinochoroidal
biopsy. Ophthalmology 2013;120:2565-2572. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24053996
.
240. Jensen OA, Prause JU, Scherfig E. Transvitreal retino-choroidal
biopsy of suspected malignant lesions of the choroid. Follow-up of cases
over 7 years. Acta Ophthalmol Scand 1997;75:409-411. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9374250
.
241. Seregard S, All-Ericsson C, Hjelmqvist L, et al. Diagnostic incisional
biopsies in clinically indeterminate choroidal tumours. Eye (Lond)
2013;27:115-118. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23154498
.
242. Akgul H, Otterbach F, Bornfeld N, Jurklies B. Intraocular biopsy
using special forceps: a new instrument and refined surgical technique.
Br J Ophthalmol 2011;95:79-82. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21163820
.
243. Kvanta A, Seregard S, Kopp ED, et al. Choroidal biopsies for
intraocular tumors of indeterminate origin. Am J Ophthalmol
2005;140:1002-1006. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16376642
.
244. Bagger M, Smidt-Nielsen I, Andersen MK, et al. Long-Term
Metastatic Risk after Biopsy of Posterior Uveal Melanoma.
Ophthalmology 2018;125:1969-1976. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29705056
.
245. Char DH, Miller T. Accuracy of presumed uveal melanoma
diagnosis before alternative therapy. Br J Ophthalmol 1995;79:692-696.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/7662638
.
246. McCannel TA, Chang MY, Burgess BL. Multi-year follow-up of fine-
needle aspiration biopsy in choroidal melanoma. Ophthalmology
2012;119:606-610. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22226884
.
247. Eide N, Walaas L. Fine-needle aspiration biopsy and other biopsies
in suspected intraocular malignant disease: a review. Acta Ophthalmol
2009;87:588-601. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19719804
.
248. Frizziero L, Midena E, Trainiti S, et al. Uveal Melanoma Biopsy: A
Review. Cancers (Basel) 2019;11. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31366043
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-68
249. Matet A, Ait Rais K, Malaise D, et al. Comparative Cytogenetic
Abnormalities in Paired Choroidal Melanoma Samples Obtained Before
and After Proton Beam Irradiation by Transscleral Fine-Needle
Aspiration Biopsy and Endoresection. Cancers (Basel) 2019;11.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/31416209
.
250. Cohen VM, Dinakaran S, Parsons MA, Rennie IG. Transvitreal fine
needle aspiration biopsy: the influence of intraocular lesion size on
diagnostic biopsy result. Eye (Lond) 2001;15:143-147. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11339578
.
251. Eide N, Syrdalen P, Walaas L, Hagmar B. Fine needle aspiration
biopsy in selecting treatment for inconclusive intraocular disease. Acta
Ophthalmol Scand 1999;77:448-452. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10463420
.
252. Correa ZM, Augsburger JJ. Sufficiency of FNAB aspirates of
posterior uveal melanoma for cytologic versus GEP classification in 159
patients, and relative prognostic significance of these classifications.
Graefes Arch Clin Exp Ophthalmol 2014;252:131-135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24270974
.
253. Singh AD, Aziz HA, Pelayes D, Biscotti CV. Twenty-five-gauge
cannula-assisted fine-needle aspiration biopsy of choroidal melanoma:
Cytopathological Analysis. Retina 2017;37:1674-1677. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28005633
.
254. Reddy DM, Mason LB, Mason JO, 3rd, et al. Vitrectomy and
Vitrector Port Needle Biopsy of Choroidal Melanoma for Gene
Expression Profile Testing Immediately before Brachytherapy.
Ophthalmology 2017;124:1377-1382. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28461017
.
255. Kim RS, Chevez-Barrios P, Bretana ME, et al. Histopathologic
Analysis of Transvitreal Fine Needle Aspiration Biopsy Needle Tracts for
Uveal Melanoma. Am J Ophthalmol 2017;174:9-16. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27818205
.
256. Chang MY, McCannel TA. Comparison of uveal melanoma
cytopathologic sample retrieval in trans-scleral versus vitrectomy-
assisted transvitreal fine needle aspiration biopsy. Br J Ophthalmol
2014;98:1654-1658. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24997179
.
257. Augsburger JJ, Correa ZM, Trichopoulos N. Prognostic implications
of cytopathologic classification of melanocytic uveal tumors evaluated by
fine-needle aspiration biopsy. Arq Bras Oftalmol 2013;76:72-79.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/23828465
.
258. Shields CL, Materin MA, Teixeira L, et al. Small choroidal
melanoma with chromosome 3 monosomy on fine-needle aspiration
biopsy. Ophthalmology 2007;114:1919-1924. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17698199
.
259. Shields CL, Ganguly A, Materin MA, et al. Chromosome 3 analysis
of uveal melanoma using fine-needle aspiration biopsy at the time of
plaque radiotherapy in 140 consecutive cases. Trans Am Ophthalmol
Soc 2007;105:43-52; discussion 52-43. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18427593
.
260. Metz CH, Bornfeld N, Metz KA, Gok M. Suspected vitreous seeding
of uveal melanoma: relevance of diagnostic vitrectomy. Br J Ophthalmol
2016;100:660-664. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26359338
.
261. Glasgow BJ, Brown HH, Zargoza AM, Foos RY. Quantitation of
tumor seeding from fine needle aspiration of ocular melanomas. Am J
Ophthalmol 1988;105:538-546. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3369520
.
262. Karcioglu ZA, Gordon RA, Karcioglu GL. Tumor seeding in ocular
fine needle aspiration biopsy. Ophthalmology 1985;92:1763-1767.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/4088631
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-69
263. Mashayekhi A, Lim RP, Shields CL, et al. Extraocular Extension of
Ciliochoroidal Melanoma after Transscleral Fine-Needle Aspiration
Biopsy. Retin Cases Brief Rep 2016;10:289-292. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26655386
.
264. Schefler AC, Gologorsky D, Marr BP, et al. Extraocular extension of
uveal melanoma after fine-needle aspiration, vitrectomy, and open
biopsy. JAMA Ophthalmol 2013;131:1220-1224. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24030334
.
265. Caminal JM, Sanz S, Carreras M, et al. Epibulbar seeding at the
site of a transvitreal fine-needle aspiration biopsy. Arch Ophthalmol
2006;124:587-589. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16606890
.
266. Bagger M, Andersen MT, Heegaard S, et al. Transvitreal
Retinochoroidal Biopsy Provides a Representative Sample From
Choroidal Melanoma for Detection of Chromosome 3 Aberrations. Invest
Ophthalmol Vis Sci 2015;56:5917-5924. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26377078
.
267. Sen J, Groenewald C, Hiscott PS, et al. Transretinal choroidal
tumor biopsy with a 25-gauge vitrector. Ophthalmology 2006;113:1028-
1031. Available at: https://www.ncbi.nlm.nih.gov/pubmed/16751041
.
268. Bechrakis NE, Foerster MH, Bornfeld N. Biopsy in indeterminate
intraocular tumors. Ophthalmology 2002;109:235-242. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11825801
.
269. Raja V, Russo A, Coupland S, et al. Extraocular seeding of
choroidal melanoma after a transretinal biopsy with a 25-gauge vitrector.
Retin Cases Brief Rep 2011;5:194-196. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25390162
.
270. Koch KR, Hishmi AM, Ortmann M, Heindl LM. Uveal Melanoma Cell
Seeding after Transretinal Tumor Biopsy? Ocul Oncol Pathol
2017;3:164-167. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29071267
.
271. Foulds WS. The uses and limitations of intraocular biopsy. Eye
(Lond) 1992;6 ( Pt 1):11-27. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/1426393
.
272. Chang MY, McCannel TA. Local treatment failure after globe-
conserving therapy for choroidal melanoma. Br J Ophthalmol
2013;97:804-811. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23645818
.
273. Bechrakis NE, Petousis V, Willerding G, et al. Ten-year results of
transscleral resection of large uveal melanomas: local tumour control
and metastatic rate. Br J Ophthalmol 2010;94:460-466. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19965818
.
274. Damato BE, Paul J, Foulds WS. Risk factors for residual and
recurrent uveal melanoma after trans-scleral local resection. Br J
Ophthalmol 1996;80:102-108. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8814738
.
275. Shields JA, Shields CL, Shah P, Sivalingam V. Partial lamellar
sclerouvectomy for ciliary body and choroidal tumors. Ophthalmology
1991;98:971-983. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/1866153
.
276. Caminal JM, Padron-Perez N, Arias L, et al. Transscleral resection
without hypotensive anaesthesia vs iodine-125 plaque brachytherapy in
the treatment of choroidal melanoma. Eye (Lond) 2016;30:833-842.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/27034202
.
277. Krantz BA, Dave N, Komatsubara KM, et al. Uveal melanoma:
epidemiology, etiology, and treatment of primary disease. Clin
Ophthalmol 2017;11:279-289. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28203054
.
278. Puusaari I, Damato B, Kivela T. Transscleral local resection versus
iodine brachytherapy for uveal melanomas that are large because of
tumour height. Graefes Arch Clin Exp Ophthalmol 2007;245:522-533.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/17111148
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-70
279. Kivela T, Puusaari I, Damato B. Transscleral resection versus
iodine brachytherapy for choroidal malignant melanomas 6 millimeters or
more in thickness: a matched case-control study. Ophthalmology
2003;110:2235-2244. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14597535
.
280. Damato BE. Local resection of uveal melanoma. Dev Ophthalmol
2012;49:66-80. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22042014
.
281. Dogrusoz M, Jager MJ, Damato B. Uveal Melanoma Treatment and
Prognostication. Asia Pac J Ophthalmol (Phila) 2017;6:186-196.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28399342
.
282. Yang J, Manson DK, Marr BP, Carvajal RD. Treatment of uveal
melanoma: where are we now? Ther Adv Med Oncol
2018;10:1758834018757175. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29497459
.
283. Dogrusoz M, Kroes WG, van Duinen SG, et al. Radiation Treatment
Affects Chromosome Testing in Uveal Melanoma. Invest Ophthalmol Vis
Sci 2015;56:5956-5964. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26393462
.
284. Ophthalmic Oncology Task F. Local Recurrence Significantly
Increases the Risk of Metastatic Uveal Melanoma. Ophthalmology
2016;123:86-91. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26505803
.
285. Shields CL, Kaliki S, Furuta M, et al. American Joint Committee on
Cancer classification of posterior uveal melanoma (tumor size category)
predicts prognosis in 7731 patients. Ophthalmology 2013;120:2066-
2071. Available at: https://www.ncbi.nlm.nih.gov/pubmed/23664467
.
286. Isager P, Ehlers N, Overgaard J. Have choroidal and ciliary body
melanomas changed during the period 1955-2000? Acta Ophthalmol
Scand 2004;82:509-516. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15453845
.
287. Rao YJ, Sein J, Badiyan S, et al. Patterns of care and survival
outcomes after treatment for uveal melanoma in the post-coms era
(2004-2013): a surveillance, epidemiology, and end results analysis. J
Contemp Brachytherapy 2017;9:453-465. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29204166
.
288. Vidoris AAC, Maia A, Lowen M, et al. Outcomes of primary
endoresection for choroidal melanoma. Int J Retina Vitreous 2017;3:42.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29142760
.
289. Konstantinidis L, Groenewald C, Coupland SE, Damato B. Long-
term outcome of primary endoresection of choroidal melanoma. Br J
Ophthalmol 2014;98:82-85. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24169650
.
290. Caminal JM, Mejia K, Masuet-Aumatell C, et al. Endoresection
versus iodine-125 plaque brachytherapy for the treatment of choroidal
melanoma. Am J Ophthalmol 2013;156:334-342 e331. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23677135
.
291. Garcia-Arumi J, Zapata MA, Balaguer O, et al. Endoresection in
high posterior choroidal melanomas: long-term outcome. Br J
Ophthalmol 2008;92:1040-1045. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18653597
.
292. Kertes PJ, Johnson JC, Peyman GA. Internal resection of posterior
uveal melanomas. Br J Ophthalmol 1998;82:1147-1153. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9924302
.
293. Damato B, Groenewald C, McGalliard J, Wong D. Endoresection of
choroidal melanoma. Br J Ophthalmol 1998;82:213-218. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9602614
.
294. The Collaborative Ocular Melanoma Study (COMS) randomized trial
of pre-enucleation radiation of large choroidal melanoma III: local
complications and observations following enucleation COMS report no.
11. Am J Ophthalmol 1998;126:362-372. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9744369
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-71
295. Collaborative Ocular Melanoma Study G. Assessment of metastatic
disease status at death in 435 patients with large choroidal melanoma in
the Collaborative Ocular Melanoma Study (COMS): COMS report no. 15.
Arch Ophthalmol 2001;119:670-676. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11346394
.
296. Weis E, Salopek TG, McKinnon JG, et al. Management of uveal
melanoma: a consensus-based provincial clinical practice guideline. Curr
Oncol 2016;23:e57-64. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26966414
.
297. Van Acker E, De Potter P. [Porous polyethylene (Medpor) orbital
implant. Prospective study of 75 primary implantations]. J Fr Ophtalmol
2001;24:1067-1073. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11913237
.
298. Ho VWM, Hussain RN, Czanner G, et al. Porous Versus Nonporous
Orbital Implants After Enucleation for Uveal Melanoma: A Randomized
Study. Ophthalmic Plast Reconstr Surg 2017;33:452-458. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27861329
.
299. Hope-Stone L, Brown SL, Heimann H, et al. Phantom Eye
Syndrome: Patient Experiences after Enucleation for Uveal Melanoma.
Ophthalmology 2015;122:1585-1590. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26004080
.
300. Hawkins BS, Collaborative Ocular Melanoma Study G. The
Collaborative Ocular Melanoma Study (COMS) randomized trial of pre-
enucleation radiation of large choroidal melanoma: IV. Ten-year mortality
findings and prognostic factors. COMS report number 24. Am J
Ophthalmol 2004;138:936-951. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15629284
.
301. The Collaborative Ocular Melanoma Study (COMS) randomized trial
of pre-enucleation radiation of large choroidal melanoma II: initial
mortality findings. COMS report no. 10. Am J Ophthalmol 1998;125:779-
796. Available at: https://www.ncbi.nlm.nih.gov/pubmed/9645716
.
302. Augsburger JJ, Lauritzen K, Gamel JW, et al. Matched group study
of preenucleation radiotherapy versus enucleation alone for primary
malignant melanoma of the choroid and ciliary body. Am J Clin Oncol
1990;13:382-387. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2220658
.
303. Diener-West M, Earle JD, Fine SL, et al. The COMS randomized
trial of iodine 125 brachytherapy for choroidal melanoma, III: initial
mortality findings. COMS Report No. 18. Arch Ophthalmol 2001;119:969-
982. Available at: https://www.ncbi.nlm.nih.gov/pubmed/11448319
.
304. Collaborative Ocular Melanoma Study G. The COMS randomized
trial of iodine 125 brachytherapy for choroidal melanoma: V. Twelve-year
mortality rates and prognostic factors: COMS report No. 28. Arch
Ophthalmol 2006;124:1684-1693. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17159027
.
305. Augsburger JJ, Gamel JW, Sardi VF, et al. Enucleation vs cobalt
plaque radiotherapy for malignant melanomas of the choroid and ciliary
body. Arch Ophthalmol 1986;104:655-661. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3518677
.
306. Augsburger JJ, Correa ZM, Freire J, Brady LW. Long-term survival
in choroidal and ciliary body melanoma after enucleation versus plaque
radiation therapy. Ophthalmology 1998;105:1670-1678. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9754176
.
307. Augsburger JJ, Gamel JW, Lauritzen K, Brady LW. Cobalt-60
plaque radiotherapy vs enucleation for posterior uveal melanoma. Am J
Ophthalmol 1990;109:585-592. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2333923
.
308. Adams KS, Abramson DH, Ellsworth RM, et al. Cobalt plaque
versus enucleation for uveal melanoma: comparison of survival rates. Br
J Ophthalmol 1988;72:494-497. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3415941
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-72
309. Markoe AM, Brady LW, Jr., Shields JA, et al. Malignant melanoma
of the eye: treatment of posterior uveal lesions by Co-60 plaque
radiotherapy versus enucleation. Radiology 1985;156:801-803. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/4023247
.
310. Brady LW, Hernandez JC. Brachytherapy of choroidal melanomas.
Strahlenther Onkol 1992;168:61-65. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/1542847
.
311. Seddon JM, Gragoudas ES, Egan KM, et al. Relative survival rates
after alternative therapies for uveal melanoma. Ophthalmology
1990;97:769-777. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2374681
.
312. Mosci C, Lanza FB, Barla A, et al. Comparison of clinical outcomes
for patients with large choroidal melanoma after primary treatment with
enucleation or proton beam radiotherapy. Ophthalmologica
2012;227:190-196. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22269846
.
313. Dinca EB, Yianni J, Rowe J, et al. Survival and complications
following gamma knife radiosurgery or enucleation for ocular melanoma:
a 20-year experience. Acta Neurochir (Wien) 2012;154:605-610.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/22231777
.
314. Furdova A, Slezak P, Chorvath M, et al. No differences in outcome
between radical surgical treatment (enucleation) and stereotactic
radiosurgery in patients with posterior uveal melanoma. Neoplasma
2010;57:377-381. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20429631
.
315. Cohen VM, Carter MJ, Kemeny A, et al. Metastasis-free survival
following treatment for uveal melanoma with either stereotactic
radiosurgery or enucleation. Acta Ophthalmol Scand 2003;81:383-388.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/12859266
.
316. Melia M, Moy CS, Reynolds SM, et al. Quality of life after iodine 125
brachytherapy vs enucleation for choroidal melanoma: 5-year results
from the Collaborative Ocular Melanoma Study: COMS QOLS Report
No. 3. Arch Ophthalmol 2006;124:226-238. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16476893
.
317. van Beek JGM, Buitendijk GHS, Timman R, et al. Quality of life:
fractionated stereotactic radiotherapy versus enucleation treatment in
uveal melanoma patients. Acta Ophthalmol 2018;96:841-848. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/30284368
.
318. Damato B, Hope-Stone L, Cooper B, et al. Patient-reported
Outcomes and Quality of Life After Treatment of Choroidal Melanoma: A
Comparison of Enucleation Versus Radiotherapy in 1596 Patients. Am J
Ophthalmol 2018;193:230-251. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29555484
.
319. Brandberg Y, Kock E, Oskar K, et al. Psychological reactions and
quality of life in patients with posterior uveal melanoma treated with
ruthenium plaque therapy or enucleation: a one year follow-up study.
Eye (Lond) 2000;14:839-846. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11584839
.
320. Klingenstein A, Furweger C, Muhlhofer AK, et al. Quality of life in
the follow-up of uveal melanoma patients after enucleation in comparison
to CyberKnife treatment. Graefes Arch Clin Exp Ophthalmol
2016;254:1005-1012. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26573389
.
321. Cruickshanks KJ, Fryback DG, Nondahl DM, et al. Treatment choice
and quality of life in patients with choroidal melanoma. Arch Ophthalmol
1999;117:461-467. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10206573
.
322. Available at: http://documents.cap.org/protocols/cp-uveal-
melanoma-17protocol-4000.pdf. Accessed May 21st, 2021.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-73
323. Abrams MJ, Gagne NL, Melhus CS, Mignano JE. Brachytherapy vs.
external beam radiotherapy for choroidal melanoma: Survival and
patterns-of-care analyses. Brachytherapy 2016;15:216-223. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26846381
.
324. Desjardins L, Levy-Gabriel C, Lumbroso-Lerouic L, et al.
[Prognostic factors for malignant uveal melanoma. Retrospective study
on 2,241 patients and recent contribution of monosomy-3 research]. J Fr
Ophtalmol 2006;29:741-749. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16988624
.
325. Echegaray JJ, Bechrakis NE, Singh N, et al. Iodine-125
Brachytherapy for Uveal Melanoma: A Systematic Review of Radiation
Dose. Ocul Oncol Pathol 2017;3:193-198. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29071269
.
326. Rospond-Kubiak I, Kociecki J, Damato B. Clinical evaluation of a
paper chart for predicting ruthenium plaque placement in relation to
choroidal melanoma. Eye (Lond) 2018;32:421-425. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28960216
.
327. Sanchez-Tabernero S, Garcia-Alvarez C, Munoz-Moreno MF, et al.
Pattern of Local Recurrence After I-125 Episcleral Brachytherapy for
Uveal Melanoma in a Spanish Referral Ocular Oncology Unit. Am J
Ophthalmol 2017;180:39-45. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28572063
.
328. McCannel TA, Kamrava M, Demanes J, et al. 23-mm iodine-125
plaque for uveal melanoma: benefit of vitrectomy and silicone oil on
visual acuity. Graefes Arch Clin Exp Ophthalmol 2016;254:2461-2467.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/27638702
.
329. Collaborative Ocular Melanoma Study G. Incidence of cataract and
outcomes after cataract surgery in the first 5 years after iodine 125
brachytherapy in the Collaborative Ocular Melanoma Study: COMS
Report No. 27. Ophthalmology 2007;114:1363-1371. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17337065
.
330. Pilotto E, Vujosevic S, De Belvis V, et al. Long-term choroidal
vascular changes after iodine brachytherapy versus transpupillary
thermotherapy for choroidal melanoma. Eur J Ophthalmol 2009;19:646-
653. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19551682
.
331. Sobrin L, Schiffman JC, Markoe AM, Murray TG. Outcomes of
iodine 125 plaque radiotherapy after initial observation of suspected
small choroidal melanomas: a pilot study. Ophthalmology
2005;112:1777-1783. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16095708
.
332. Seregard S. Long-term survival after ruthenium plaque radiotherapy
for uveal melanoma. A meta-analysis of studies including 1,066 patients.
Acta Ophthalmol Scand 1999;77:414-417. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10463412
.
333. Finger PT, Berson A, Szechter A. Palladium-103 plaque
radiotherapy for choroidal melanoma: results of a 7-year study.
Ophthalmology 1999;106:606-613. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10080222
.
334. Miguel D, de Frutos-Baraja JM, Lopez-Lara F, et al. Visual outcome
after posterior uveal melanoma episcleral brachytherapy including
radiobiological doses. J Contemp Brachytherapy 2018;10:123-131.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29789761
.
335. Tsui I, Beardsley RM, McCannel TA, et al. Visual Acuity, Contrast
Sensitivity and Color Vision Three Years After Iodine-125 Brachytherapy
for Choroidal and Ciliary Body Melanoma. Open Ophthalmol J
2015;9:131-135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26312123
.
336. Boldt HC, Melia BM, Liu JC, et al. I-125 brachytherapy for choroidal
melanoma photographic and angiographic abnormalities: the
Collaborative Ocular Melanoma Study: COMS Report No. 30.
Ophthalmology 2009;116:106-115 e101. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19118701
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-74
337. Finger PT, Chin KJ, Tena LB. A five-year study of slotted eye
plaque radiation therapy for choroidal melanoma: near, touching, or
surrounding the optic nerve. Ophthalmology 2012;119:415-422.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/22133796
.
338. Almony A, Breit S, Zhao H, et al. Tilting of radioactive plaques after
initial accurate placement for treatment of uveal melanoma. Arch
Ophthalmol 2008;126:65-70. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18195220
.
339. Rivard MJ, Chiu-Tsao S-T, Finger PT, et al. Comparison of dose
calculation methods for brachytherapy of intraocular tumors. Med Phys
2010;38:306-316. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21361199
.
340. Meeralakshmi P, Shah PK, Narendran V. Experiences of two
different modalities in the management of choroidal melanoma in the
Asian Indian population. South Asian J Cancer 2017;6:134-136.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28975125
.
341. Karlovits B, Trombetta MG, Verstraeten T, et al. Local control and
visual acuity following treatment of medium-sized ocular melanoma using
a contact eye plaque: a single surgeon experience. Brachytherapy
2011;10:228-231. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20932809
.
342. Wagner A, Chen A, Cook T, et al. Outcomes and control rates for I-
125 plaque brachytherapy for uveal melanoma: a community-based
institutional experience. ISRN Ophthalmol 2014;2014:950975. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/24734198
.
343. Berry JL, Dandapani SV, Stevanovic M, et al. Outcomes of
choroidal melanomas treated with eye physics: a 20-year review. JAMA
Ophthalmol 2013;131:1435-1442. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24008431
.
344. Krema H, Heydarian M, Beiki-Ardakani A, et al. A comparison
between (1)(2)(5)Iodine brachytherapy and stereotactic radiotherapy in
the management of juxtapapillary choroidal melanoma. Br J Ophthalmol
2013;97:327-332. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23335213
.
345. Wilson MW, Hungerford JL. Comparison of episcleral plaque and
proton beam radiation therapy for the treatment of choroidal melanoma.
Ophthalmology 1999;106:1579-1587. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10442907
.
346. Char DH, Kroll S, Phillips TL, Quivey JM. Late radiation failures
after iodine 125 brachytherapy for uveal melanoma compared with
charged-particle (proton or helium ion) therapy. Ophthalmology
2002;109:1850-1854. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12359605
.
347. Char DH, Quivey JM, Castro JR, et al. Helium ions versus iodine
125 brachytherapy in the management of uveal melanoma. A
prospective, randomized, dynamically balanced trial. Ophthalmology
1993;100:1547-1554. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8414414
.
348. Harbour JW, Char DH, Kroll S, et al. Metastatic risk for distinct
patterns of postirradiation local recurrence of posterior uveal melanoma.
Ophthalmology 1997;104:1785-1792; discussion 1792-1783. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/9373108
.
349. Wang Z, Nabhan M, Schild SE, et al. Charged particle radiation
therapy for uveal melanoma: a systematic review and meta-analysis. Int
J Radiat Oncol Biol Phys 2013;86:18-26. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23040219
.
350. Badiyan SN, Rao RC, Apicelli AJ, et al. Outcomes of iodine-125
plaque brachytherapy for uveal melanoma with intraoperative
ultrasonography and supplemental transpupillary thermotherapy. Int J
Radiat Oncol Biol Phys 2014;88:801-805. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24462385
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-75
351. Jensen AW, Petersen IA, Kline RW, et al. Radiation complications
and tumor control after 125I plaque brachytherapy for ocular melanoma.
Int J Radiat Oncol Biol Phys 2005;63:101-108. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16111577
.
352. Quivey JM, Augsburger J, Snelling L, Brady LW. 125I plaque
therapy for uveal melanoma. Analysis of the impact of time and dose
factors on local control. Cancer 1996;77:2356-2362. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8635107
.
353. Oellers P, Mowery YM, Perez BA, et al. Efficacy and Safety of Low-
Dose Iodine Plaque Brachytherapy for Juxtapapillary Choroidal
Melanoma. Am J Ophthalmol 2018;186:32-40. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29199010
.
354. Perez BA, Mettu P, Vajzovic L, et al. Uveal melanoma treated with
iodine-125 episcleral plaque: an analysis of dose on disease control and
visual outcomes. Int J Radiat Oncol Biol Phys 2014;89:127-136.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/24613808
.
355. Aziz HA, Singh N, Bena J, et al. Vision Loss Following Episcleral
Brachytherapy for Uveal Melanoma: Development of a Vision
Prognostication Tool. JAMA Ophthalmol 2016;134:615-620. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27101414
.
356. Murray TG, Markoe AM, Gold AS, et al. Long-term followup
comparing two treatment dosing strategies of (125) I plaque radiotherapy
in the management of small/medium posterior uveal melanoma. J
Ophthalmol 2013;2013:517032. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23533708
.
357. Khan N, Khan MK, Bena J, et al. Plaque brachytherapy for uveal
melanoma: a vision prognostication model. Int J Radiat Oncol Biol Phys
2012;84:e285-290. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22658514
.
358. Puusaari I, Heikkonen J, Kivela T. Effect of radiation dose on ocular
complications after iodine brachytherapy for large uveal melanoma:
empirical data and simulation of collimating plaques. Invest Ophthalmol
Vis Sci 2004;45:3425-3434. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15452045
.
359. Char DH, Kroll S, Quivey JM, Castro J. Long term visual outcome of
radiated uveal melanomas in eyes eligible for randomisation to
enucleation versus brachytherapy. Br J Ophthalmol 1996;80:117-124.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/8814740
.
360. Wisely CE, Hadziahmetovic M, Reem RE, et al. Long-term visual
acuity outcomes in patients with uveal melanoma treated with 125I
episcleral OSU-Nag plaque brachytherapy. Brachytherapy 2016;15:12-
22. Available at: https://www.ncbi.nlm.nih.gov/pubmed/26525215
.
361. De Caluwe A, Termote K, Van Gestel D, Van Limbergen E. Dose-
response in choroidal melanoma. Radiother Oncol 2018;127:374-378.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29680322
.
362. Danish H, Ferris MJ, Balagamwala E, et al. Comparative outcomes
and toxicities for ruthenium-106 versus palladium-103 in the treatment of
choroidal melanoma. Melanoma Res 2018;28:120-125. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29303890
.
363. Cho Y, Chang JS, Yoon JS, et al. Ruthenium-106 Brachytherapy
with or without Additional Local Therapy Shows Favorable Outcome for
Variable-Sized Choroidal Melanomas in Korean Patients. Cancer Res
Treat 2018;50:138-147. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28343376
.
364. Siedlecki J, Reiterer V, Leicht S, et al. Incidence of secondary
glaucoma after treatment of uveal melanoma with robotic radiosurgery
versus brachytherapy. Acta Ophthalmol 2017;95:e734-e739. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/28387434
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-76
365. Tagliaferri L, Pagliara MM, Masciocchi C, et al. Nomogram for
predicting radiation maculopathy in patients treated with Ruthenium-106
plaque brachytherapy for uveal melanoma. J Contemp Brachytherapy
2017;9:540-547. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29441098
.
366. Marinkovic M, Horeweg N, Fiocco M, et al. Ruthenium-106
brachytherapy for choroidal melanoma without transpupillary
thermotherapy: Similar efficacy with improved visual outcome. Eur J
Cancer 2016;68:106-113. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27741435
.
367. Kaiserman I, Anteby I, Chowers I, et al. Post-brachytherapy initial
tumour regression rate correlates with metastatic spread in posterior
uveal melanoma. Br J Ophthalmol 2004;88:892-895. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15205232
.
368. Georgopoulos M, Zehetmayer M, Ruhswurm I, et al. Tumour
regression of uveal melanoma after ruthenium-106 brachytherapy or
stereotactic radiotherapy with gamma knife or linear accelerator.
Ophthalmologica 2003;217:315-319. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12913319
.
369. Coleman DJ, Silverman RH, Rondeau MJ, et al. Ultrasonic tissue
characterization of uveal melanoma and prediction of patient survival
after enucleation and brachytherapy. Am J Ophthalmol 1991;112:682-
688. Available at: https://www.ncbi.nlm.nih.gov/pubmed/1957904
.
370. Gragoudas ES, Lane AM, Regan S, et al. A randomized controlled
trial of varying radiation doses in the treatment of choroidal melanoma.
Arch Ophthalmol 2000;118:773-778. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10865313
.
371. Lin AJ, Rao YJ, Acharya S, et al. Patterns of care and outcomes of
proton and eye plaque brachytherapy for uveal melanoma: Review of the
National Cancer Database. Brachytherapy 2017;16:1225-1231. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/28966081
.
372. Castro JR, Char DH, Petti PL, et al. 15 years experience with
helium ion radiotherapy for uveal melanoma. Int J Radiat Oncol Biol
Phys 1997;39:989-996. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9392536
.
373. Tsuji H, Ishikawa H, Yanagi T, et al. Carbon-ion radiotherapy for
locally advanced or unfavorably located choroidal melanoma: a Phase
I/II dose-escalation study. Int J Radiat Oncol Biol Phys 2007;67:857-862.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/17161555
.
374. Hrbacek J, Mishra KK, Kacperek A, et al. Practice Patterns Analysis
of Ocular Proton Therapy Centers: The International OPTIC Survey. Int J
Radiat Oncol Biol Phys 2016;95:336-343. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27084651
.
375. Hartsell WF, Kapur R, Hartsell SO, et al. Feasibility of Proton Beam
Therapy for Ocular Melanoma Using a Novel 3D Treatment Planning
Technique. Int J Radiat Oncol Biol Phys 2016;95:353-359. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27084652
.
376. Krema H, Heydarian M, Beiki-Ardakani A, et al. Dosimetric and late
radiation toxicity comparison between iodine-125 brachytherapy and
stereotactic radiation therapy for juxtapapillary choroidal melanoma. Int J
Radiat Oncol Biol Phys 2013;86:510-515. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23507292
.
377. Sikuade MJ, Salvi S, Rundle PA, et al. Outcomes of treatment with
stereotactic radiosurgery or proton beam therapy for choroidal
melanoma. Eye (Lond) 2015;29:1194-1198. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26160531
.
378. Furdova A, Horkovicova K, Justusova P, Sramka M. Is it sufficient
to repeat LINEAR accelerator stereotactic radiosurgery in choroidal
melanoma? Bratisl Lek Listy 2016;117:456-462. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27546698
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-77
379. Muller K, Naus N, Nowak PJ, et al. Fractionated stereotactic
radiotherapy for uveal melanoma, late clinical results. Radiother Oncol
2012;102:219-224. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21864922
.
380. Modorati G, Miserocchi E, Galli L, et al. Gamma knife radiosurgery
for uveal melanoma: 12 years of experience. Br J Ophthalmol
2009;93:40-44. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18757470
.
381. Fakiris AJ, Lo SS, Henderson MA, et al. Gamma-knife-based
stereotactic radiosurgery for uveal melanoma. Stereotact Funct
Neurosurg 2007;85:106-112. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17228176
.
382. Simonova G, Novotny J, Jr., Liscak R, Pilbauer J. Leksell gamma
knife treatment of uveal melanoma. J Neurosurg 2002;97:635-639.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/12507111
.
383. Mueller AJ, Talies S, Schaller UC, et al. Stereotactic radiosurgery of
large uveal melanomas with the gamma-knife. Ophthalmology
2000;107:1381-1387; discussion 1387-1388. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10889116
.
384. Zehetmayer M, Kitz K, Menapace R, et al. Local tumor control and
morbidity after one to three fractions of stereotactic external beam
irradiation for uveal melanoma. Radiother Oncol 2000;55:135-144.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/10799725
.
385. Haji Mohd Yasin NA, Gray AR, Bevin TH, et al. Choroidal
melanoma treated with stereotactic fractionated radiotherapy and
prophylactic intravitreal bevacizumab: The Dunedin Hospital experience.
J Med Imaging Radiat Oncol 2016;60:756-763. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27378522
.
386. Wackernagel W, Holl E, Tarmann L, et al. Local tumour control and
eye preservation after gamma-knife radiosurgery of choroidal
melanomas. Br J Ophthalmol 2014;98:218-223. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24169651
.
387. Dunavoelgyi R, Zehetmayer M, Gleiss A, et al. Hypofractionated
stereotactic photon radiotherapy of posteriorly located choroidal
melanoma with five fractions at ten Gy--clinical results after six years of
experience. Radiother Oncol 2013;108:342-347. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24044800
.
388. Dunavoelgyi R, Dieckmann K, Gleiss A, et al. Local tumor control,
visual acuity, and survival after hypofractionated stereotactic photon
radiotherapy of choroidal melanoma in 212 patients treated between
1997 and 2007. Int J Radiat Oncol Biol Phys 2011;81:199-205. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/20675066
.
389. Dieckmann K, Georg D, Zehetmayer M, et al. LINAC based
stereotactic radiotherapy of uveal melanoma: 4 years clinical experience.
Radiother Oncol 2003;67:199-206. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12812851
.
390. Suesskind D, Scheiderbauer J, Buchgeister M, et al. Retrospective
evaluation of patients with uveal melanoma treated by stereotactic
radiosurgery with and without tumor resection. JAMA Ophthalmol
2013;131:630-637. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23494018
.
391. Kosydar S, Robertson JC, Woodfin M, et al. Systematic Review and
Meta-Analysis on the Use of Photon-based Stereotactic Radiosurgery
Versus Fractionated Stereotactic Radiotherapy for the Treatment of
Uveal Melanoma. Am J Clin Oncol 2020. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/33208706
.
392. Yazici G, Kiratli H, Ozyigit G, et al. Stereotactic Radiosurgery and
Fractionated Stereotactic Radiation Therapy for the Treatment of Uveal
Melanoma. Int J Radiat Oncol Biol Phys 2017;98:152-158. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28586956
.
393. Furdova A, Babal P, Kobzova D, et al. Uveal melanoma survival
rates after single dose stereotactic radiosurgery. Neoplasma
2018;65:965-971. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30334446
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-78
394. van den Bosch T, Vaarwater J, Verdijk R, et al. Risk factors
associated with secondary enucleation after fractionated stereotactic
radiotherapy in uveal melanoma. Acta Ophthalmol 2015;93:555-560.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/25879399
.
395. Sarici AM, Pazarli H. Gamma-knife-based stereotactic radiosurgery
for medium- and large-sized posterior uveal melanoma. Graefes Arch
Clin Exp Ophthalmol 2013;251:285-294. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22944897
.
396. Eibl-Lindner K, Furweger C, Nentwich M, et al. Robotic radiosurgery
for the treatment of medium and large uveal melanoma. Melanoma Res
2016;26:51-57. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26484738
.
397. Zahorjanova P, Furdova A, Waczulikova I, et al. Radiation
Maculopathy after One-Day Session Stereotactic Radiosurgery in
Patients with Ciliary Body and Choroidal Melanoma. Cesk Slov Oftalmol
2019;75:3-10. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31382751
.
398. Horwath-Winter J, Schneider MR, Wackernagel W, et al. Influence
of single-fraction Gamma-Knife radiosurgery on ocular surface and tear
function in choroidal melanoma patients. Br J Ophthalmol 2013;97:466-
470. Available at: https://www.ncbi.nlm.nih.gov/pubmed/23349246
.
399. Dunavoelgyi R, Dieckmann K, Gleiss A, et al. Radiogenic side
effects after hypofractionated stereotactic photon radiotherapy of
choroidal melanoma in 212 patients treated between 1997 and 2007. Int
J Radiat Oncol Biol Phys 2012;83:121-128. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21945109
.
400. Gigliotti CR, Modorati G, Di Nicola M, et al. Predictors of radio-
induced visual impairment after radiosurgery for uveal melanoma. Br J
Ophthalmol 2018;102:833-839. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28903963
.
401. Dunavoelgyi R, Georg D, Zehetmayer M, et al. Dose-response of
critical structures in the posterior eye segment to hypofractioned
stereotactic photon radiotherapy of choroidal melanoma. Radiother
Oncol 2013;108:348-353. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24044791
.
402. Augsburger JJ, Roth SE, Magargal LE, Shields JA. Panretinal
photocoagulation for radiation-induced ocular ischemia. Ophthalmic Surg
1987;18:589-593. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3658314
.
403. Konstantinidis L, Groenewald C, Coupland SE, Damato B. Trans-
scleral local resection of toxic choroidal melanoma after proton beam
radiotherapy. Br J Ophthalmol 2014;98:775-779. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24568869
.
404. Finger PT, Chin KJ. Antivascular endothelial growth factor
bevacizumab for radiation optic neuropathy: secondary to plaque
radiotherapy. Int J Radiat Oncol Biol Phys 2012;82:789-798. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21277107
.
405. Finger PT, Chin KJ, Semenova EA. Intravitreal anti-VEGF therapy
for macular radiation retinopathy: a 10-year study. Eur J Ophthalmol
2016;26:60-66. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26391167
.
406. Roelofs K, Larocque MP, Murtha A, Weis E. The Use of Intravitreal
Anti-VEGF and Triamcinolone in the Treatment of Radiation
Papillopathy. Ocul Oncol Pathol 2018;4:395-400. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30574493
.
407. Russo A, Reibaldi M, Avitabile T, et al. Dexamethasone Intravitreal
Implant Vs Ranibizumab in the Treatment of Macular Edema Secondary
to Brachytherapy for Choroidal Melanoma. Retina 2018;38:788-794.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28272283
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-79
408. Daruich A, Matet A, Schalenbourg A, Zografos L. Intravitreal anti-
vascular endothelial growth factor treatment at 2-month intervals reduces
foveal avascular zone enlargement and vision loss in radiation
maculopathy: A Pilot Study. Retina 2019;39:1519-1526. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29746413
.
409. Murray TG, Latiff A, Villegas VM, Gold AS. Aflibercept for Radiation
Maculopathy Study: A Prospective, Randomized Clinical Study.
Ophthalmol Retina 2019;3:561-566. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31277797
.
410. Caminal JM, Flores-Moreno I, Arias L, et al. Intravitreal
Dexamethasone Implant for Radiation Maculopathy Secondary to Plaque
Brachytherapy in Choroidal Melanoma. Retina 2015;35:1890-1897.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/26035401
.
411. Mashayekhi A, Rojanaporn D, Al-Dahmash S, et al. Monthly
intravitreal bevacizumab for macular edema after iodine-125 plaque
radiotherapy of uveal melanoma. Eur J Ophthalmol 2014;24:228-234.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/23934823
.
412. Horgan N, Shields CL, Mashayekhi A, et al. Periocular
triamcinolone for prevention of macular edema after iodine 125 plaque
radiotherapy of uveal melanoma. Retina 2008;28:987-995. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18698302
.
413. Horgan N, Shields CL, Mashayekhi A, et al. Periocular
triamcinolone for prevention of macular edema after plaque radiotherapy
of uveal melanoma: a randomized controlled trial. Ophthalmology
2009;116:1383-1390. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19481812
.
414. Kim IK, Lane AM, Jain P, et al. Ranibizumab for the Prevention of
Radiation Complications in Patients Treated With Proton Beam
Irradiation for Choroidal Melanoma. Trans Am Ophthalmol Soc
2016;114:T2. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27630373
.
415. Shields JA, Glazer LC, Mieler WF, et al. Comparison of xenon arc
and argon laser photocoagulation in the treatment of choroidal
melanomas. Am J Ophthalmol 1990;109:647-655. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2346193
.
416. Munzenrider JE, Verhey LJ, Gragoudas ES, et al. Conservative
treatment of uveal melanoma: local recurrence after proton beam
therapy. Int J Radiat Oncol Biol Phys 1989;17:493-498. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2550395
.
417. Shields CL, Shields JA, Gunduz K, et al. Radiation therapy for uveal
malignant melanoma. Ophthalmic Surg Lasers 1998;29:397-409.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/9599365
.
418. Sagoo MS, Shields CL, Mashayekhi A, et al. Plaque radiotherapy
for choroidal melanoma encircling the optic disc (circumpapillary
choroidal melanoma). Arch Ophthalmol 2007;125:1202-1209. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/17846359
.
419. Peyman GA, Gremillion CM. Eye wall resection in the management
of uveal neoplasms. Jpn J Ophthalmol 1989;33:458-471. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2625780
.
420. Shields JA, Shields CL, Peairs R, Racciato P. Laser ablation of an
enlarging small melanocytic choroidal tumor: 16-year follow-up and
rationale for treatment. Ophthalmic Surg Lasers Imaging 2006;37:79-81.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16468559
.
421. Qiang Z, Cairns JD. Laser photocoagulation treatment of choroidal
melanoma. Aust N Z J Ophthalmol 1993;21:87-92. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8333939
.
422. Shields JA, Shields CL. Current management of posterior uveal
melanoma. Mayo Clin Proc 1993;68:1196-1200. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8246622
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-80
423. Riechardt AI, Cordini D, Dobner B, et al. Salvage proton beam
therapy in local recurrent uveal melanoma. Am J Ophthalmol
2014;158:948-956. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25038327
.
424. Brewington BY, Shao YF, Davidorf FH, Cebulla CM. Brachytherapy
for patients with uveal melanoma: historical perspectives and future
treatment directions. Clin Ophthalmol 2018;12:925-934. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29844657
.
425. Rodriguez A, Duenas-Gonzalez A, Delgado-Pelayo S. Clinical
presentation and management of uveal melanoma. Mol Clin Oncol
2016;5:675-677. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28101347
.
426. Augsburger JJ, Mullen D, Kleineidam M. Planned combined I-125
plaque irradiation and indirect ophthalmoscope laser therapy for
choroidal malignant melanoma. Ophthalmic Surg 1993;24:76-81.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/8446358
.
427. Tse DT. Laser photocoagulation of choroidal malignant melanoma.
Biomed Pharmacother 1986;40:323-325. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3828478
.
428. Brancato R, Menchini U, Pece A. Enucleation after argon laser
photocoagulation for choroidal melanoma. Ann Ophthalmol 1988;20:296-
298. Available at: https://www.ncbi.nlm.nih.gov/pubmed/3056206
.
429. Beaumont P. Laser photocoagulation treatment of choroidal
melanoma. Aust N Z J Ophthalmol 1994;22:85-86. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8037923
.
430. Aaberg TM, Jr. Laser for choroidal melanoma. Int Ophthalmol Clin
2006;46:15-26. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16365552
.
431. Harbour JW, Meredith TA, Thompson PA, Gordon ME.
Transpupillary thermotherapy versus plaque radiotherapy for suspected
choroidal melanomas. Ophthalmology 2003;110:2207-2214; discussion
2215. Available at: https://www.ncbi.nlm.nih.gov/pubmed/14597531
.
432. De Potter P, Jamart J. Adjuvant indocyanine green in transpupillary
thermotherapy for choroidal melanoma. Ophthalmology 2003;110:406-
413; discussion 413-404. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12578788
.
433. Chojniak MM, Chojniak R, Nishimoto IN, et al. Primary
transpupillary thermotherapy for small choroidal melanoma. Graefes
Arch Clin Exp Ophthalmol 2011;249:1859-1865. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21713533
.
434. Parrozzani R, Boccassini B, De Belvis V, et al. Long-term outcome
of transpupillary thermotherapy as primary treatment of selected
choroidal melanoma. Acta Ophthalmol 2009;87:789-792. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18778335
.
435. Win PH, Robertson DM, Buettner H, et al. Extended follow-up of
small melanocytic choroidal tumors treated with transpupillary
thermotherapy. Arch Ophthalmol 2006;124:503-506. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16606875
.
436. Spire M, Devouassoux MS, Kodjikian L, et al. Primary transpupillary
thermotherapy for 18 small posterior pole uveal melanomas. Am J
Ophthalmol 2006;141:840-849. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16678505
.
437. Horgan N, Shields CL, Mashayekhi A, et al. Early macular
morphological changes following plaque radiotherapy for uveal
melanoma. Retina 2008;28:263-273. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18301032
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-81
438. Desjardins L, Lumbroso-Le Rouic L, Levy-Gabriel C, et al.
Combined proton beam radiotherapy and transpupillary thermotherapy
for large uveal melanomas: a randomized study of 151 patients.
Ophthalmic Res 2006;38:255-260. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16888407
.
439. Bartlema YM, Oosterhuis JA, Journee-De Korver JG, et al.
Combined plaque radiotherapy and transpupillary thermotherapy in
choroidal melanoma: 5 years' experience. Br J Ophthalmol
2003;87:1370-1373. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14609837
.
440. Girvigian MR, Astrahan MA, Lim JI, et al. Episcleral plaque 125I
radiotherapy with episcleral LCF hyperthermia: a prospective
randomized trial. Brachytherapy 2003;2:229-239. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15062131
.
441. Shields CL, Cater J, Shields JA, et al. Combined plaque
radiotherapy and transpupillary thermotherapy for choroidal melanoma:
tumor control and treatment complications in 270 consecutive patients.
Arch Ophthalmol 2002;120:933-940. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12096964
.
442. Petrovich Z, Astrahan MA, Luxton G, et al. Episcleral plaque
thermoradiotherapy in patients with choroidal melanoma. Int J Radiat
Oncol Biol Phys 1992;23:599-603. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/1612961
.
443. Shields CL, Shields JA, Perez N, et al. Primary transpupillary
thermotherapy for small choroidal melanoma in 256 consecutive cases:
outcomes and limitations. Ophthalmology 2002;109:225-234. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/11825800
.
444. Yarovoy AA, Magaramov DA, Bulgakova ES. The comparison of
ruthenium brachytherapy and simultaneous transpupillary thermotherapy
of choroidal melanoma with brachytherapy alone. Brachytherapy
2012;11:224-229. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22104351
.
445. Gunduz K, Kurt RA, Akmese HE, et al. Ruthenium-106 plaque
radiotherapy alone or in combination with transpupillary thermotherapy in
the management of choroidal melanoma. Jpn J Ophthalmol 2010;54:338-
343. Available at: https://www.ncbi.nlm.nih.gov/pubmed/20700803
.
446. Godfrey DG, Waldron RG, Capone A, Jr. Transpupillary
thermotherapy for small choroidal melanoma. Am J Ophthalmol
1999;128:88-93. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10482099
.
447. Croughs P, Baguette MN. [Treatment of choroidal nevi and
melanomas using cryo-applications: preliminary study]. Bull Soc Belge
Ophtalmol 1992;243:87-93. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/1302157
.
448. Brovkina AF, Ziangirova GG, Komarov BA. [Cryodestruction of
choroidal melanomas]. Vestn Oftalmol 1977:61-63. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/860372
.
449. Abramson DH, Lisman RD. Cryopexy of a choroidal melanoma. Ann
Ophthalmol 1979;11:1418-1421. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/556166
.
450. Lincoff H, McLean J, Long R. The cryosurgical treatment of
intraocular tumors. Am J Ophthalmol 1967;63:389-399. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/6019526
.
451. Wilson DJ, Klein ML. Choroidal melanoma treated with cryotherapy.
Arch Ophthalmol 2002;120:393-395. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11879148
.
452. Wilson DJ, Klein ML. Cryotherapy as a primary treatment for
choroidal melanoma. Arch Ophthalmol 2002;120:400-403. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11879152
.
453. Jalkh AE, Trempe CL, Nasrallah FP, et al. Treatment of small
choroidal melanomas with photocoagulation. Ophthalmic Surg
1988;19:738-742. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3194108
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-82
454. Shields JA. Current approaches to the diagnosis and management
of choroidal melanomas. Surv Ophthalmol 1977;21:443-463. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/331533
.
455. Bellmann C, Lumbroso-Le Rouic L, Levy C, et al. Uveal melanoma:
management and outcome of patients with extraocular spread. Br J
Ophthalmol 2010;94:569-574. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19965816
.
456. Shields CL, Sioufi K, Robbins JS, et al. LARGE UVEAL
MELANOMA (>/=10 MM THICKNESS): Clinical Features and Millimeter-
by-Millimeter Risk of Metastasis in 1311 Cases. The 2018 Albert E.
Finley Lecture. Retina 2018;38:2010-2022. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29528980
.
457. Toth J. [Retrospective study of uveal melanoma]. Magy Onkol
2005;49:19-25. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15902329
.
458. van Beek JG, Koopmans AE, Vaarwater J, et al. The prognostic
value of extraocular extension in relation to monosomy 3 and gain of
chromosome 8q in uveal melanoma. Invest Ophthalmol Vis Sci
2014;55:1284-1291. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24508790
.
459. Blanco G. Diagnosis and treatment of orbital invasion in uveal
melanoma. Can J Ophthalmol 2004;39:388-396. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15327104
.
460. Pereira PR, Odashiro AN, Lim LA, et al. Current and emerging
treatment options for uveal melanoma. Clin Ophthalmol 2013;7:1669-
1682. Available at: https://www.ncbi.nlm.nih.gov/pubmed/24003303
.
461. Mirkiewicz-Sieradzka B, Zygulska-Mach H, Romanowska B, et al.
[Attempts of orbit irradiation after enucleation of the eye with malignant
choroidal melanoma. Part I]. Klin Oczna 1999;101:287-290. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10581897
.
462. Shammas HF, Blodi FC. Orbital extension of Choroidal and ciliary
body melanomas. Arch Ophthalmol 1977;95:2002-2005. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/921579
.
463. Kersten RC, Tse DT, Anderson RL, Blodi FC. The role of orbital
exenteration in choroidal melanoma with extrascleral extension.
Ophthalmology 1985;92:436-443. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3991131
.
464. Pach JM, Robertson DM, Taney BS, et al. Prognostic factors in
choroidal and ciliary body melanomas with extrascleral extension. Am J
Ophthalmol 1986;101:325-331. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3953727
.
465. Hykin PG, McCartney AC, Plowman PN, Hungerford JL.
Postenucleation orbital radiotherapy for the treatment of malignant
melanoma of the choroid with extrascleral extension. Br J Ophthalmol
1990;74:36-39. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2106340
.
466. Gunduz K, Shields CL, Shields JA, et al. Plaque radiotherapy for
management of ciliary body and choroidal melanoma with extraocular
extension. Am J Ophthalmol 2000;130:97-102. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11004266
.
467. Ang KK, Peters LJ, Weber RS, et al. Postoperative radiotherapy for
cutaneous melanoma of the head and neck region. Int J Radiat Oncol
Biol Phys 1994;30:795-798. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7960981
.
468. Shields CL, Naseripour M, Cater J, et al. Plaque radiotherapy for
large posterior uveal melanomas (> or =8-mm thick) in 354 consecutive
patients. Ophthalmology 2002;109:1838-1849. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12359604
.
469. Eskelin S, Pyrhonen S, Summanen P, et al. Tumor doubling times
in metastatic malignant melanoma of the uvea: tumor progression before
and after treatment. Ophthalmology 2000;107:1443-1449. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10919885
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-83
470. Kreusel KM, Bechrakis N, Riese J, et al. Combined brachytherapy
and transpupillary thermotherapy for large choroidal melanoma: tumor
regression and early complications. Graefes Arch Clin Exp Ophthalmol
2006;244:1575-1580. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16738857
.
471. Sanke RF, Collin JR, Garner A, Packard RB. Local recurrence of
choroidal malignant melanoma following enucleation. Br J Ophthalmol
1981;65:846-849. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7317332
.
472. Boudinet M, Berges O, Le Huerou JY, et al. Quantitative
echography in the follow-up of patients treated with proton-beam
irradiation for primary choroidal melanomas. Ultrasound Med Biol
2007;33:1046-1056. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17448588
.
473. Gragoudas ES, Lane AM, Munzenrider J, et al. Long-term risk of
local failure after proton therapy for choroidal/ciliary body melanoma.
Trans Am Ophthalmol Soc 2002;100:43-48; discussion 48-49. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/12545676
.
474. Kim TW, Choi E, Park J, et al. Clinical Outcomes of Proton Beam
Therapy for Choroidal Melanoma at a Single Institute in Korea. Cancer
Res Treat 2018;50:335-344. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28421723
.
475. Naeser P, Blomquist E, Montelius A, Thoumas KA. Proton
irradiation of malignant uveal melanoma. A five year follow-up of patients
treated in Uppsala, Sweden. Ups J Med Sci 1998;103:203-211. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/10052109
.
476. Egger E, Zografos L, Schalenbourg A, et al. Eye retention after
proton beam radiotherapy for uveal melanoma. Int J Radiat Oncol Biol
Phys 2003;55:867-880. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12605964
.
477. Design and methods of a clinical trial for a rare condition: the
Collaborative Ocular Melanoma Study. COMS Report No. 3. Control Clin
Trials 1993;14:362-391. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8222668
.
478. Marchini G, Gerosa M, Piovan E, et al. Gamma Knife stereotactic
radiosurgery for uveal melanoma: clinical results after 2 years. Stereotact
Funct Neurosurg 1996;66 Suppl 1:208-213. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9032863
.
479. Salvi SM, Aziz HA, Dar S, et al. Uveal Melanoma Regression after
Brachytherapy: Relationship with Chromosome 3 Monosomy Status.
Ocul Oncol Pathol 2017;3:87-94. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28868276
.
480. Diener-West M, Reynolds SM, Agugliaro DJ, et al. Screening for
metastasis from choroidal melanoma: the Collaborative Ocular
Melanoma Study Group Report 23. J Clin Oncol 2004;22:2438-2444.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/15197206
.
481. Diener-West M, Reynolds SM, Agugliaro DJ, et al. Development of
metastatic disease after enrollment in the COMS trials for treatment of
choroidal melanoma: Collaborative Ocular Melanoma Study Group
Report No. 26. Arch Ophthalmol 2005;123:1639-1643. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16344433
.
482. Collaborative Ocular Melanoma Study G. Ten-year follow-up of
fellow eyes of patients enrolled in Collaborative Ocular Melanoma Study
randomized trials: COMS report no. 22. Ophthalmology 2004;111:966-
976. Available at: https://www.ncbi.nlm.nih.gov/pubmed/15121376
.
483. Scott JF, Vyas R, Galvin J, et al. Primary bilateral uveal melanoma:
a population-based study and systematic review. Clin Exp Ophthalmol
2018;46:502-510. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29219254
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-84
484. Khetan V, Ambatipudi S, Gopal L. Presumed metastasis of
choroidal melanoma to the contralateral choroid. Retin Cases Brief Rep
2010;4:387-389. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25390926
.
485. Shields JA, Shields CL, Shakin EP, Kobetz LE. Metastasis of
choroidal melanoma to the contralateral choroid, orbit, and eyelid. Br J
Ophthalmol 1988;72:456-460. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3390423
.
486. Singh AD, Shields JA, Shields CL, Sato T. Choroidal melanoma
metastatic to the contralateral choroid. Am J Ophthalmol 2001;132:941-
943. Available at: https://www.ncbi.nlm.nih.gov/pubmed/11730671
.
487. Torossian NM, Wallace RT, Hwu WJ, Bedikian AY. Metastasis of
ciliary body melanoma to the contralateral eye: a case report and review
of uveal melanoma literature. Case Rep Oncol Med 2015;2015:427163.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/25874144
.
488. Bourla DH, Young TA. Treatment considerations for primary uveal
melanoma with choroidal metastasis to the fellow eye. Semin
Ophthalmol 2007;22:175-177. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17763240
.
489. Caminal Mitjana JM, Vila Grane N, Adan Civera A, et al. Clinical
Course of a Presumed Metastatic Uveal Melanoma to the Contralateral
Choroid. Semin Ophthalmol 2015;30:417-419. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24171794
.
490. Coupland SE, Sidiki S, Clark BJ, et al. Metastatic choroidal
melanoma to the contralateral orbit 40 years after enucleation. Arch
Ophthalmol 1996;114:751-756. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8639093
.
491. George S, Cooke CA, Mc Ginnity GF, et al. Treated choroidal
melanoma with late metastases to the contralateral orbit. Clin Med
Pathol 2009;2:5-8. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21151543
.
492. Belfort RN, Muller MS, Isenberg J, et al. Metastatic choroidal
melanoma to the contralateral eye: a rare case. Arq Bras Oftalmol
2018;81:517-519. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30328940
.
493. Abrahamsson M. Malignant melanoma of the choroid and the ciliary
body 1956-1975 in Halland and Gothenburg. Incidence, histopathology
and prognosis. Acta Ophthalmol (Copenh) 1983;61:600-610. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/6637421
.
494. Crowley NJ, Seigler HF. Late recurrence of malignant melanoma.
Analysis of 168 patients. Ann Surg 1990;212:173-177. Available at:
http://www.ncbi.nlm.nih.gov/pubmed/2375648
.
495. Gragoudas ES, Egan KM, Seddon JM, et al. Survival of patients
with metastases from uveal melanoma. Ophthalmology 1991;98:383-
389; discussion 390. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/2023760
.
496. Eskelin S, Pyrhonen S, Summanen P, et al. Screening for
metastatic malignant melanoma of the uvea revisited. Cancer
1999;85:1151-1159. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10091801
.
497. Eskelin S, Pyrhonen S, Hahka-Kemppinen M, et al. A prognostic
model and staging for metastatic uveal melanoma. Cancer 2003;97:465-
475. Available at: https://www.ncbi.nlm.nih.gov/pubmed/12518371
.
498. Finger PT, Kurli M, Reddy S, et al. Whole body PET/CT for initial
staging of choroidal melanoma. Br J Ophthalmol 2005;89:1270-1274.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16170114
.
499. Kurli M, Reddy S, Tena LB, et al. Whole body positron emission
tomography/computed tomography staging of metastatic choroidal
melanoma. Am J Ophthalmol 2005;140:193-199. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15992753
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-85
500. Piperno-Neumann S, Servois V, Mariani P, et al. Prospective study
of surveillance testing for metastasis in 100 high-risk uveal melanoma
patients. J Fr Ophtalmol 2015;38:526-534. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25978872
.
501. Kim JH, Shin SJ, Heo SJ, et al. Prognoses and Clinical Outcomes
of Primary and Recurrent Uveal Melanoma. Cancer Res Treat
2018;50:1238-1251. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29281872
.
502. Correa ZM, Augsburger JJ. Independent Prognostic Significance of
Gene Expression Profile Class and Largest Basal Diameter of Posterior
Uveal Melanomas. Am J Ophthalmol 2016;162:20-27 e21. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26596399
.
503. Cai L, Paez-Escamilla M, Walter SD, et al. Gene Expression
Profiling and PRAME Status Versus Tumor-Node-Metastasis Staging for
Prognostication in Uveal Melanoma. Am J Ophthalmol 2018;195:154-
160. Available at: https://www.ncbi.nlm.nih.gov/pubmed/30092184
.
504. Szalai E, Wells JR, Ward L, Grossniklaus HE. Uveal Melanoma
Nuclear BRCA1-Associated Protein-1 Immunoreactivity Is an Indicator of
Metastasis. Ophthalmology 2018;125:203-209. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28823399
.
505. Delgado-Ramos GM, Thomas F, VanderWalde A, et al. Risk
factors, clinical outcomes, and natural history of uveal melanoma: a
single-institution analysis. Med Oncol 2019;36:17. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30666496
.
506. Drabarek W, Yavuzyigitoglu S, Obulkasim A, et al. Multi-Modality
Analysis Improves Survival Prediction in Enucleated Uveal Melanoma
Patients. Invest Ophthalmol Vis Sci 2019;60:3595-3605. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31425584
.
507. Mazloumi M, Vichitvejpaisal P, Dalvin LA, et al. Accuracy of The
Cancer Genome Atlas Classification vs American Joint Committee on
Cancer Classification for Prediction of Metastasis in Patients With Uveal
Melanoma. JAMA Ophthalmol 2020;138:260-267. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31944225
.
508. Mooy C, Vissers K, Luyten G, et al. DNA flow cytometry in uveal
melanoma: the effect of pre-enucleation irradiation. Br J Ophthalmol
1995;79:174-177. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7696240
.
509. Staby KM, Gravdal K, Mork SJ, et al. Prognostic impact of
chromosomal aberrations and GNAQ, GNA11 and BAP1 mutations in
uveal melanoma. Acta Ophthalmol 2018;96:31-38. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28444874
.
510. Robertson AG, Shih J, Yau C, et al. Integrative Analysis Identifies
Four Molecular and Clinical Subsets in Uveal Melanoma. Cancer Cell
2017;32:204-220 e215. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28810145
.
511. Prescher G, Bornfeld N, Hirche H, et al. Prognostic implications of
monosomy 3 in uveal melanoma. Lancet 1996;347:1222-1225. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/8622452
.
512. Petrausch U, Martus P, Tonnies H, et al. Significance of gene
expression analysis in uveal melanoma in comparison to standard risk
factors for risk assessment of subsequent metastases. Eye (Lond)
2008;22:997-1007. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17384575
.
513. Trolet J, Hupe P, Huon I, et al. Genomic profiling and identification
of high-risk uveal melanoma by array CGH analysis of primary tumors
and liver metastases. Invest Ophthalmol Vis Sci 2009;50:2572-2580.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/19151381
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-86
514. Shields CL, Ganguly A, Bianciotto CG, et al. Prognosis of uveal
melanoma in 500 cases using genetic testing of fine-needle aspiration
biopsy specimens. Ophthalmology 2011;118:396-401. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20869116
.
515. Vaarwater J, van den Bosch T, Mensink HW, et al. Multiplex
ligation-dependent probe amplification equals fluorescence in-situ
hybridization for the identification of patients at risk for metastatic
disease in uveal melanoma. Melanoma Res 2012;22:30-37. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22157614
.
516. Ewens KG, Kanetsky PA, Richards-Yutz J, et al. Genomic profile of
320 uveal melanoma cases: chromosome 8p-loss and metastatic
outcome. Invest Ophthalmol Vis Sci 2013;54:5721-5729. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23821189
.
517. Barnhill RL, Ye M, Batistella A, et al. The biological and prognostic
significance of angiotropism in uveal melanoma. Lab Invest 2017.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28240745
.
518. Vaquero-Garcia J, Lalonde E, Ewens KG, et al. PRiMeUM: A Model
for Predicting Risk of Metastasis in Uveal Melanoma. Invest Ophthalmol
Vis Sci 2017;58:4096-4105. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28828481
.
519. Ewens KG, Lalonde E, Richards-Yutz J, et al. Comparison of
Germline versus Somatic BAP1 Mutations for Risk of Metastasis in Uveal
Melanoma. BMC Cancer 2018;18:1172. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30477459
.
520. Patrone S, Maric I, Rutigliani M, et al. Prognostic value of
chromosomal imbalances, gene mutations, and BAP1 expression in
uveal melanoma. Genes Chromosomes Cancer 2018;57:387-400.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29689622
.
521. Afshar AR, Damato BE, Stewart JM, et al. Next-Generation
Sequencing of Uveal Melanoma for Detection of Genetic Alterations
Predicting Metastasis. Transl Vis Sci Technol 2019;8:18. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31024753
.
522. Shields CL, Dalvin LA, Vichitvejpaisal P, et al. Prognostication of
uveal melanoma is simple and highly predictive using The Cancer
Genome Atlas (TCGA) classification: A review. Indian J Ophthalmol
2019;67:1959-1963. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31755428
.
523. Rodrigues M, Ait Rais K, Salviat F, et al. Association of Partial
Chromosome 3 Deletion in Uveal Melanomas With Metastasis-Free
Survival. JAMA Ophthalmol 2020;138:182-188. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31895446
.
524. Versluis M, de Lange MJ, van Pelt SI, et al. Digital PCR validates
8q dosage as prognostic tool in uveal melanoma. PLoS One
2015;10:e0116371. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25764247
.
525. Chang MY, Rao NP, Burgess BL, et al. Heterogeneity of monosomy
3 in fine needle aspiration biopsy of choroidal melanoma. Mol Vis
2013;19:1892-1900. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24049435
.
526. Onken MD, Worley LA, Ehlers JP, Harbour JW. Gene expression
profiling in uveal melanoma reveals two molecular classes and predicts
metastatic death. Cancer Res 2004;64:7205-7209. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15492234
.
527. Tschentscher F, Husing J, Holter T, et al. Tumor classification
based on gene expression profiling shows that uveal melanomas with
and without monosomy 3 represent two distinct entities. Cancer Res
2003;63:2578-2584. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12750282
.
528. Onken MD, Worley LA, Davila RM, et al. Prognostic testing in uveal
melanoma by transcriptomic profiling of fine needle biopsy specimens. J
Mol Diagn 2006;8:567-573. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17065425
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-87
529. Worley LA, Onken MD, Person E, et al. Transcriptomic versus
chromosomal prognostic markers and clinical outcome in uveal
melanoma. Clin Cancer Res 2007;13:1466-1471. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17332290
.
530. Onken MD, Worley LA, Tuscan MD, Harbour JW. An accurate,
clinically feasible multi-gene expression assay for predicting metastasis
in uveal melanoma. J Mol Diagn 2010;12:461-468. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20413675
.
531. Chappell MC, Char DH, Cole TB, et al. Uveal melanoma: molecular
pattern, clinical features, and radiation response. Am J Ophthalmol
2012;154:227-232 e222. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22541662
.
532. Decatur CL, Ong E, Garg N, et al. Driver Mutations in Uveal
Melanoma: Associations With Gene Expression Profile and Patient
Outcomes. JAMA Ophthalmol 2016;134:728-733. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27123562
.
533. Field MG, Decatur CL, Kurtenbach S, et al. PRAME as an
Independent Biomarker for Metastasis in Uveal Melanoma. Clin Cancer
Res 2016;22:1234-1242. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26933176
.
534. Plasseraud KM, Cook RW, Tsai T, et al. Clinical Performance and
Management Outcomes with the DecisionDx-UM Gene Expression
Profile Test in a Prospective Multicenter Study. J Oncol
2016;2016:5325762. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27446211
.
535. Walter SD, Chao DL, Feuer W, et al. Prognostic Implications of
Tumor Diameter in Association With Gene Expression Profile for Uveal
Melanoma. JAMA Ophthalmol 2016;134:734-740. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27123792
.
536. Koopmans AE, Verdijk RM, Brouwer RW, et al. Clinical significance
of immunohistochemistry for detection of BAP1 mutations in uveal
melanoma. Mod Pathol 2014;27:1321-1330. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24633195
.
537. Jha J, Singh MK, Singh L, et al. Expression of BAP1 and ATM
proteins: Association with AJCC tumor category in uveal melanoma. Ann
Diagn Pathol 2019;44:151432. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31864162
.
538. Szalai E, Jiang Y, van Poppelen NM, et al. Association of Uveal
Melanoma Metastatic Rate With Stochastic Mutation Rate and Type of
Mutation. JAMA Ophthalmol 2018;136:1115-1120. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30073324
.
539. Yavuzyigitoglu S, Koopmans AE, Verdijk RM, et al. Uveal
Melanomas with SF3B1 Mutations: A Distinct Subclass Associated with
Late-Onset Metastases. Ophthalmology 2016;123:1118-1128. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/26923342
.
540. Yavuzyigitoglu S, Mensink HW, Smit KN, et al. Metastatic Disease
in Polyploid Uveal Melanoma Patients Is Associated With BAP1
Mutations. Invest Ophthalmol Vis Sci 2016;57:2232-2239. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27116551
.
541. Schefler AC, Koca E, Bernicker EH, Correa ZM. Relationship
between clinical features, GEP class, and PRAME expression in uveal
melanoma. Graefes Arch Clin Exp Ophthalmol 2019;257:1541-1545.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/31065847
.
542. Gombos DS, Van Quill KR, Uusitalo M, O'Brien JM. Geographic
disparities in diagnostic screening for metastatic uveal melanoma.
Ophthalmology 2004;111:2254-2258. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15582082
.
543. Felberg NT, Shields JA, Maguire J, et al. Gamma-glutamyl
transpeptidase in the prognosis of patients with uveal malignant
melanoma. Am J Ophthalmol 1983;95:467-473. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/6132555
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-88
544. Hicks C, Foss AJ, Hungerford JL. Predictive power of screening
tests for metastasis in uveal melanoma. Eye (Lond) 1998;12 ( Pt 6):945-
948. Available at: https://www.ncbi.nlm.nih.gov/pubmed/10325992
.
545. Kaiserman I, Amer R, Pe'er J. Liver function tests in metastatic
uveal melanoma. Am J Ophthalmol 2004;137:236-243. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14962411
.
546. Patel M, Winston CB, Marr BP, et al. Characterization of computed
tomography scan abnormalities in patients with biopsy-proven hepatic
metastases from uveal melanoma. Arch Ophthalmol 2011;129:1576-
1582. Available at: https://www.ncbi.nlm.nih.gov/pubmed/22159677
.
547. Mouriaux F, Diorio C, Bergeron D, et al. Liver function testing is not
helpful for early diagnosis of metastatic uveal melanoma. Ophthalmology
2012;119:1590-1595. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22683062
.
548. Wagoner MD, Albert DM. The incidence of metastases from
untreated ciliary body and choroidal melanoma. Arch Ophthalmol
1982;100:939-940. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/7092632
.
549. Hendler K, Pe'er J, Kaiserman I, et al. Trends in liver function tests:
a comparison with serum tumor markers in metastatic uveal melanoma
(part 2). Anticancer Res 2011;31:351-357. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21273623
.
550. Semelka RC, Martin DR, Balci C, Lance T. Focal liver lesions:
comparison of dual-phase CT and multisequence multiplanar MR
imaging including dynamic gadolinium enhancement. J Magn Reson
Imaging 2001;13:397-401. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11241813
.
551. Francken AB, Fulham MJ, Millward MJ, Thompson JF. Detection of
metastatic disease in patients with uveal melanoma using positron
emission tomography. Eur J Surg Oncol 2006;32:780-784. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16765562
.
552. Maeda T, Tateishi U, Suzuki S, et al. Magnetic resonance screening
trial for hepatic metastasis in patients with locally controlled choroidal
melanoma. Jpn J Clin Oncol 2007;37:282-286. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17553818
.
553. Strobel K, Bode B, Dummer R, et al. Limited value of 18F-FDG
PET/CT and S-100B tumour marker in the detection of liver metastases
from uveal melanoma compared to liver metastases from cutaneous
melanoma. Eur J Nucl Med Mol Imaging 2009;36:1774-1782. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/19495748
.
554. Servois V, Mariani P, Malhaire C, et al. Preoperative staging of liver
metastases from uveal melanoma by magnetic resonance imaging (MRI)
and fluorodeoxyglucose-positron emission tomography (FDG-PET). Eur
J Surg Oncol 2010;36:189-194. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19775851
.
555. Orcurto V, Denys A, Voelter V, et al. (18)F-fluorodeoxyglucose
positron emission tomography/computed tomography and magnetic
resonance imaging in patients with liver metastases from uveal
melanoma: results from a pilot study. Melanoma Res 2012;22:63-69.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/22027909
.
556. Marshall E, Romaniuk C, Ghaneh P, et al. MRI in the detection of
hepatic metastases from high-risk uveal melanoma: a prospective study
in 188 patients. Br J Ophthalmol 2013;97:159-163. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23159448
.
557. Wagner M, Mariani P, Bidard FC, et al. Diffusion-weighted MRI for
uveal melanoma liver metastasis detection. Eur Radiol 2015;25:2263-
2273. Available at: https://www.ncbi.nlm.nih.gov/pubmed/25712144
.
558. Choudhary MM, Gupta A, Bena J, et al. Hepatic Ultrasonography
for Surveillance in Patients With Uveal Melanoma. JAMA Ophthalmol
2016;134:174-180. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26633182
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-89
559. Cohen VML, Pavlidou E, DaCosta J, et al. Staging Uveal Melanoma
with Whole-Body Positron-Emission Tomography/Computed
Tomography and Abdominal Ultrasound: Low Incidence of Metastatic
Disease, High Incidence of Second Primary Cancers. Middle East Afr J
Ophthalmol 2018;25:91-95. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30122854
.
560. Rantala ES, Peltola E, Helminen H, et al. Hepatic Ultrasonography
Compared With Computed Tomography and Magnetic Resonance
Imaging at Diagnosis of Metastatic Uveal Melanoma. Am J Ophthalmol
2020;216:156-164. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32278769
.
561. Wen JC, Sai V, Straatsma BR, McCannel TA. Radiation-related
cancer risk associated with surveillance imaging for metastasis from
choroidal melanoma. JAMA Ophthalmol 2013;131:56-61. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23307209
.
562. Kodjikian L, Grange JD, Baldo S, et al. Prognostic factors of liver
metastases from uveal melanoma. Graefes Arch Clin Exp Ophthalmol
2005;243:985-993. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15891893
.
563. Rivoire M, Kodjikian L, Baldo S, et al. Treatment of liver metastases
from uveal melanoma. Ann Surg Oncol 2005;12:422-428. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15886904
.
564. Kim IK, Lane AM, Gragoudas ES. Survival in patients with
presymptomatic diagnosis of metastatic uveal melanoma. Arch
Ophthalmol 2010;128:871-875. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20625048
.
565. Chin K, Finger PT, Kurli M, et al. Second cancers discovered by
(18)FDG PET/CT imaging for choroidal melanoma. Optometry
2007;78:396-401. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17662928
.
566. Diener-West M, Reynolds SM, Agugliaro DJ, et al. Second primary
cancers after enrollment in the COMS trials for treatment of choroidal
melanoma: COMS Report No. 25. Arch Ophthalmol 2005;123:601-604.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/15883277
.
567. Kujala E, Makitie T, Kivela T. Very long-term prognosis of patients
with malignant uveal melanoma. Invest Ophthalmol Vis Sci
2003;44:4651-4659. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14578381
.
568. Caminal JM, Martinez J, Arias LL, et al. Multiple neoplasms in
patients with uveal melanoma. Arch Soc Esp Oftalmol 2007;82:535-540.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/17846942
.
569. Augsburger JJ, Correa ZM, Shaikh AH. Effectiveness of treatments
for metastatic uveal melanoma. Am J Ophthalmol 2009;148:119-127.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/19375060
.
570. Valpione S, Moser JC, Parrozzani R, et al. Development and
external validation of a prognostic nomogram for metastatic uveal
melanoma. PLoS One 2015;10:e0120181. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25780931
.
571. Nicholas MN, Khoja L, Atenafu EG, et al. Prognostic factors for first-
line therapy and overall survival of metastatic uveal melanoma: The
Princess Margaret Cancer Centre experience. Melanoma Res
2018;28:571-577. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30067547
.
572. Lorenzo D, Piulats JM, Ochoa M, et al. Clinical predictors of survival
in metastatic uveal melanoma. Jpn J Ophthalmol 2019;63:197-209.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/30796549
.
573. Mariani P, Dureau S, Savignoni A, et al. Development of a
Prognostic Nomogram for Liver Metastasis of Uveal Melanoma Patients
Selected by Liver MRI. Cancers (Basel) 2019;11. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31234340
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-90
574. Jochems A, van der Kooij MK, Fiocco M, et al. Metastatic Uveal
Melanoma: Treatment Strategies and Survival-Results from the Dutch
Melanoma Treatment Registry. Cancers (Basel) 2019;11. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31323802
.
575. Gragoudas ES, Egan KM, Seddon JM, et al. Intraocular recurrence
of uveal melanoma after proton beam irradiation. Ophthalmology
1992;99:760-766. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/1594223
.
576. Marucci L, Lane AM, Li W, et al. Conservation treatment of the eye:
Conformal proton reirradiation for recurrent uveal melanoma. Int J Radiat
Oncol Biol Phys 2006;64:1018-1022. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16376492
.
577. Marucci L, Ancukiewicz M, Lane AM, et al. Uveal melanoma
recurrence after fractionated proton beam therapy: comparison of
survival in patients treated with reirradiation or with enucleation. Int J
Radiat Oncol Biol Phys 2011;79:842-846. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20472356
.
578. Seibel I, Cordini D, Rehak M, et al. Local Recurrence After Primary
Proton Beam Therapy in Uveal Melanoma: Risk Factors, Retreatment
Approaches, and Outcome. Am J Ophthalmol 2015;160:628-636.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/26133249
.
579. King B, Morales-Tirado VM, Wynn HG, et al. Repeat Episcleral
Plaque Brachytherapy: Clinical Outcomes in Patients Treated for Locally
Recurrent Posterior Uveal Melanoma. Am J Ophthalmol 2017;176:40-45.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28048976
.
580. Rantala ES, Hernberg M, Kivela TT. Overall survival after treatment
for metastatic uveal melanoma: a systematic review and meta-analysis.
Melanoma Res 2019;29:561-568. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30664106
.
581. Rowcroft A, Loveday BPT, Thomson BNJ, et al. Systematic review
of liver directed therapy for uveal melanoma hepatic metastases. HPB
(Oxford) 2020;22:497-505. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31791894
.
582. Frenkel S, Nir I, Hendler K, et al. Long-term survival of uveal
melanoma patients after surgery for liver metastases. Br J Ophthalmol
2009;93:1042-1046. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19429579
.
583. Kivela TT, Piperno-Neumann S, Desjardins L, et al. Validation of a
Prognostic Staging for Metastatic Uveal Melanoma: A Collaborative
Study of the European Ophthalmic Oncology Group. Am J Ophthalmol
2016;168:217-226. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27296487
.
584. Xu LT, Funchain PF, Bena JF, et al. Uveal Melanoma Metastatic to
the Liver: Treatment Trends and Outcomes. Ocul Oncol Pathol
2019;5:323-332. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31559243
.
585. Hsueh EC, Essner R, Foshag LJ, et al. Prolonged survival after
complete resection of metastases from intraocular melanoma. Cancer
2004;100:122-129. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/14692032
.
586. Augsburger JJ, Correa ZM, Shaikh AH. Quality of evidence about
effectiveness of treatments for metastatic uveal melanoma. Trans Am
Ophthalmol Soc 2008;106:128-135; discussion 135-127. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19277228
.
587. Buder K, Gesierich A, Gelbrich G, Goebeler M. Systemic treatment
of metastatic uveal melanoma: review of literature and future
perspectives. Cancer Med 2013;2:674-686. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24403233
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-91
588. Barnhill R, Vermeulen P, Daelemans S, et al. Replacement and
desmoplastic histopathological growth patterns: A pilot study of
prediction of outcome in patients with uveal melanoma liver metastases.
J Pathol Clin Res 2018;4:227-240. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29917326
.
589. Agarwala SS, Eggermont AM, O'Day S, Zager JS. Metastatic
melanoma to the liver: a contemporary and comprehensive review of
surgical, systemic, and regional therapeutic options. Cancer
2014;120:781-789. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24301420
.
590. Leyvraz S, Piperno-Neumann S, Suciu S, et al. Hepatic intra-arterial
versus intravenous fotemustine in patients with liver metastases from
uveal melanoma (EORTC 18021): a multicentric randomized trial. Ann
Oncol 2014;25:742-746. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24510314
.
591. Salmon RJ, Levy C, Plancher C, et al. Treatment of liver
metastases from uveal melanoma by combined surgery-chemotherapy.
Eur J Surg Oncol 1998;24:127-130. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9591028
.
592. Mariani P, Piperno-Neumann S, Servois V, et al. Surgical
management of liver metastases from uveal melanoma: 16 years'
experience at the Institut Curie. Eur J Surg Oncol 2009;35:1192-1197.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/19329272
.
593. Ryu SW, Saw R, Scolyer RA, et al. Liver resection for metastatic
melanoma: equivalent survival for cutaneous and ocular primaries. J
Surg Oncol 2013;108:129-135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23813600
.
594. Gomez D, Wetherill C, Cheong J, et al. The Liverpool uveal
melanoma liver metastases pathway: outcome following liver resection. J
Surg Oncol 2014;109:542-547. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24357463
.
595. Akyuz M, Yazici P, Dural C, et al. Laparoscopic management of
liver metastases from uveal melanoma. Surg Endosc 2016;30:2567-
2571. Available at: https://www.ncbi.nlm.nih.gov/pubmed/26310535
.
596. Mariani P, Almubarak MM, Kollen M, et al. Radiofrequency ablation
and surgical resection of liver metastases from uveal melanoma. Eur J
Surg Oncol 2016;42:706-712. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26968227
.
597. Aoyama T, Mastrangelo MJ, Berd D, et al. Protracted survival after
resection of metastatic uveal melanoma. Cancer 2000;89:1561-1568.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/11013372
.
598. Adam R, Chiche L, Aloia T, et al. Hepatic resection for
noncolorectal nonendocrine liver metastases: analysis of 1,452 patients
and development of a prognostic model. Ann Surg 2006;244:524-535.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16998361
.
599. de Ridder J, van Walsum M, Verhoef C, et al. Hepatic resection for
metastatic melanoma in The Netherlands: survival and prognostic
factors. Melanoma Res 2013;23:27-32. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23164992
.
600. Yang XY, Xie F, Tao R, et al. Treatment of liver metastases from
uveal melanoma: a retrospective single-center analysis. Hepatobiliary
Pancreat Dis Int 2013;12:602-606. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24322745
.
601. Servois V, Bouhadiba T, Dureau S, et al. Iterative treatment with
surgery and radiofrequency ablation of uveal melanoma liver metastasis:
Retrospective analysis of a series of very long-term survivors. Eur J Surg
Oncol 2019;45:1717-1722. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31266665
.
602. Pawlik TM, Zorzi D, Abdalla EK, et al. Hepatic resection for
metastatic melanoma: distinct patterns of recurrence and prognosis for
ocular versus cutaneous disease. Ann Surg Oncol 2006;13:712-720.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/16538410
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-92
603. Eschelman DJ, Gonsalves CF, Sato T. Transhepatic therapies for
metastatic uveal melanoma. Semin Intervent Radiol 2013;30:39-48.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/24436516
.
604. Glazer ES, Zager JS. Chemosaturation With Percutaneous Hepatic
Perfusion in Unresectable Hepatic Metastases. Cancer Control
2017;24:96-101. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28178721
.
605. Broman KK, Zager JS. Intra-arterial perfusion-based therapies for
regionally metastatic cutaneous and uveal melanoma. Melanoma Manag
2019;6:MMT26. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31807277
.
606. Breedis C, Young G. The blood supply of neoplasms in the liver.
Am J Pathol 1954;30:969-977. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/13197542
.
607. Reddy SK, Kesmodel SB, Alexander HR, Jr. Isolated hepatic
perfusion for patients with liver metastases. Ther Adv Med Oncol
2014;6:180-194. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25057304
.
608. Alexander HR, Libutti SK, Bartlett DL, et al. A phase I-II study of
isolated hepatic perfusion using melphalan with or without tumor
necrosis factor for patients with ocular melanoma metastatic to liver. Clin
Cancer Res 2000;6:3062-3070. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10955785
.
609. Alexander HR, Jr., Libutti SK, Pingpank JF, et al. Hyperthermic
isolated hepatic perfusion using melphalan for patients with ocular
melanoma metastatic to liver. Clin Cancer Res 2003;9:6343-6349.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/14695133
.
610. Noter SL, Rothbarth J, Pijl ME, et al. Isolated hepatic perfusion with
high-dose melphalan for the treatment of uveal melanoma metastases
confined to the liver. Melanoma Res 2004;14:67-72. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15091197
.
611. Olofsson R, Cahlin C, All-Ericsson C, et al. Isolated hepatic
perfusion for ocular melanoma metastasis: registry data suggests a
survival benefit. Ann Surg Oncol 2014;21:466-472. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24141377
.
612. Varghese S, Xu H, Bartlett D, et al. Isolated hepatic perfusion with
high-dose melphalan results in immediate alterations in tumor gene
expression in patients with metastatic ocular melanoma. Ann Surg Oncol
2010;17:1870-1877. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20221901
.
613. Ben-Shabat I, Belgrano V, Ny L, et al. Long-Term Follow-Up
Evaluation of 68 Patients with Uveal Melanoma Liver Metastases
Treated with Isolated Hepatic Perfusion. Ann Surg Oncol 2016;23:1327-
1334. Available at: https://www.ncbi.nlm.nih.gov/pubmed/26628434
.
614. van Iersel LB, Hoekman EJ, Gelderblom H, et al. Isolated hepatic
perfusion with 200 mg melphalan for advanced noncolorectal liver
metastases. Ann Surg Oncol 2008;15:1891-1898. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18470571
.
615. Ben-Shabat I, Belgrano V, Hansson C, Olofsson Bagge R. The
effect of perfusate buffering on toxicity and response in isolated hepatic
perfusion for uveal melanoma liver metastases. Int J Hyperthermia
2017;33:483-488. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28110582
.
616. de Leede EM, Burgmans MC, Kapiteijn E, et al. Isolated (hypoxic)
hepatic perfusion with high-dose chemotherapy in patients with
unresectable liver metastases of uveal melanoma: results from two
experienced centres. Melanoma Res 2016;26:588-594. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27513071
.
617. Cantore M, Fiorentini G, Aitini E, et al. Intra-arterial hepatic
carboplatin-based chemotherapy for ocular melanoma metastatic to the
liver. Report of a phase II study. Tumori 1994;80:37-39. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/8191596
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-93
618. Leyvraz S, Spataro V, Bauer J, et al. Treatment of ocular melanoma
metastatic to the liver by hepatic arterial chemotherapy. J Clin Oncol
1997;15:2589-2595. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9215829
.
619. Egerer G, Lehnert T, Max R, et al. Pilot study of hepatic intraarterial
fotemustine chemotherapy for liver metastases from uveal melanoma: a
single-center experience with seven patients. Int J Clin Oncol 2001;6:25-
28. Available at: https://www.ncbi.nlm.nih.gov/pubmed/11706523
.
620. Vera-Aguilera J, Bedikian AY, Bassett RL, et al. Phase I/II Study of
Hepatic Arterial Infusion of Nab-paclitaxel in Patients With Metastatic
Melanoma to the Liver. Am J Clin Oncol 2018;41:1132-1136. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/29509591
.
621. Jansen YJL, Verset G, Schats K, et al. Phase I clinical trial of
decitabine (5-aza-2'-deoxycytidine) administered by hepatic arterial
infusion in patients with unresectable liver-predominant metastases.
ESMO Open 2019;4:e000464. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30962963
.
622. Peters S, Voelter V, Zografos L, et al. Intra-arterial hepatic
fotemustine for the treatment of liver metastases from uveal melanoma:
experience in 101 patients. Ann Oncol 2006;17:578-583. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16469752
.
623. Siegel R, Hauschild A, Kettelhack C, et al. Hepatic arterial
Fotemustine chemotherapy in patients with liver metastases from
cutaneous melanoma is as effective as in ocular melanoma. Eur J Surg
Oncol 2007;33:627-632. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17196362
.
624. Melichar B, Voboril Z, Lojik M, Krajina A. Liver metastases from
uveal melanoma: clinical experience of hepatic arterial infusion of
cisplatin, vinblastine and dacarbazine. Hepatogastroenterology
2009;56:1157-1162. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19760961
.
625. Farolfi A, Ridolfi L, Guidoboni M, et al. Liver metastases from
melanoma: hepatic intra-arterial chemotherapy. A retrospective study. J
Chemother 2011;23:300-305. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22005064
.
626. Heusner TA, Antoch G, Wittkowski-Sterczewski A, et al.
Transarterial hepatic chemoperfusion of uveal melanoma metastases:
survival and response to treatment. Rofo 2011;183:1151-1160. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/22033849
.
627. de Leede EM, Burgmans MC, Meijer TS, et al. Prospective Clinical
and Pharmacological Evaluation of the Delcath System's Second-
Generation (GEN2) Hemofiltration System in Patients Undergoing
Percutaneous Hepatic Perfusion with Melphalan. Cardiovasc Intervent
Radiol 2017;40:1196-1205. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28451811
.
628. Hughes MS, Zager J, Faries M, et al. Results of a Randomized
Controlled Multicenter Phase III Trial of Percutaneous Hepatic Perfusion
Compared with Best Available Care for Patients with Melanoma Liver
Metastases. Ann Surg Oncol 2016;23:1309-1319. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26597368
.
629. Meijer TS, Burgmans MC, Fiocco M, et al. Safety of Percutaneous
Hepatic Perfusion with Melphalan in Patients with Unresectable Liver
Metastases from Ocular Melanoma Using the Delcath Systems' Second-
Generation Hemofiltration System: A Prospective Non-randomized
Phase II Trial. Cardiovasc Intervent Radiol 2019;42:841-852. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/30767147
.
630. Vogl TJ, Koch SA, Lotz G, et al. Percutaneous Isolated Hepatic
Perfusion as a Treatment for Isolated Hepatic Metastases of Uveal
Melanoma: Patient Outcome and Safety in a Multi-centre Study.
Cardiovasc Intervent Radiol 2017;40:864-872. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28144756
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-94
631. Karydis I, Gangi A, Wheater MJ, et al. Percutaneous hepatic
perfusion with melphalan in uveal melanoma: A safe and effective
treatment modality in an orphan disease. J Surg Oncol 2018;117:1170-
1178. Available at: https://www.ncbi.nlm.nih.gov/pubmed/29284076
.
632. Artzner C, Mossakowski O, Hefferman G, et al. Chemosaturation
with percutaneous hepatic perfusion of melphalan for liver-dominant
metastatic uveal melanoma: a single center experience. Cancer Imaging
2019;19:31. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31146793
.
633. Mavligit GM, Charnsangavej C, Carrasco CH, et al. Regression of
ocular melanoma metastatic to the liver after hepatic arterial
chemoembolization with cisplatin and polyvinyl sponge. JAMA
1988;260:974-976. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/3398202
.
634. Bedikian AY, Legha SS, Mavligit G, et al. Treatment of uveal
melanoma metastatic to the liver: a review of the M. D. Anderson Cancer
Center experience and prognostic factors. Cancer 1995;76:1665-1670.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/8635073
.
635. Dayani PN, Gould JE, Brown DB, et al. Hepatic metastasis from
uveal melanoma: angiographic pattern predictive of survival after hepatic
arterial chemoembolization. Arch Ophthalmol 2009;127:628-632.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/19433711
.
636. Schuster R, Lindner M, Wacker F, et al. Transarterial
chemoembolization of liver metastases from uveal melanoma after
failure of systemic therapy: toxicity and outcome. Melanoma Res
2010;20:191-196. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20335820
.
637. Gupta S, Bedikian AY, Ahrar J, et al. Hepatic artery
chemoembolization in patients with ocular melanoma metastatic to the
liver: response, survival, and prognostic factors. Am J Clin Oncol
2010;33:474-480. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19935383
.
638. Duran R, Chapiro J, Frangakis C, et al. Uveal Melanoma Metastatic
to the Liver: The Role of Quantitative Volumetric Contrast-Enhanced MR
Imaging in the Assessment of Early Tumor Response after Transarterial
Chemoembolization. Transl Oncol 2014;7:447-455. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24953419
.
639. Gonsalves CF, Eschelman DJ, Thornburg B, et al. Uveal Melanoma
Metastatic to the Liver: Chemoembolization With 1,3-Bis-(2-Chloroethyl)-
1-Nitrosourea. AJR Am J Roentgenol 2015;205:429-433. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25905562
.
640. Shibayama Y, Namikawa K, Sone M, et al. Efficacy and toxicity of
transarterial chemoembolization therapy using cisplatin and gelatin
sponge in patients with liver metastases from uveal melanoma in an
Asian population. Int J Clin Oncol 2017;22:577-584. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28144882
.
641. Patel K, Sullivan K, Berd D, et al. Chemoembolization of the hepatic
artery with BCNU for metastatic uveal melanoma: results of a phase II
study. Melanoma Res 2005;15:297-304. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16034309
.
642. Huppert PE, Fierlbeck G, Pereira P, et al. Transarterial
chemoembolization of liver metastases in patients with uveal melanoma.
Eur J Radiol 2010;74:e38-44. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19467811
.
643. Yamamoto A, Chervoneva I, Sullivan KL, et al. High-dose
immunoembolization: survival benefit in patients with hepatic metastases
from uveal melanoma. Radiology 2009;252:290-298. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19561263
.
644. Edelhauser G, Schicher N, Berzaczy D, et al. Fotemustine
chemoembolization of hepatic metastases from uveal melanoma: a
retrospective single-center analysis. AJR Am J Roentgenol
2012;199:1387-1392. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23169735
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-95
645. Vogl T, Eichler K, Zangos S, et al. Preliminary experience with
transarterial chemoembolization (TACE) in liver metastases of uveal
malignant melanoma: local tumor control and survival. J Cancer Res Clin
Oncol 2007;133:177-184. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/17021903
.
646. Fiorentini G, Aliberti C, Del Conte A, et al. Intra-arterial hepatic
chemoembolization (TACE) of liver metastases from ocular melanoma
with slow-release irinotecan-eluting beads. Early results of a phase II
clinical study. In Vivo 2009;23:131-137. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19368137
.
647. Rostas JW, Tam AL, Sato T, et al. Health-related quality of life
during trans-arterial chemoembolization with drug-eluting beads loaded
with doxorubicin (DEBDOX) for unresectable hepatic metastases from
ocular melanoma. Am J Surg 2017;214:884-890. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28754534
.
648. Carling U, Dorenberg EJ, Haugvik SP, et al. Transarterial
Chemoembolization of Liver Metastases from Uveal Melanoma Using
Irinotecan-Loaded Beads: Treatment Response and Complications.
Cardiovasc Intervent Radiol 2015;38:1532-1541. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25832764
.
649. Valpione S, Aliberti C, Parrozzani R, et al. A retrospective analysis
of 141 patients with liver metastases from uveal melanoma: a two-cohort
study comparing transarterial chemoembolization with CPT-11 charged
microbeads and historical treatments. Melanoma Res 2015;25:164-168.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/25521594
.
650. Sharma KV, Gould JE, Harbour JW, et al. Hepatic arterial
chemoembolization for management of metastatic melanoma. AJR Am J
Roentgenol 2008;190:99-104. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/18094299
.
651. Sato T, Eschelman DJ, Gonsalves CF, et al. Immunoembolization of
malignant liver tumors, including uveal melanoma, using granulocyte-
macrophage colony-stimulating factor. J Clin Oncol 2008;26:5436-5442.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/18838710
.
652. Valsecchi ME, Terai M, Eschelman DJ, et al. Double-blinded,
randomized phase II study using embolization with or without
granulocyte-macrophage colony-stimulating factor in uveal melanoma
with hepatic metastases. J Vasc Interv Radiol 2015;26:523-532 e522.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/25678394
.
653. Kennedy AS, Nutting C, Jakobs T, et al. A first report of
radioembolization for hepatic metastases from ocular melanoma. Cancer
Invest 2009;27:682-690. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19219675
.
654. Gonsalves CF, Eschelman DJ, Sullivan KL, et al. Radioembolization
as salvage therapy for hepatic metastasis of uveal melanoma: a single-
institution experience. AJR Am J Roentgenol 2011;196:468-473.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/21257902
.
655. Klingenstein A, Haug AR, Zech CJ, Schaller UC. Radioembolization
as locoregional therapy of hepatic metastases in uveal melanoma
patients. Cardiovasc Intervent Radiol 2013;36:158-165. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22526099
.
656. Eldredge-Hindy H, Ohri N, Anne PR, et al. Yttrium-90 Microsphere
Brachytherapy for Liver Metastases From Uveal Melanoma: Clinical
Outcomes and the Predictive Value of Fluorodeoxyglucose Positron
Emission Tomography. Am J Clin Oncol 2016;39:189-195. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24441583
.
657. Tulokas S, Maenpaa H, Peltola E, et al. Selective internal radiation
therapy (SIRT) as treatment for hepatic metastases of uveal melanoma:
a Finnish nation-wide retrospective experience. Acta Oncol
2018;57:1373-1380. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29683787
.
658. Ponti A, Denys A, Digklia A, et al. First-Line Selective Internal
Radiation Therapy in Patients with Uveal Melanoma Metastatic to the
Liver. J Nucl Med 2020;61:350-356. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31481579
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-96
659. Gonsalves CF, Eschelman DJ, Adamo RD, et al. A Prospective
Phase II Trial of Radioembolization for Treatment of Uveal Melanoma
Hepatic Metastasis. Radiology 2019;293:223-231. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31453767
.
660. Putzer D, Schullian P, Bale R. Locoregional ablative treatment of
melanoma metastases. Int J Hyperthermia 2019;36:59-63. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31537161
.
661. Holzwanger DJ, Madoff DC. Role of interventional radiology in the
management of hepatocellular carcinoma: current status. Chin Clin
Oncol 2018;7:49. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30180753
.
662. Glassberg MB, Ghosh S, Clymer JW, et al. Microwave ablation
compared with radiofrequency ablation for treatment of hepatocellular
carcinoma and liver metastases: a systematic review and meta-analysis.
Onco Targets Ther 2019;12:6407-6438. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31496742
.
663. Poulou LS, Botsa E, Thanou I, et al. Percutaneous microwave
ablation vs radiofrequency ablation in the treatment of hepatocellular
carcinoma. World J Hepatol 2015;7:1054-1063. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26052394
.
664. Lewandowski RJ, Salem R. Microwave or radiofrequency ablation:
clinically equivalent? Lancet Gastroenterol Hepatol 2018;3:291-292.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29503248
.
665. Facciorusso A, Di Maso M, Muscatiello N. Microwave ablation
versus radiofrequency ablation for the treatment of hepatocellular
carcinoma: A systematic review and meta-analysis. Int J Hyperthermia
2016;32:339-344. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26794414
.
666. Huo YR, Eslick GD. Microwave Ablation Compared to
Radiofrequency Ablation for Hepatic Lesions: A Meta-Analysis. J Vasc
Interv Radiol 2015;26:1139-1146 e1132. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26027937
.
667. Vietti Violi N, Duran R, Guiu B, et al. Efficacy of microwave ablation
versus radiofrequency ablation for the treatment of hepatocellular
carcinoma in patients with chronic liver disease: a randomised controlled
phase 2 trial. Lancet Gastroenterol Hepatol 2018;3:317-325. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29503247
.
668. Mahnken AH, Konig AM, Figiel JH. Current Technique and
Application of Percutaneous Cryotherapy. Rofo 2018;190:836-846.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29665588
.
669. Shiina S, Sato K, Tateishi R, et al. Percutaneous Ablation for
Hepatocellular Carcinoma: Comparison of Various Ablation Techniques
and Surgery. Can J Gastroenterol Hepatol 2018;2018:4756147.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/29974040
.
670. Bala MM, Riemsma RP, Wolff R, et al. Cryotherapy for liver
metastases. Cochrane Database Syst Rev 2019;7:CD009058. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/31291464
.
671. Wu S, Hou J, Ding Y, et al. Cryoablation Versus Radiofrequency
Ablation for Hepatic Malignancies: A Systematic Review and Literature-
Based Analysis. Medicine (Baltimore) 2015;94:e2252. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26656371
.
672. Pearson AS, Izzo F, Fleming RY, et al. Intraoperative
radiofrequency ablation or cryoablation for hepatic malignancies. Am J
Surg 1999;178:592-599. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/10670879
.
673. Huang YZ, Zhou SC, Zhou H, Tong M. Radiofrequency ablation
versus cryosurgery ablation for hepatocellular carcinoma: a meta-
analysis. Hepatogastroenterology 2013;60:1131-1135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23321123
.
674. Eichler K, Zangos S, Gruber-Rouh T, et al. MR-guided laser-
induced thermotherapy (LITT) in patients with liver metastases of uveal
melanoma. J Eur Acad Dermatol Venereol 2014;28:1756-1760. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/24593299
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-97
675. Bale R, Schullian P, Schmuth M, et al. Stereotactic Radiofrequency
Ablation for Metastatic Melanoma to the Liver. Cardiovasc Intervent
Radiol 2016;39:1128-1135. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27055850
.
676. White ML, Atwell TD, Kurup AN, et al. Recurrence and Survival
Outcomes After Percutaneous Thermal Ablation of Oligometastatic
Melanoma. Mayo Clin Proc 2016;91:288-296. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26827235
.
677. Rozeman EA, Prevoo W, Meier MAJ, et al. Phase Ib/II trial testing
combined radiofrequency ablation and ipilimumab in uveal melanoma
(SECIRA-UM). Melanoma Res 2020. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31895753
.
678. Bedikian AY, Papadopoulos N, Plager C, et al. Phase II evaluation
of temozolomide in metastatic choroidal melanoma. Melanoma Res
2003;13:303-306. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12777987
.
679. Homsi J, Bedikian AY, Papadopoulos NE, et al. Phase 2 open-label
study of weekly docosahexaenoic acid-paclitaxel in patients with
metastatic uveal melanoma. Melanoma Res 2010;20:507-510. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/20881508
.
680. Bedikian AY, Plager C, Papadopoulos N, et al. Phase II evaluation
of paclitaxel by short intravenous infusion in metastatic melanoma.
Melanoma Res 2004;14:63-66. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15091196
.
681. Schmittel A, Schmidt-Hieber M, Martus P, et al. A randomized
phase II trial of gemcitabine plus treosulfan versus treosulfan alone in
patients with metastatic uveal melanoma. Ann Oncol 2006;17:1826-
1829. Available at: https://www.ncbi.nlm.nih.gov/pubmed/16971664
.
682. Schmidt-Hieber M, Schmittel A, Thiel E, Keilholz U. A phase II study
of bendamustine chemotherapy as second-line treatment in metastatic
uveal melanoma. Melanoma Res 2004;14:439-442. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15577312
.
683. Bedikian AY, Papadopoulos NE, Kim KB, et al. A pilot study with
vincristine sulfate liposome infusion in patients with metastatic
melanoma. Melanoma Res 2008;18:400-404. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/19011511
.
684. Carvajal RD, Piperno-Neumann S, Kapiteijn E, et al. Selumetinib in
Combination With Dacarbazine in Patients With Metastatic Uveal
Melanoma: A Phase III, Multicenter, Randomized Trial (SUMIT). J Clin
Oncol 2018;36:1232-1239. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29528792
.
685. Carvajal RD, Sosman JA, Quevedo JF, et al. Effect of selumetinib
vs chemotherapy on progression-free survival in uveal melanoma: a
randomized clinical trial. JAMA 2014;311:2397-2405. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24938562
.
686. Sacco JJ, Nathan PD, Danson S, et al. Sunitinib versus
dacarbazine as first-line treatment in patients with metastatic uveal
melanoma. J Clin Oncol 2013;31:9031-9031. Available at:
https://ascopubs.org/doi/abs/10.1200/jco.2013.31.15_suppl.9031
.
687. Luke JJ, Olson DJ, Allred JB, et al. Randomized Phase II Trial and
Tumor Mutational Spectrum Analysis from Cabozantinib versus
Chemotherapy in Metastatic Uveal Melanoma (Alliance A091201). Clin
Cancer Res 2019. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31558480
.
688. Kim KB, Bedikian AY, Camacho LH, et al. A phase II trial of arsenic
trioxide in patients with metastatic melanoma. Cancer 2005;104:1687-
1692. Available at: https://www.ncbi.nlm.nih.gov/pubmed/16130126
.
689. Hofmann UB, Kauczok-Vetter CS, Houben R, Becker JC.
Overexpression of the KIT/SCF in uveal melanoma does not translate
into clinical efficacy of imatinib mesylate. Clin Cancer Res 2009;15:324-
329. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19118061
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-98
690. Penel N, Delcambre C, Durando X, et al. O-Mel-Inib: a Cancero-
pole Nord-Ouest multicenter phase II trial of high-dose imatinib mesylate
in metastatic uveal melanoma. Invest New Drugs 2008;26:561-565.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/18551246
.
691. Mouriaux F, Servois V, Parienti JJ, et al. Sorafenib in metastatic
uveal melanoma: efficacy, toxicity and health-related quality of life in a
multicentre phase II study. Br J Cancer 2016;115:20-24. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27253171
.
692. Mahipal A, Tijani L, Chan K, et al. A pilot study of sunitinib malate in
patients with metastatic uveal melanoma. Melanoma Res 2012;22:440-
446. Available at: https://www.ncbi.nlm.nih.gov/pubmed/23114504
.
693. Shoushtari AN, Kudchadkar RR, Panageas K, et al. A randomized
phase 2 study of trametinib with or without GSK2141795 in patients with
advanced uveal melanoma. J Clin Oncol 2016;34:9511-9511. Available
at: http://ascopubs.org/doi/abs/10.1200/JCO.2016.34.15_suppl.9511
.
694. Falchook GS, Lewis KD, Infante JR, et al. Activity of the oral MEK
inhibitor trametinib in patients with advanced melanoma: a phase 1 dose-
escalation trial. Lancet Oncol 2012;13:782-789. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/22805292
.
695. Daud A, Kluger HM, Kurzrock R, et al. Phase II randomised
discontinuation trial of the MET/VEGF receptor inhibitor cabozantinib in
metastatic melanoma. Br J Cancer 2017;116:432-440. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28103611
.
696. Shah S, Luke JJ, Jacene HA, et al. Results from phase II trial of
HSP90 inhibitor, STA-9090 (ganetespib), in metastatic uveal melanoma.
Melanoma Res 2018;28:605-610. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30211813
.
697. Shoushtari AN, Ong LT, Schoder H, et al. A phase 2 trial of
everolimus and pasireotide long-acting release in patients with
metastatic uveal melanoma. Melanoma Res 2016;26:272-277. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/26795274
.
698. Tarhini AA, Frankel P, Margolin KA, et al. Aflibercept (VEGF Trap)
in inoperable stage III or stage iv melanoma of cutaneous or uveal origin.
Clin Cancer Res 2011;17:6574-6581. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21880788
.
699. Ellerhorst JA, Bedikian AY, Smith TM, et al. Phase II trial of 9-
nitrocamptothecin (RFS 2000) for patients with metastatic cutaneous or
uveal melanoma. Anticancer Drugs 2002;13:169-172. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/11901310
.
700. Sato T, Nathan PD, Hernandez-Aya LF, et al. Intra-patient
escalation dosing strategy with IMCgp100 results in mitigation of T-cell
based toxicity and preliminary efficacy in advanced uveal melanoma. J
Clin Oncol 2017;35:9531-9531. Available at:
https://ascopubs.org/doi/abs/10.1200/JCO.2017.35.15_suppl.9531
.
701. Chandran SS, Somerville RPT, Yang JC, et al. Treatment of
metastatic uveal melanoma with adoptive transfer of tumour-infiltrating
lymphocytes: a single-centre, two-stage, single-arm, phase 2 study.
Lancet Oncol 2017;18:792-802. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28395880
.
702. Bol KF, Mensink HW, Aarntzen EH, et al. Long overall survival after
dendritic cell vaccination in metastatic uveal melanoma patients. Am J
Ophthalmol 2014;158:939-947. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25038326
.
703. Borden EC, Jacobs B, Hollovary E, et al. Gene regulatory and
clinical effects of interferon beta in patients with metastatic melanoma: a
phase II trial. J Interferon Cytokine Res 2011;31:433-440. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21235385
.
704. Zimmer L, Vaubel J, Mohr P, et al. Phase II DeCOG-study of
ipilimumab in pretreated and treatment-naive patients with metastatic
uveal melanoma. PLoS One 2015;10:e0118564. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25761109
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-99
705. Joshua AM, Monzon JG, Mihalcioiu C, et al. A phase 2 study of
tremelimumab in patients with advanced uveal melanoma. Melanoma
Res 2015;25:342-347. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26050146
.
706. Piulats Rodriguez JM, Ochoa de Olza M, Codes M, et al. Phase II
study evaluating ipilimumab as a single agent in the first-line treatment of
adult patients (Pts) with metastatic uveal melanoma (MUM): The GEM-1
trial. J Clin Oncol 2014;32:9033-9033. Available at:
https://ascopubs.org/doi/abs/10.1200/jco.2014.32.15_suppl.9033
.
707. Johnson DB, Bao R, Ancell KK, et al. Response to Anti-PD-1 in
Uveal Melanoma Without High-Volume Liver Metastasis. J Natl Compr
Canc Netw 2019;17:114-117. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30787124
.
708. van der Kooij MK, Joosse A, Speetjens FM, et al. Anti-PD1
treatment in metastatic uveal melanoma in the Netherlands. Acta Oncol
2017;56:101-103. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27911126
.
709. Rossi E, Pagliara MM, Orteschi D, et al. Pembrolizumab as first-line
treatment for metastatic uveal melanoma. Cancer Immunol Immunother
2019;68:1179-1185. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/31175402
.
710. Schadendorf D, Ascierto PA, Haanen JBAG, et al. Efficacy and
safety of nivolumab (NIVO) in patients with advanced melanoma (MEL)
and poor prognostic factors who progressed on or after ipilimumab (IPI):
Results from a phase II study (CheckMate 172). J Clin Oncol
2017;35:9524-9524. Available at:
https://ascopubs.org/doi/abs/10.1200/JCO.2017.35.15_suppl.9524
.
711. Pelster M, Gruschkus SK, Bassett R, et al. Phase II study of
ipilimumab and nivolumab (ipi/nivo) in metastatic uveal melanoma (UM).
J Clin Oncol 2019;37:9522-9522. Available at:
https://ascopubs.org/doi/abs/10.1200/JCO.2019.37.15_suppl.9522
.
712. Piulats JM, Ochoa-de-Olza M, Lopez-Martin JA, et al. Phase II
study evaluating Ipilimumab monotherapy in the firstline treatment of
adult patients with metastatic uveal melanoma (MUM): The GEM1 trial
[abstract]. Pigment Cell Melanoma Res 2014;27:1209. Available at:
https://onlinelibrary.wiley.com/doi/10.1111/pcmr.12317
.
713. Piulats JM, De La Cruz-Merino L, Curiel Garcia MT, et al. Phase II
multicenter, single arm, open label study of nivolumab (NIVO) in
combination with ipilimumab (IPI) as first line in adult patients (pts) with
metastatic uveal melanoma (MUM): GEM1402 NCT02626962. J Clin
Oncol 2017;35:9533-9533. Available at:
http://ascopubs.org/doi/abs/10.1200/JCO.2017.35.15_suppl.9533
.
714. Schmittel A, Schuster R, Bechrakis NE, et al. A two-cohort phase II
clinical trial of gemcitabine plus treosulfan in patients with metastatic
uveal melanoma. Melanoma Res 2005;15:447-451. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16179873
.
715. Corrie PG, Shaw J, Spanswick VJ, et al. Phase I trial combining
gemcitabine and treosulfan in advanced cutaneous and uveal melanoma
patients. Br J Cancer 2005;92:1997-2003. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15886706
.
716. Keilholz U, Schuster R, Schmittel A, et al. A clinical phase I trial of
gemcitabine and treosulfan in uveal melanoma and other solid tumours.
Eur J Cancer 2004;40:2047-2052. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15341977
.
717. Terheyden P, Brocker EB, Becker JC. Clinical evaluation of in vitro
chemosensitivity testing: the example of uveal melanoma. J Cancer Res
Clin Oncol 2004;130:395-399. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15160290
.
718. Pfohler C, Cree IA, Ugurel S, et al. Treosulfan and gemcitabine in
metastatic uveal melanoma patients: results of a multicenter feasibility
study. Anticancer Drugs 2003;14:337-340. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12782938
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-100
719. Schmittel A, Scheulen ME, Bechrakis NE, et al. Phase II trial of
cisplatin, gemcitabine and treosulfan in patients with metastatic uveal
melanoma. Melanoma Res 2005;15:205-207. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/15917703
.
720. O'Neill PA, Butt M, Eswar CV, et al. A prospective single arm phase
II study of dacarbazine and treosulfan as first-line therapy in metastatic
uveal melanoma. Melanoma Res 2006;16:245-248. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/16718271
.
721. Schinzari G, Rossi E, Cassano A, et al. Cisplatin, dacarbazine and
vinblastine as first line chemotherapy for liver metastatic uveal
melanoma in the era of immunotherapy: a single institution phase II
study. Melanoma Res 2017;27:591-595. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29076951
.
722. Lee CK, Jung M, Choi HJ, et al. Results of a Phase II Study to
Evaluate the Efficacy of Docetaxel and Carboplatin in Metastatic
Malignant Melanoma Patients Who Failed First-Line Therapy Containing
Dacarbazine. Cancer Res Treat 2015;47:781-789. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/25687848
.
723. Bedikian AY, Legha SS, Eton O, et al. Phase II trial of tirapazamine
combined with cisplatin in chemotherapy of advanced malignant
melanoma. Ann Oncol 1997;8:363-367. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9209666
.
724. Kivela T, Suciu S, Hansson J, et al. Bleomycin, vincristine,
lomustine and dacarbazine (BOLD) in combination with recombinant
interferon alpha-2b for metastatic uveal melanoma. Eur J Cancer
2003;39:1115-1120. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12736111
.
725. Pyrhonen S, Hahka-Kemppinen M, Muhonen T, et al.
Chemoimmunotherapy with bleomycin, vincristine, lomustine,
dacarbazine (BOLD), and human leukocyte interferon for metastatic
uveal melanoma. Cancer 2002;95:2366-2372. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12436444
.
726. Becker JC, Terheyden P, Kampgen E, et al. Treatment of
disseminated ocular melanoma with sequential fotemustine, interferon
alpha, and interleukin 2. Br J Cancer 2002;87:840-845. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/12373596
.
727. Nathan FE, Berd D, Sato T, et al. BOLD+interferon in the treatment
of metastatic uveal melanoma: first report of active systemic therapy. J
Exp Clin Cancer Res 1997;16:201-208. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9261748
.
728. McWilliams RR, Allred JB, Slostad JA, et al. NCCTG N0879
(Alliance): A randomized phase 2 cooperative group trial of carboplatin,
paclitaxel, and bevacizumab +/- everolimus for metastatic melanoma.
Cancer 2018;124:537-545. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29044496
.
729. Guenterberg KD, Grignol VP, Relekar KV, et al. A pilot study of
bevacizumab and interferon-alpha2b in ocular melanoma. Am J Clin
Oncol 2011;34:87-91. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/20458209
.
730. Solti M, Berd D, Mastrangelo MJ, Sato T. A pilot study of low-dose
thalidomide and interferon alpha-2b in patients with metastatic
melanoma who failed prior treatment. Melanoma Res 2007;17:225-231.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/17625452
.
731. Bhatia S, Moon J, Margolin KA, et al. Phase II trial of sorafenib in
combination with carboplatin and paclitaxel in patients with metastatic
uveal melanoma: SWOG S0512. PLoS One 2012;7:e48787. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23226204
.
732. Piperno-Neumann S, Diallo A, Etienne-Grimaldi MC, et al. Phase II
Trial of Bevacizumab in Combination With Temozolomide as First-Line
Treatment in Patients With Metastatic Uveal Melanoma. Oncologist
2016;21:281-282. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26911405
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-101
733. Flaherty LE, Unger JM, Liu PY, et al. Metastatic melanoma from
intraocular primary tumors: the Southwest Oncology Group experience in
phase II advanced melanoma clinical trials. Am J Clin Oncol
1998;21:568-572. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/9856657
.
734. Heppt MV, Steeb T, Schlager JG, et al. Immune checkpoint
blockade for unresectable or metastatic uveal melanoma: A systematic
review. Cancer Treat Rev 2017;60:44-52. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28881222
.
735. Steeb T, Wessely A, Ruzicka T, et al. How to MEK the best of uveal
melanoma: A systematic review on the efficacy and safety of MEK
inhibitors in metastatic or unresectable uveal melanoma. Eur J Cancer
2018;103:41-51. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30205280
.
736. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with
ipilimumab in patients with metastatic melanoma. N Engl J Med
2010;363:711-723. Available at:
http://www.ncbi.nlm.nih.gov/pubmed/20525992
.
737. Larkin J, Minor D, D'Angelo S, et al. Overall Survival in Patients
With Advanced Melanoma Who Received Nivolumab Versus
Investigator's Choice Chemotherapy in CheckMate 037: A Randomized,
Controlled, Open-Label Phase III Trial. J Clin Oncol 2018;36:383-390.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/28671856
.
738. Ascierto PA, Long GV, Robert C, et al. Survival Outcomes in
Patients With Previously Untreated BRAF Wild-Type Advanced
Melanoma Treated With Nivolumab Therapy: Three-Year Follow-up of a
Randomized Phase 3 Trial. JAMA Oncol 2019;5:187-194. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/30422243
.
739. Hodi FS, Chiarion-Sileni V, Gonzalez R, et al. Nivolumab plus
ipilimumab or nivolumab alone versus ipilimumab alone in advanced
melanoma (CheckMate 067): 4-year outcomes of a multicentre,
randomised, phase 3 trial. Lancet Oncol 2018;19:1480-1492. Available
at: https://www.ncbi.nlm.nih.gov/pubmed/30361170
.
740. Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus
ipilimumab for advanced melanoma: final overall survival results of a
multicentre, randomised, open-label phase 3 study (KEYNOTE-006).
Lancet 2017;390:1853-1862. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28822576
.
741. Long GV, Flaherty KT, Stroyakovskiy D, et al. Dabrafenib plus
trametinib versus dabrafenib monotherapy in patients with metastatic
BRAF V600E/K-mutant melanoma: long-term survival and safety
analysis of a phase 3 study. Ann Oncol 2017;28:1631-1639. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/28475671
.
742. Robert C, Karaszewska B, Schachter J, et al. Improved overall
survival in melanoma with combined dabrafenib and trametinib. N Engl J
Med 2015;372:30-39. Available at:
http://www.ncbi.nlm.nih.gov/pubmed/25399551
.
743. Ascierto PA, McArthur GA, Dreno B, et al. Cobimetinib combined
with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM):
updated efficacy results from a randomised, double-blind, phase 3 trial.
Lancet Oncol 2016;17:1248-1260. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27480103
.
744. Dummer R, Ascierto PA, Gogas HJ, et al. Encorafenib plus
binimetinib versus vemurafenib or encorafenib in patients with BRAF-
mutant melanoma (COLUMBUS): a multicentre, open-label, randomised
phase 3 trial. Lancet Oncol 2018;19:603-615. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29573941
.
745. Zeldis JB, Heller C, Seidel G, et al. A randomized phase II trial
comparing two doses of lenalidomide for the treatment of stage IV ocular
melanoma. J Clin Oncol 2009;27:e20012-e20012. Available at:
https://ascopubs.org/doi/abs/10.1200/jco.2009.27.15_suppl.e20012
.
746. Atzpodien J, Terfloth K, Fluck M, Reitz M. Cisplatin, gemcitabine
and treosulfan is effective in chemotherapy-pretreated relapsed stage IV
uveal melanoma patients. Cancer Chemother Pharmacol 2008;62:685-
688. Available at: https://www.ncbi.nlm.nih.gov/pubmed/18084763
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-102
747. Nathan PD, Marshall E, Smith CT, et al. A Cancer Research UK
two-stage multicenter phase II study of imatinib in the treatment of
patients with c-kit positive metastatic uveal melanoma (ITEM). J Clin
Oncol 2012;30:8523-8523. Available at:
https://ascopubs.org/doi/abs/10.1200/jco.2012.30.15_suppl.8523
.
748. Kaempgen E, Schmid M, Erdmann M, et al. Predictable clinical
responses to sorafenib in stage IV uveal melanoma. J Clin Oncol
2012;30:e19032-e19032. Available at:
https://ascopubs.org/doi/abs/10.1200/jco.2012.30.15_suppl.e19032
.
749. Wallander ML, Layfield LJ, Emerson LL, et al. KIT mutations in
ocular melanoma: frequency and anatomic distribution. Mod Pathol
2011;24:1031-1035. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21478825
.
750. Dogrusoz M, Jager MJ. Genetic prognostication in uveal melanoma.
Acta Ophthalmol 2018;96:331-347. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/29105334
.
751. Atkinson TM, Hay JL, Shoushtari A, et al. Relationship between
physician-adjudicated adverse events and patient-reported health-related
quality of life in a phase II clinical trial (NCT01143402) of patients with
metastatic uveal melanoma. J Cancer Res Clin Oncol 2017;143:439-445.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/27921276
.
752. Maio M, Danielli R, Chiarion-Sileni V, et al. Efficacy and safety of
ipilimumab in patients with pre-treated, uveal melanoma. Ann Oncol
2013;24:2911-2915. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/24067719
.
753. Khattak MA, Fisher R, Hughes P, et al. Ipilimumab activity in
advanced uveal melanoma. Melanoma Res 2013;23:79-81. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23211837
.
754. Kelderman S, van der Kooij MK, van den Eertwegh AJ, et al.
Ipilimumab in pretreated metastastic uveal melanoma patients. Results
of the Dutch Working group on Immunotherapy of Oncology (WIN-O).
Acta Oncol 2013;52:1786-1788. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/23607756
.
755. Danielli R, Ridolfi R, Chiarion-Sileni V, et al. Ipilimumab in
pretreated patients with metastatic uveal melanoma: safety and clinical
efficacy. Cancer Immunol Immunother 2012;61:41-48. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/21833591
.
756. Ahmad SS, Qian W, Ellis S, et al. Ipilimumab in the real world: the
UK expanded access programme experience in previously treated
advanced melanoma patients. Melanoma Res 2015;25:432-442.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/26225580
.
757. Shaw H, Larkin J, Corrie P, et al. Ipilimumab for Advanced
Melanoma in an Expanded Access Programme (EAP): Ocular, Mucosal
and Acral Subtype UK Experience. Ann Oncol 2012;23:ix374. Available
at: http://www.sciencedirect.com/science/article/pii/S0923753420337042
.
758. Jung M, Lee J, Kim TM, et al. Ipilimumab Real-World Efficacy and
Safety in Korean Melanoma Patients from the Korean Named-Patient
Program Cohort. Cancer Res Treat 2017;49:44-53. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/27121719
.
759. Kottschade LA, McWilliams RR, Markovic SN, et al. The use of
pembrolizumab for the treatment of metastatic uveal melanoma.
Melanoma Res 2016;26:300-303. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/26848796
.
760. Middleton MR, McAlpine C, Woodcock VK, et al. Tebentafusp, A
TCR/Anti-CD3 Bispecific Fusion Protein Targeting gp100, Potently
Activated Antitumor Immune Responses in Patients with Metastatic
Melanoma. Clin Cancer Res 2020. Available at:
https://www.ncbi.nlm.nih.gov/pubmed/32816891
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.
Version 2.2021 © 2021 National Comprehensive Cancer Network
©
(NCCN
©
), All rights reserved. NCCN Guidelines
®
and this illustration may not be reproduced in any form without the express written permission of NCCN.
NCCN Guidelines Version 2.2021
Melanoma: Uveal
MS-103
761. Huguenin PU, Kieser S, Glanzmann C, et al. Radiotherapy for
metastatic carcinomas of the kidney or melanomas: an analysis using
palliative end points. Int J Radiat Oncol Biol Phys 1998;41:401-405.
Available at: https://www.ncbi.nlm.nih.gov/pubmed/9607358
.
Printed by on 7/4/2021 10:28:36 AM. For personal use only. Not approved for distribution. Copyright © 2021 National Comprehensive Cancer Network, Inc., All Rights Reserved.