Absolute Clinical Radiation Oncology Review (PDFDrive) PDF
Absolute Clinical Radiation Oncology Review (PDFDrive) PDF
Absolute Clinical Radiation Oncology Review (PDFDrive) PDF
Radiation Oncology
Review
Daniel M. Trifiletti
Nicholas G. Zaorsky
Editors
https://t.me/MBS_MedicalBooksStore
123
Absolute Clinical Radiation
Oncology Review
Daniel M. Trifiletti • Nicholas G. Zaorsky
Editors
Absolute Clinical
Radiation Oncology
Review
Editors
Daniel M. Trifiletti Nicholas G. Zaorsky
Department of Radiation Oncology Department of Radiation Oncology
Mayo Clinic Penn State Cancer Institute
Jacksonville, FL Hershey, PA
USA USA
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Contents
v
vi Contents
vii
Abbreviations
2D Two-dimensional
3D Three-dimensional
3D-CRT Three-dimensional conformal radiation therapy
5-FU 5-Fluorouracil
ABMT Autologous bone marrow transplant
APBI Accelerated partial breast irradiation
abnl Abnormal
ACTH Adrenocorticotropic hormone
ADH Antidiuretic hormone
adj Adjuvant
Adr Adriamycin
AFP Alpha-fetoprotein
AIDS Acquired immune deficiency syndrome
AJCC American Joint Committee on Cancer
aka Also known as
alk phos Alkaline phosphatase
Alt Alternated with
am Morning (ante meridian)
ANC Absolute neutrophil count (lab)
ant Anterior
anterolat Anterolateral
AP Anterior-posterior
APC Adenomatous polyposis coli (gene mutation)
appx Approximately
APR Abdominoperineal resection
ARUBA A Randomized Trial of Unruptured Brain Arteriovenous
Malformations
ASCUS Atypical squamous cells of undetermined significance
ASTRO American Society for Radiation Oncology
AUC Area under the curve
avg Average
BAT B-mode acquisition and targeting
b/c Because
b/t Between
bFFP Biochemical freedom from progression
b-HCG Beta-human chorionic gonadotropin
bid Twice daily
ix
x Abbreviations
bilat Bilateral
BM Bone marrow
BMI Body mass index
BMP Basic metabolic panel
BMT Bone marrow transplant
BTSG Brain Tumor Study Group
BWS Beckwith-Wiedemann Syndrome
Bx Biopsy/biopsies
C Cervical (spine level)
c/w Compared with
CA19-9 Cancer antigen 19-9
CA 125 Cancer antigen 125
CALGB Cancer and Leukemia Group B
C/A/P Chest/abdomen/pelvis
CBC Complete blood count (lab)
CCCG Colorectal Cancer Collaborative Group
cCR Clinical complete response
CD Cone-down
CD4 Cluster of differentiation 4 (for immune cells)
CEA Carcinoembryonic antigen
CESS Cooperative Ewing Sarcoma Study
CHART Continuous Hyperfractionated Accelerated Radiotherapy Trial
chemo Chemotherapy
CHF Congestive heart failure
CIN Cervical intraepithelial neoplasia
CIS Carcinoma in situ
cm Centimeter/centimeters
CMP Complete metabolic panel (lab)
c-myc Proto-oncogene, part of the Myc gene family
cN0 Clinically node-negative
CN Cranial nerve
CNS Central nervous system
Co-60 Cobalt-60
COG Children’s Oncology Group
contralat Contralateral
CPT Common procedural terminology
Cr Creatinine
CR Complete response
CRT Chemoradiation
CSF Cerebrospinal fluid
CSI Craniospinal irradiation
CSM Cancer-specific mortality
CSS Cause-specific survival
CT Computed tomography
cT Clinical T-stage
CTV Clinical target volume
Cx Cervical (spine level)
CXR Chest x-ray
Abbreviations xi
D/C Discontinue/discontinued
D&C Dilation and curettage
DCC Deleted in colorectal cancer (gene)
DDx Differential diagnosis
DFS Disease free survival
DI Diabetes insipidus
DLBCL Diffuse large-B cell lymphoma
DLCO Lung diffusion capacity testing
DM Distant metastasis
DMFS Distant metastasis free survival
DOI Depth of invasion
DRE Digital rectal examination
DSS Disease-specific survival
d/t Due to
DVH Dose volume histogram
DVT Deep venous thrombosis
Dx Diagnosis/diagnoses
Dz Disease/diseases
EB External beam
EBRT External beam radiation therapy
EBUS Endobronchial ultrasound
EBV Epstein-Barr virus
ECE Extracapsular extension
ECOG Eastern Cooperative Oncology Group
EFRT Extended field radiotherapy
EFS Event free survival
e.g. For example
EGFR Epidermal growth factor receptor
EM Electron microscopy
ENI Elective nodal irradiation
EORTC European Organisation for Research and Treatment of Cancer
Epo Erythropoietin
ESR Erythrocyte sedimentation rate (lab)
et al. And others
EUA Exam under anesthesia
EUS Endoscopic ultrasound
EWS Ewing sarcoma
exam Examination
f/b Followed by
FAP Familial adenomatous polyposis
FDA Food and Drug Administration
FDG Fluorine-18 2-fluoro-2-deoxy-D-glucose
FEV Forced expiratory volume
FFS Failure-free survival
FFTF Freedom from treatment failure
FIGO International Federation of Gynecology and Obstetrics
FH Favorable histology
FHIT Fragile histidine triad
xii Abbreviations
IFN Interferon
IgA Immunoglobulin A
IGF Insulin-like growth factor
IgG Immunoglobulin G
IGRT Image-guided radiation therapy
IJROBP International Journal of Radiation Oncology, Biology, and
Physics
IMA Inferior mesenteric artery
IMRT Intensity-modulated radiation therapy
inf Inferior
INR International normalized ratio
intraop Intraoperative
IORT Intraoperative radiation therapy
ipsi Ipsilateral
IQ Intelligence quotient
ITV Internal target volume
IVC Inferior vena cava
JAMA Journal of the American Medical Association
JCO Journal of Clinical Oncology
JCOG Japan Clinical Oncology Group
JCRT Joint Center for Radiation Therapy
JHH Johns Hopkins Hospital
JNCI Journal of the National Cancer Institute
JPA Juvenile pilocytic astrocytoma
KPS Karnofsky Performance Status
L Lumbar (spine level)
LA Lymphadenopathy
lab Laboratory/laboratory test
LAD Lymphadenopathy
LAMP Locally Advanced Multimodality Protocol
LAO Left anterior oblique
lat Lateral
LC Local control
LDH Lactate dehydrogenase
LDR Low dose rate
LE Lower extremity
LEEP Loop electrosurgical excision procedure
LF Local failure
LFT Liver function test
LGSIL Low-grade squamous intraepithelial lesion
LH Luteinizing hormone
LINAC Linear accelerator
LLL Left lower lobe
LML Left middle lobe
LN Lymph node
LND Lymph node dissection
LOH Loss of heterozygosity
LP Lumbar puncture
xiv Abbreviations
PT Prothrombin time
pT Pathologic tumor stage
PTV Planning target volume
PUVA Psoralen and long-wave ultraviolet radiation
q Every
qd Daily
QOL Quality of life
QUANTEC Quantitative analysis of normal tissue effect in the clinic
R1 Microscopically positive margin
R2 Macroscopically positive margin
RAO Right anterior oblique
RASSFIA Ras association (RalGDS/AF-6) domain family member 1A
RB Retinoblastoma
RBE Relative biologic effectiveness
RCC Renal cell carcinoma
RCT Randomized controlled trial
rcv Receive/received
RFS Relapse free survival
RLL Right lower lobe
RML Right middle lobe
RMS Rhabdomyosarcoma
r/o Rule out
ROM Range of motion
RPO Right posterior oblique
RR Relative risk
RT Radiation or radiation therapy
RTOG Radiation Therapy Oncology Group
RUL Right upper lobe
RUQ Right upper quadrant
Rx Prescription/prescriptions
S Sacral (spine level)
SBO Small bowel obstruction
SC Spinal cord
SCC (or SCCa) Squamous cell carcinoma
SCV Supraclavicular
Sg Surgery
SEER Surveillance Epidemiology and End Results (data)
SFOP French Society of Pediatric Oncology
Sg Surgery
SIADH Syndrome of inappropriate secretion of antidiuretic hormone
SIL Squamous intraepithelial lesion
SQ Subcutaneous
s/p Status post
SPECT Single photon emission computed tomography
SRS Stereotactic radiosurgery
SS Statistically significant
SSD Source to skin distance
ST Soft tissue (as in sarcoma)
Abbreviations xvii
Symbols
Abstract
This chapter discusses the general manage-
ment and thought process used by radiation
oncologists. Several broad and basic principles
of radiation oncology are discussed.
N. G. Zaorsky (*)
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
D. M. Trifiletti
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
D. W. Golden
University of Chicago, Chicago, IL, USA
e-mail: dgolden@radonc.uchicago.edu
WHO IV (GBM) Standard (Stupp): MRI in 48h of surg. CTV46 = Post-op T2 FLAIR + cavity + 2 cm + 3 mm w daily 46 Gy to FLAIR, 60 Gy to +TMZ 75 mg/m2 QD, then 1m GBM
and WHO III (AA, AO, AOA) < ~65–70 yo, KPS > Send tissue for MGMT, CTV60 = post-op cavity + CBCT enhancement break, then 150 mg x 5d q 28d x 6c. RPA III: 22 m
1p19q non-codel; Unidel; AA ~60–70 IDH1/2, TERT promoter, residual enhancement + 2 cm (cropped to 5 mm at Consider TTF RPA IV: 15 m
Rim-enhancing (signifying EGFR natural barriers) No Avastin upfront RPA V: 10 m
central necrosis), irregular.
Pathology: pseudo- >65–70 yo and good Decadron + PPI CTV = T1post enhancement + 2 cm + 3 mm w daily 40 Gy / 10 @ 4 + TMZ → TMZ (Perry) No TMZ in Canada/Roa, Perry: MST 9.3 vs 7.6 m
palisading necrosis. High KPS Keppra 500 mg BID if sz CBCT 40.05 Gy / 15 @2.7 Gy (Roa 1; also WHO yes TMZ in Nordic/Perry
grade = Mitoses, Endothelial III KPS≤60), 75% of dose
prolif, Atypia, Necrosis. 25 Gy / 5 @ 5 Gy (Roa 2: 40/15=25/5)
IDH 1 WT = molecular GBM 34 Gy / 10 @ 3.4 Gy (Nordic: 34/10 better
than 60/30)
IDH1 = 2ndary GBM (LGG
=>HGG) KPS ≤ 60 TMZ alone (Nordic) for poor KPS
Recurrence GTV + 3 mm w daily 35 Gy / 10 Bev alone
CBCT
WHO III (AA, AO, AOA); 1p19q co-del (all are MRI in 48h of surg. CTV45 = Post-op T2 FLAIR + cavity + 2 cm + 3 mm w daily 45 Gy to FLAIR, and 59.4 Gy to Adjuvant PCV q6w x 6c 94–02 GIII AO, AOA:
IDH1mut); Send tissue for 1p/19q and (cropped to 5 mm at natural barriers) CBCT conedown / 1.8 Gy RT vs RT, adj PCV
others per WHO IV IDH1 for oligo (all 1p19q CTV59.4 = If enhancing: post-op cavity + 1p/19q co-del (all IDH1 mut): 7yr vs. 14.9yr
standard codel are also IDH1 mut) residual enhancement + 2 cm (cropped to 1p/19q intact, IDH1 mut: 3.3 vs. 5.5
5 mm at natural barriers). If non- Intact/WT: 1.3 vs. 1.0
enhancing: T2-FLAIR +5mm
LGG (diffuse astro: oligo, New hi risk: > 40 yo or options: CTV = T2 FLAIR + tumor bed + 1 cm + 3 mm w daily 50.4–54 Gy High Risk options: Low risk: OS5 = 90%; PFS5 = 50%
astro, mixed oligoastro; STR Old hi risk (also): RT + adjuvant PCV CBCT RT + adjuvant procarbazine High risk: OS5 = 70%; PFS5 = 50%
pilomyxoid; pleomorphic size 6+ cm, astrocytoma, RT + concurrent TMZ (prefer POD8–21, CCNU d1,
neuro deficit preop, for MGMTm) vincristine (PCV)
General Principles of Radiation Oncology
xanthoastro) G1 subtypes:
cross midline, 1p19q
JPA, pleomorphic non-co-del, IDH1/2 Decadron + PPI RT + concurrent TMZ
xanthoastro, SEGC non-mut Keppra 500 mg BID if sz
astrocytoma, Low risk: <40 yo Options: Obs vs RT alone if Low risk option:
subependymoma, AND GTR sx or difficult to salvage vs. TMZ alone if 1p19q co
gangliogliomas TMZ alone if 1p19q codel deleted
Ependymoma, brain G1 = subependymomas MRI in 48h of surg. CTV = initial tumor vol, tumor bed + 3 mm w daily G2: 54 Gy Chemo no proven benefit EFS5 GTR: 80%
and myxopapillary RT in 8w. + 1 cm CBCT G3: 59.4 Gy. If MRI spine/LP+, CSI EFS5 STR: 40%
G2 = classic to 36 Gy. Boost spine GTVs to 45 Gy
Ependymoma, spine ependymomas R0 = observe (only G1/2) + 3 mm w daily Limited field = 50.4 Gy. If MRI spine/LP+,
G3 = anaplastic R+ = limited field RT CBCT CSI to 36 Gy. Boost spine GTVs to
ependymomas Spine mets = CSI + boost 45 Gy
Ependymoblastoma treat like STPNET/high CSI 36 Gy. Boost to 55.8 Gy
risk medullo
Meningioma G1 Simpson1–3 (R0): obs Do not cover dural tail!!! + 3 mm w daily RTOG 0539: G1: 90% LC in def EBRT, R0, R1+EBRT
G1 Simpson4–5 (R+): obs G1: CTV54 = GTV + bed + 1 cm CBCT PTV54 to tumor/bed +1–2 cm G2: MST 12y. LC 60% if R0, 70% LC surg
vs RT hi risk vol G2/3: PTV60 to G2/3 tumor bed + 1 cm + EBRT
Recur: surg vs RT CTV54 = GTV + bed + 2 cm Consider SRS 13 Gy (up to 17 Gy if G3), G3: MST 3y. OS5 50%.
G2-3 (<10%): EBRT CTV60 = GTV + bed + 1 cm but not 1st choise. Optic n sheath: 45 Gy
Pituitary adenoma: RT if: Inoperable; TSH Endo panel: TSH, FSH, LH, CTV= GTV + bed + 2 mm + 3 mm w daily Functioning: 54 Gy. Or SRS 20 Gy GH or TSH: Somatostatin If no hormone normalization after surg, RT,
Macro: ³ 1 cm, micro < 1 cm secreting (always post- GH/IGF-1, cortisol, ACTH, CBCT Non-functioning 50.4 Gy. Or SRS ACTH: Ketoconazole meds,:
PRL (30%), GH (25%), op); R+ with persistent PRL. Dex text. 18 Gy Cushing’s => adrenalectomy
ACTH (15%). PRL: hypersecretion or TSH => thyroidectomy
amenorrhea, galactorrhea.
Cushing (ACTH): obesity, residual near critical
HTN, DM, hirsutism, skin, structures;
osteoporosis. Acromegaly Recur after surg
(GH): bones, HA, cardiac dz
AVM SRS best if < 3 cm, + 1 mm SRS 16–20 Gy None SRS reduces bleed risk 50% in latency period,
deep, defined focus 88% after obliteration. 2y obliteration = 90%
Acoustic neuroma No mass effect. If mass Audiometry to check SRS: if < 3 cm. GTV = PTV. SRS = none SRS = 12.5 Gy No
effect, needs surg serviceable hearing FSRT: if > 3 cm. PTV = GTV + 3 mm. FSRT = +3 mm w FSRT = 50.4 Gy in 1.8 Gy or 25
daily CBCRT Gy / 5
PCNSL Vz exam. Slip lamp exam If no ocular involvement, iso at bony Chemo. If CR, WBRT to 23.4 Gy. If R-MPV x 7c + IT MTX first 80% CR to chemo
(20% ocular involvement). canthi and ½ beam block. Cover post PR, WBRT to 30, boost GTV to 45 (per RTOG 93–10) OS5: 80% for pts who received WBRT
CBC, CMP, HIV, EBV, 1/3 orbit if slit lamp neg. Cover brain Gy If orbit involved, and if CR, 23.4 PCNSL = Ocular involvement in 20%
testicular exam, MRI spine. and cribriform, down to C2/3. Gy to orbit; if PR 36 Gy. Ocular lymphoma = 80% risk of PCNSL
Bx first. No steroids bc If no chemo: WBRT 36 Gy, boost GTV to later
40% response. 45 Gy. Avoid RT if > 60yo
Primary ocular DLBCL 36 Gy / 1.8 Gy fx. Half beam block
post at post optic canal (tx CN II).
If only 1 eye, tx ipsi globe, CN II.
Trigeminal neuralgia MRI thin slices Single 4-mm isocenter; target is 3–6 SRS 80 Gy to 100 % IDL. Median time to pain relief: 1–2m
mm anterior from junction of the 60% pain free, 20% have decrease, 15%
trigeminal nerve and pons no change. <10% facial numbness
Brian mets MRI w contrast. Always do Intact = GTV + 1 mm RTOG 90–05. SRS to 80% IDL on
workup for primary: breast, Post-op = cavity + 2 mm linac. 0–2cm: 24 Gy, but use 22 Gy,
lung, RCC most common >2–3 cm: 18 Gy, >3–4 cm: 15 Gy
3
4
Disease Association Workup Treatment
HL CD15/30+, I-II F MC = EBV CBC, CMP, LDH, HIV, pregnancy ABVD x 2, restage w/ PET , Deauville 1–3, ISRT 20 Gy, Deauville 4, ISRT 30 Gy
LD = HIV, old test, EBV, albumin, ESR, UK RAPID: ABVD x3, interim PET. D1–2: observe or ABVDx1; D3–4 ABVDx1, then ISRT 30 Gy.
albumin, echo, excisional bx,
CSF if HIV+/testicular
HL CD15/30+, I-II UF + BM bx ABVD x 4, restage w/ PET,
Deauville 1–3 = ISRT 30 Gy
Deauville 4 , ABVD x 2, restage w/ PET
Deauville 1–3, ISRT 30 Gy
Persistent Deauville 4 or 5 needs bx, if (+) it is refractory
HL III-IV (MASH ALL): Male, Age ≥45, Stage IV, + BM bx ABVD x 2, restage w/ PET
Hgb <10.5, Alb <4, Leukocytosis >15k, Deauville 1–3 = ABVD x4, then observe
lymphocytopenia <0.6k/mL (or <8%)] Deauville 4 = ABVD x 4, restage w/ PET Deauville 1–3 ISRT to bulky sites to 36 Gy
NLPHL CD20/45+, I-II ISRT 30 Gy (larger margin than HL), then PET. CR = observe. PR = R-CHOP or ABVD.
NHL relapse/refractory RT after salvage chemo in pero-txp prior. Pre transplant 24–30 Gy / 20 fx BID 6 hours apart. Post txp 30 Gy in
17–20 Gy. If non-txp candidate, then 30 Gy for CR.
NHL, high grade: t(8:14): Burkitt’s + beta2 micro, + LDH, + da-R-EPOCH or R-CHOP x 6c, then PET-CT, then 30 Gy ISRT for PR
• Burkitt, lymphoblastic lymphoma HBV, + EGD, + CSF, + BM bx
NHL, G2, Stage I–II: t(11:14): BCL-1, mantle Above, but no CSF I-II (non-X, i.e. < 7.5 cm): For IPI 0-1, RCHOP x 3c. Then PET. Then ISRT 30–36 Gy. Boost FDG-avid residual dz to 40–46 Gy.
• follicular (G3B), mantle cell, DLBCL, T/NK cell, cell lymphoma + c-scope for mantle For IPI 2–4 or X, RCHOP x 6c. Then PET. Then +/–ISRT 30–36 Gy.
peripheral T cell, anaplastic large cell “Double hit”: bcl-2, c-myc If unable to tolerate chemo, then 36–45 Gy ISRT,boost to 40–46 Gy.
IPI: APLES: Age > 60, Performance >2, LDH > “Triple hit”: above + bcl-6 If high IPI, clinical trial for high dose chemo with autologous stem cell rescue. ISRT 36 Gy only to initial bulk dz.
ULN, 2+ E sites, Stage III/IV
NHL, intermediate grade, III–IV III-IV: If high IPI, clinical trial for high dose chemo with autologous stem cell rescue.
R-CHOP x 4, restage w/ PET. If CR, R-CHOP x 2, observe. Consider ISRT only to initial bulk (36 Gy), per German group. If PR,
R-CHOP x 2, restage w/ PET. If PET negative: ISRT only to initial bulk (36 Gy)
If PET positive: refractory, go to transplant.
• DLBCL, gastric R-CHOP x 3, ISRT 30 Gy
• DLBCL, testicular Orchiectomy, R-CHOP x 6 w/ IT-MTX, scrotal RT to 30 Gy. Electrons 9–12 MeV. No bolus.
• DLBCL, bone R-CHOP x3c, ISRT 30 Gy if CR, 40 if PR
• DLBCL, breast R-CHOP x 6c + RT 36 Gy to whole breast
Primary mediastinal B cell lymphoma (PMBCL) 35yo woman DA-EPOCH-R x 6c. NO RT unless persistent focal dz, then 30 Gy.
Mediastinal gray zone lymphoma (GZL) 35yo man DA-EPOCH-R or R-EPOCH x 6c. Always consolidate 30 Gy + 10 Gy boost for PR.
NHL, low grade / indolent: t(14:18), BCL 2, follicular Stage I–II, G1, no X: ISRT, 24–30 Gy / 2 Gy fx
• FL G1–2, MZL, MALT, SLL/CLL, MF lymphoma Stage I–II, X, or unfav molecular: Chemo w R-bendamustine, then ISRT, 24–30 Gy / 2 Gy fx
FLIPI for FL (“NoooooLASH”): Sites: ≥5, LDH > del13, t(14,19), tri12, Stage I–II G1, non-X and non-contiguous: chemo alone
ULN, Age ≥60, Stage III/IV, Hgb < 12. FLIPI-2: SLL, CLL Stage III–IV: no cure. Palliate PRN. 2 Gy x 2
B2 micro, BM, Hgb < 12, LN > 6 cm, age ≥ 60
• MALT, gastric t(11:18) usually abx EGD, urea breath test, Bismuth, lansoprazole, tetracycline, metronidazole. Wait 1 year. 30 Gy / 20 (@1.5) ISRT if refractory.
refractory bacteria test Tx planning: NPO, 4D, 100 cc PO contrast.
• MALT, orbital Chlamydia psittaci bacteria test Doxycycline. RT if refractory, 24 Gy / 12 fx. Treat whole orbit.
• MALT, salivary Sjogren Abx. Surg. If R0, observe. Else, RT 24 Gy / 12 fx.
• MALT, small bowel Campylobacter jejuni bacteria test Abx. Surg. If R0, observe. Else, RT 24 Gy / 12 fx.
• MALT, testis RT 24 Gy / 12 fx to contralattestis.
• MALT, thyroid Hashimoto Surg. If R0, observe. RT 24 Gy / 12 fx.
• MALT, spleen HCV Surg. If R0, observe. RT 24 Gy / 12 fx.
• MALT, breast RT 24 Gy / 12 fx. WBI.
• MALT, lung Surg. If R0, observe. RT 24 Gy / 12 fx.
Advanced stage: chemo + ISRT
• MALT, skin (Primary cutaneous MZL) Borrelia burgdorferi Surg. If R0, observe. RT 24 Gy / 12 fx. CTV = GTV + 1 cm. Palliate 2 Gy x 2.
Plasmacytoma
• SEP Lesion visible, CRP, LDH, ALP, albumin, B2 PTV1 to 50.4 Gy, ENI to 40 Gy.
plasmacytoma by bx micro, Ca, SPEP/UPEP, serum
• SBP (<5% plasma cells), no Bone with 2–3cm margin. Spine: whole VB
free light chain. Bx. No Tc99 BS.
CRAB Standard tx is RT, 40–50 Gy
MM BM bx > 10% plasma Bortezomib and dexamethasone x 2 c. Palliative 30 Gy / 10 fx
cells, M-spike on SPEP
or UPEP, CRAB
MF T1: PUVA, steroids, topical chemo, local electrons 24 Gy / 2 Gy
T2–4: TSEBT 24 Gy / 12 fx, 4 days per week, 3 positions QOD.
Nasal NK/T cell NHL EBV Concurrent Chemo-RT (54 Gy) to involved dz w/ concurrent cisplatinum, 3c DeVIC
Chloroma / myeloid sarcoma 24 Gy / 12 fx
N. G. Zaorsky et al.
Site Work-Up Scenario Treatment CTV PTV Dose Chemo Follow-Up Outcomes
Esophageal - H/P: GERD, alcohol, T2–4 or N+ Neoadjuvant CRT (followed ITV + 4cm sup/inf and 1cm CTV + 0.5cm 50.4Gy [can also do 45Gy Neoadjuvant: Carbo H/P Q 3–6mos for yrs 5-Yr OS:
smoking, dysphagia, by restaging PET/CT +/– radial + 5.4Gy CD if preop – CD AUC 2/ Taxol 50mg/m2 1–2, then Q6–12mos for Stage I: > 90%
weight loss EGD w/ Bx in 4 weeks) volume = GTV + 1cm per weekly yrs 3–5, then annually Stage II: 40%
- EGD + Bx surgery ENI: SCLV if proximal 1/3; celiac RTOG 1010] Stage III: 10%
-CT C/A/P +C if distal 1/3; paraesophageal for Definitive: cisplatin CT C/A/P Q4–6mos yr 1, Stage IV: < 5%
-EUS if M0 Or everyone 75mg/m2 + 5FU then decr freq
-PET/CT 1000mg/m2 on weeks pCR=30%
-Smoking Cessation Definitive CRT 1, 5, 8, 11 EGD Q 3–6mos for yrs R0=90%
-PEG/PEJ 1–2, then Q6mos yr 3
-Bronch if above carina
(25cm)
Gastric -H/P: satiety, hematemesis, High risk T2 Surgery Adjuvant CRT Gastric Bed CTV = 0.5cm 45Gy in 25 fractions 1c of capecitabine H/P Q 3–6mos for yrs 5-yr OS:
detailed nodal exam [G3, LVSI, PNI, Anastomosis (1000mg/m2 BID)D1– 1–2, then Q6–12mos for Stage I: 65%
(cervical axillary, no D2 LNs: SMA, perigastric, celiac for 5.4Gy boost if +SM 14 followed by yrs 3–5, then annually Stage II: 40%
periumbilical, SCLV), dissection] all; add 825mg/m2 BID Stage III: 15%
ascites, HSM paraesophageal + splenic If 9Gy boost if GRD capecitabine Monitor for B12 and iron Stage IV: 5%
-EGD + Bx T3–4 or N+ proximal/GEJ; add concurrent with RT deficiency
-CT C/A/P +C pancreaticoduodenal and followed by 2c of
-PET/CT if >T2,N+,M0 splenic if middle; add capecitabine Labs, imaging and EGD
-EUS if M0 pancreaticoduodenal + (1000mg/m2 BID)D1 as clinically indicated
General Principles of Radiation Oncology
Pancreas -H/P: painless jaundice, abdo Any If resectable: resect Resected: CTV + 0.5cm 50.4Gy in 28Fx Capecitabine H/P Q3–6mos for 2 yrs, Resectable: MS 2 yrs ,
exam, weight loss chemo (4c Gem/abraxane or CTV: [(proximal 1.5cm of CA, 825mg/m2 BID on then Q6–12mos w/ CA 3-yr OS=30%, LC
-Pancreatic protocol CT FOLFOX) CRT prox 3cm of SMA, PV, PJ, preop radiation days 19–9 and CT (cat 2B) 70–80%
-EUS + FNA Bx tumor volume) + 1cm] + [Aorta
-ERCP w/ stent if obstructed If unresectable: 4c chemo + (3cm to the R + 1cm to the L + Pancreatic enzyme Unresectable: MS
-CT chest CRT 2cm anteriorly + 0.2cm replacement 10mos, 2-yr
posteriorly)] OS 10–20%
Rectal -H/P: anemia, prior RT, IBD, [TAE alone if: Neoadjuvant CRT Surgery Mesorectum up to CTV + 0.7cm 45Gy to whole pelvis f/b Concurrent H/P Q3mos for 2yrs w/ 50% LRF reduction w/
DRE, pelvic exam in women, <8cm from adjuvant CT rectosigmoid or 2cm superior 5.4Gy boost to GTV + Capecitabine CEA if elevated at dx, RT
FHx verge, <30% to gross dz (whichever is more Fields: 2cm 825mg/m2 BID on then Q6 mos for yrs 3–5 LRR < 10% at 10yrs
-CEA circum, <3cm, proximal) and 2cm distal to PA: L5/S1 radiation days f/b 4 with naCRT; 30% DMs
-colonoscopy, proctoscopy G1, margin gross dz 3cm below months of CAPOX or Colonoscopy in 1 yr then
-EUS or MRI >3mm, no tumor + 2cm on FOLFOX [total of 6 Q5yrs 5-yr OS:
-CT C/A/P +C LVSI] If cT1/2 taken for surgery Presacral and internal iliac LNs pelvic brim months of chemo] Stage I: 90%
and found to be T3/4 or N+ laterally CT C/A/P annually for T3 or N+: 75%
T3–4 or N+ or +SM, give postop CRT If T4 : ext iliac Lats: behind 5yrs T4N+: 50%
CT PS (in front if M1, resectable liver:
If anal canal involved:+/– T4) behind 30–40%
inguinals sacrum
Anal -H/P: sphincter tone, HIV, IBD, Any Definitive CRT CTV_A: [GTV + anal canal] + CTV + 1cm T2N0: 50.4 Gy in 28Fx to Concurrent MMC H/P in 8–12 weeks. If CR, 5-yr OS:
prior RT, pelvic exam with pap 2–2.5cm CTV_A and 42Gy in 28Fx 10mg/m2 and 5-FU DRE and inguinal nodal Stage I: 85%
and HPV testing in women to CTV_C 1000mg/m2 exam Q3–6mos for 5 yrs; Stage II: 75%
-Labs: HIV CTV_B: Involved LN(s) + 1cm on D1 and D29 Anoscopy Q6–12mos for Stage III: 50%
-Anoscopy + Biopsy T3/4N0: 54Gy in 30Fx to 3 yrs, CT C/A/P annually Stage IV: 5%
-CT C/A/P +C CTV_C: Elective LNs (internal CTV_A and 45GY in 30Fx for 3 yrs
-PET/CT iliac, external iliac, inguinal) + to CTV_C
1cm If persistent disease at
N+: 54Gy in 30Fx to CTV_ 8–12 weeks, re-evaluate
A., 54Gy in 30Fx to at 4 weeks. If persistent
CTV_B if LN > 3cm OR disease, restage.
50.4Gy in 30Fx to CTV_B
if LN < 3cm; 45Gy in 30Fx If progressive dz at 8–12
to CTV_C weeks, re-stage.
5
6
Prostate PSA, MRI, Biopsy VLR, LR AS PSA q 6m, DRE/bx no more than q No
12m
After 12-month bx, alternate
mpMRI w/ bx. Indications to tx: in #
cores, % involvement, ≥ G4;
PSA DT < 3 years
LR (T1-T2a, GS6, PSA <10) EBRT Prostate + prox SVs + 5 mm 78 Gy No
+ BS if T2 and PSA >10 IR (T2b-c, GS7, PSA 10–20) EBRT Prostate + prox SVs + 5 mm PTV1 = 80 Gy Consider ADT 6m in unfav IR
Distal SVs PTV2 = 46 Gy
+ BS HR (GS 8+, T3+, PSA >20) EBRT Prostate + prox SVs + 5 mm PTV1 = 80 Gy ADT x 2–3y
+ T, LFTs, CBC, CMP, Dexa, + ADT Distal SVs + pelvic LNs PTV2 = 46 Gy Lupron (7.5 mg = 1 month; 22.5 mg = 3 months) = GnRH
Ca, Vit D agonist (Degarelix = GnRH antagonist). Menopausal sx.
Bicalutamide (50 mg/day) = anti-androgen. Check LFTs
monthly. Diarrhea.
> 60 cc, < 20 cc (if too large give LDR-BT Prostate 0 mm I125, 145 Gy mono 110/50.4 Gy boost
ADT to shrink by 1/3) Pd103, 110 Gy mono, 100/50.4 Gy
TURP defect boost
Large median lobe
Pubic arch interference HDR-BT Prostate 0 mm 13.5 x 2, one week apart
IPSS > 15 (> 20 absolute) 15 Gy + 50.4 Gy
Anesthesia/sedation risk
IBD/prior RT
MRI Post RP 1 cm below UVA, lat to + 5 mm 68 Gy. 74 Gy if GTV aRT, N0 = RT alone (shared decision making about ADT)
BS if PSA > 10 Adjuvant: T3, R+ obturator internus, post to aRT, N+ = RT + 2y of ADT (Messing was life)
Salvage: Persistent or rectum, ant entire bladder sRT, N0 = RT + 6 months ADT if pre-RT PSA <0.7 ng/mL;
rising PSA post-RP below pubic symphysis, and else, 2y
1 m of it superior to it. sRT, N+ = RT + 2y of ADT
Pelvic LNs if pLN+
IIA: T1–4 N1 (all ≤ 2cm (no 20 Gy DL + 10 Gy CD From T11/T12, contour PTV = CTV + 0.7cm to
more than 5 total pN+) S0-2 aorta and IVC down to the block edge
ipsilateral iliac arteries and
IIB: T1–4 N2 (all 2 -≤ 5cm, or 20 Gy DL + 16 Gy CD veins stopping at the top of Block edge CD = LN + 2 cm
more than 5 pN+, or ECE) the acetabulum
S0-2
III BEP 3c
N. G. Zaorsky et al.
General Principles of Radiation Oncology 7
Radiobiology
he “4 Rs” of Radiobiology
T
–– Redistribution
–– Repopulation
–– Repair
–– Reoxygenation
–– Lethal damage = DNA double-strand breaks
–– 1 Gy of radiation = ~20–40 DSBs
–– Majority of radiation-induced cell
death = mitotic catastrophe
Ipilimumab
Tremelimumab
Atezolizumab Depression
durvalumab
avelumab
Symptoms
MHC B7 B7 PD-L1
Five symptoms × 2 weeks:
1 2 3 4
GI – diarrhea <4 stools over 4–6 stools/d over baseline, 7+ stools/d over Acute or subacute
baseline, mild increase mod increase ostomy baseline, obstruction, fistula
ostomy output output, incontinence; or perforation’ GIB
antidiarrheal med hospitalized, severe requiring
increase in ostomy transfusion
output,
parenteral support
GI – constipation Occasional sx, Persistent sx with regular Obstipation with Urgent intervention
occasional stool use of laxatives or enema manual evacuation indicated
softeners, laxatives, indicated
enema
GI – LGIB Mild, no intervention Moderate, medical Transfusion, Urgent care needed
intervention, or minor radiologic,
cauterization endoscopic, or
elective operation
indicated
GI – fistula Altered GI function TPN or hospitalized, Life threatening
elective operation
GU renal (units eGFR < LLN – 60 59–30 29–15 <15
for CrCl are ml/ or proteinuria 2+
min/1.73 m2)
GU sexual Meds or penile pump Penile prosthesis
GU frequency Present Meds indicated N/A N/A
GU ejaculation Diminished Anejaculation or
retrograde
GU sperm N/A N/A Azoospermia N/A
(absence of sperm in
ejaculate)
GYN – Menopause: ovarian
menopause failure prior to 40 y/o
GYN – Mild pain, relieved Moderate pain, partially Severe discomfort or
dyspareunia with lubricants or relieved with lubricants or pain, unrelieved with
estrogen estrogen lubricants or estrogen
Weight Loss 5–10% Loss 10–20%, nutritional Loss 20 + %, TF or N/A
support indicated TPN needed
WBC < LLN – 1500/mm3 <1500–1000/mm3 <1000–500/mm3 <500/mm3
Limb length < 2 cm discrepancy 2–5 cm discrepancy, shoe >5 cm discrepancy,
lift indicated operation indicated
Lethargy Reduced alertness and Moderate, limited N/A N/A
awareness instrumental ADLs
Somnolence Mild drowsiness or Moderate sedation, Obtundation or stupor
sleepiness limiting instrumental
ADLs
Hypohidrosis, N/A Symptomatic, limiting Increase in body Heat stroke
body temp instrumental ADL temp
16 N. G. Zaorsky et al.
Dose Constraints
Daniel M. Trifiletti
Mayo Clinic, Jacksonville, FL, USA
Nicholas G. Zaorsky
Penn State Cancer Institute, Hershey, PA, USA
Daniel W. Golden
University of Chicago, Chicago, IL, USA
General Principles of Radiation Oncology 17
Dose
Site Organ at risk Dose constraint 1 Dose constraint 2 constraint 3 Notes
Pediatrics Long bone V40 < 64% Mean < 37 Gy Max 59 Gy
Pediatrics Kidney V10 < 80%
Pediatrics Flat bone Minimize V35
Pediatrics VB V18 all or none 18 Gy over entirety if
receiving any RT; this
will prevent scoliosis
Pediatrics Skin/lymphedema Keep longitudinal
2–3 cm strip of
extremity to <15 Gy
Pediatrics Soft tissue (to V60
prevent
RT-induced
sarcoma)
CNS Cauda equina 0.03 cc < 16 Gy 5 cc < 14 Gy
CNS (SRS) Brain stem 0.5 cc < 10 Gy
CNS (SRS) Optic nerves/ 0.2 cc < 8 Gy
Chiasm
CNS (SRS) Spinal cord 0.35 cc < 10 Gy Max 18 Gy / 3 fx Kilpatrick (2010) [1]
(RTOG 0236,
0618)
H&N Mandible 0.1 cc < 70 Gy
H&N Temporal Lobe 0.03 cc < 68 Gy 1 cc < 58 Gy
H&N/Lung Brachial Plexus 0.1 cc < 66 Gy
H&N Brainstem 0.1 cc < 55 Gy
H&N Optic Nerves/ 0.1 cc < 55 Gy
Chiasm
H&N Spinal cord (+ 0.1 cc < 50 Gy PRV
3 mm)
H&N Uninvolved Mean < 45 Gy
constrictor
H&N Contralateral Mean < 39 Gy
SMG
H&N GSL Mean < 35 Gy
H&N Cochlea [2] Mean < 35 Gy <12–14 Gy/1 fx For mean < 45 Gy, risk
of hearing loss <30%.
H&N Oral cavity Mean < 30–40 Gy
H&N Lacrimal gland Mean < 34 Gy
H&N Combination Mean < 25 Gy
parotid [3]
H&N Contralateral Mean < 20 Gy
parotid
GI Small bowel/ 1 cc < 60 Gy V50 < 2%
Stomach
GI Duodenum 2 cc < 55 Gy
GI Liver V30 < 60% Mean < 25 Gy
GI (SBRT) Liver 700 cc < 21 Gy
GI (SBRT) Duodenum V15 < 9cc/1 fx Murphy (2010) [4]
V20 < 3cc/1 fx
Dmax <23 Gy
GI Kidney [3]
GI Kidney V18 < 66%
GI Bladder V40 < 35% Same as prostate
GI Femoral heads 1 cc < 45 Gy V40 < 35%
GI Heart V40 < 100% V50 < 33%
20 N. G. Zaorsky et al.
Dose
Site Organ at risk Dose constraint 1 Dose constraint 2 constraint 3 Notes
GI Lung V5 < 50% V20 < 20% Mean < 12 Gy
Thoracic Lung (−PTV) V5 < 60% V20 < 30% Mean < 18 Gy Lung V5 not
associated with
toxicity per secondary
analysis of RTOG
0617 [5]
Thoracic Lung [3] <20% risk of
pneumonitis
Thoracic Contra lung after V20 < 7% Rice (2007) [6]
EPP [6]
Thoracic Heart V45 < 2/3 V50 < 25% V60 < 30% RTOG 0617
Thoracic Heart V30 < 50% V45 < 35% RTOG 1308
Thoracic Heart
Thoracic Esophagus V60 < 30% Mean < 34 Gy
Thoracic Spinal cord 0.1 cc < 45 Gy Max dose 54 Gy has
<1% RT myelopathy
per QUANTEC
Breast Heart V30 < 1% Mean < 4 Gy NSABP B51
Breast Lung V20 < 15%
Breast Breast 0.03 cc < 115% NSABP B51
GU Bladder V40 < 50% V65 < 25%
GU Femoral heads V50 < 10%
GU Rectum V40 < 35% V65 < 17%
GU Rectum V50 < 3cc (50 Gy V24 < 35% of Kim (2014). Using
SBRT) rectal wall 45–50 Gy/5 fractions
circumference for prostate cancer [7]
GU Penile bulb
GU Kidney
GU (HDR) Bladder V75 < 1 cc
GU (HDR) Rectum V100 < 1 cc
GU (HDR) Urethra V125 < 1 cc V150 = 0 cc
GYN Rectum V40 < 60% RTOG 0724 (postop
cervix)
GYN Rectum V40 < 80% RTOG 1203 (cervix +
endometrial)
GYN Bowel space (SB, V40 < 30% RTOG 0724, RTOG
colon, sigmoid) 1203
GYN Bladder V45 < 35% RTOG 0724, RTOG
1203
GYN Bone marrow V10 < 90% V40 < 37% RTOG 1203
GYN Bladder pt <75 Gy, <80% of D2cc < 90 Gy
dose,
GYN Rectal pt <70 Gy, <70% of D2cc < 75 Gy
dose
GYN Sigmoid D2cc <75 Gy
GYN Small bowel D2cc < 60 Gy V55 < 15 cc Verma (2014) [8]
Gyn (HDR) Rectum/Sigmoid 2cc < 75 Gy D2cc = the minimum
dose to the maximally
irradiated 2cc
Gyn (HDR) Bladder 2cc < 90 Gy
Gyn (HDR) Upper vagina Point <120 Gy
Gyn (HDR) Lower vagina Point <90 Gy
General Principles of Radiation Oncology 21
HN, CNS
Protocols, institutional SRS (for brain)
PTV HN: Max < 110% GBM: CI < 1.5–2
Rx. D95% > 100% D95% = 60 Gy
Rx. D99% > 93% Rx D99% = 54 Gy
Temporal lobe max < 68 Gy, 1 cc < 58 Gy
Normal brain Max 60 Gy V12 < 10 cc (RN predictor)
Brainstem Max < 54 Gy (HN), 60 Gy (RTOG 0529, Max 12 Gy (required)
0825), 64 Gy (QUANTEC) Max 45 Gy (for trigem neuralgia)
V54 < 20% (ependymoma)
Chiasm; Optic N Max 54 Gy (HN) – 55 (GBM) Max 8–10 Gy (prefer), 12Gy
(required). Stay >3 mm from GTV
Pituitary max 50 Gy
Hippocampus (RTOG 0933) Max 16–17 Gy D100 < 9 Gy
Spinal cord PRV max 45–50 Gy Max 14 Gy
Retina max < 45 (HN) – 50 (GBM)
Lens max < 7–10 Gy
Lacrimal gland mean < 34 Gy (41.4in RMS)
Cochlea max 35 Gy. 55 in NPX. Max 9 Gy
mean < 35–45 Gy (also peds) Mean < 4 Gy
V30 < 50%
Inner ear max < 50 Gy
Dorsal vagal complex mean < 12 Gy
Parotid, contra mean < 20 Gy (<20% D salivation)
Parotids, combo mean < 26 Gy
SMG, contra mean < 35–39 Gy
GSL (larynx) max < 66 Gy
mean < 35–45 Gy (involved)
mean < 20 Gy (uninvolved)
V55 < 10%
Oral cavity, ant V35 < 35%
V30 < 65%
Oral cavity, uninvolved max < 60 Gy
mean < 30–40 Gy
Lips mean < 20 Gy
Mandible 0.1cc < 70 Gy, 1 cc < 75 Gy
Pharyngeal constrictor, sup V65 < 30%
V55 < 80%
Pharyngeal constrictor, mid V65 < 75%
Pharyngeal constrictor, V60 < 15%
uninvolved V50 < 33%
mean < 45 Gy
Esophagus mean < 30–34 Gy Max < 16 Gy
V11 < 5 cc
Brachial plexus (1 of 2) Max < 66–70 Gy
22 N. G. Zaorsky et al.
Lung
Lung, 5 fx SBRT Lung, 30-35 fx
(RTOG 0813) (RTOG 1306/1308, LungART)
PTV Rx to 60–90% IDL
V100 > 95%
Max < 120%
Lungs (-PTV or ITV) V20 (bilat) < 10% V20 < 35% (<25% if BID)
V20 (ipsi) < 25% V20 < 31% (if PORT)
D1.5L max 12.5 Gy V20 < 8% (if meso)
D1L max 13.5 Gy V5 < 60%
Mean < 20 (<13 if BID, <8 if
meso)
Heart 32 Gy to < 15 cc V60 < 30%
105% PTV Rx V45 < 35% (1308)
V35 < 30% (Lung ART)
V30 < 50% (1308)
Mean < 26 Gy (not 35), < 9 for
meso
heart block at 19.8 (meso)
Stomach NS Block entirely for meso
Liver Block entirely for meso
General Principles of Radiation Oncology 23
GI
Esophagus Gastric Liver SBRT, 5 fraction Pancreatic Rectal Anal
(RTOG 1010) (RTOG 0114) (RTOG 1112) (RTOG 0848) (RTOG 0822) (RTOG 0529)
IMRT IMRT IMRT 3D
PTV Max < 110%
D95 = 100
Cord Max <45 Max <45 Max 20–25 Gy Max < 45
Esophagus Max 32 Gy
Lungs (-PTV or V30 < 20% V30 < 20%
ITV) V20 < 25% V20 < 25%
V10 < 40% V10 < 40%
V5 < 50% V5 < 50%
Mean < 20 (5–8) Mean < 20 (5–8)
Heart Max 50 Gy Max 50
Mean < 30 Gy Mean < 30
V40 < 30–50% V40 < 30%
Stomach Max 30 Gy
Liver Mean < 21 V30 < 60% 700 cc < 21 Gy Mean < 21–25
V30 < 30% Mean < 15 V30 < 30%
liver < 35 (10% risk)
liver < 50 (50% risk)
Small bowel Max < 56 Max 30 Gy Max < 54 V45 < 65 cc Max < 50 Gy
D15% < 45 V40 < 100 cc V45 < 20 cc
D10% < 50 V35 < 180 cc V35 < 150 cc
V30 < 300 cc V30 < 200 cc
Large bowel Max 32 Gy V50 < 0.5 cc V45 < 20 cc
V35 < 150 cc
V30 < 200 cc
Kidneys (1 of 2) Max 45 Mean < 18 Mean < 10 Gy D30-50% < 18 V18 < 2/3 of
V20 < 30% V20 < 30% Mean < 18 kidney
Kidney (1 of 1) V20 < 20% V20 < 20% V10 < 10% D15% < 18
V20 < 20%
Femoral heads V50 < 0.5 cc V44 < 5%
V40 < 35%
V30 < 50%
Bladder V50 < 0.5 cc V50 < 5%
V40 < 40% V40 < 35%
V35 < 50%
Iliac crest V50 < 5%
V40 < 35%
V30 < 50%
Genitalia V40 < 5%
V30 < 35%
V20 < 50%
GU
24
Prostate Prostate hypofrac Prostate Prostate post- Prostate SBRT Prostate LDR Prostate HDR Bladder Seminoma
conventional 80 70.2 Gy/26 hypofrac op 36.25/7.25 Gy 145 Gy mono I125 13.5 Gy x 2 mono 45 Gy, CD to 64.8 20–36 Gy
Gy/20 (Pollack/FCCC) 70 Gy/25 68 Gy/34 (RTOG 0938) Ir192 Gy
(Pollack/FCCC) (RTOG 0415) (Pollack/RTOG
0534)
PTV D100% > 95% D100% > 95% V100 >98% D100% > 95% D0.03cc <107% D90 > 100% of dose V100 > 90–95% of
V95% > 100% V95% > 100% Max < 107% V95% > 100% Rx CK V100 > 90–95% dose
Dmax < 115% D0.03cc <120% V150 < 50–60%
Rx non-CK
V100 >95%
D0.03 >95% Rx
Bladder (-CTV) V65 <25% V50 < 25% V79 <15% V65 < 50% D1cc <105%
V40 < 50% V31 < 50% V74 <25% V40 < 70% D90% <90% Rx
V69 <35% D50% <50% Rx
V64 <50%
Rectum V65 < 17.5% V50 < 25% V74 <15% V65 < 35% D1cc <105% RV100: <1 cc on D2cc £ 75 Gy V60 < 5%
V40 <35% V31 < 50% V69 <25% V40 < 55% D90% <90% Rx day 0; and < 1.3 cc EQD2 V30 < 50%
V64 <35% D80% <80% Rx on day 30 D2cc < D1cc < 100% of
V59 <50% D50% <50% Rx prescribed dose; and dose (FCCC)
D0.1cc < 200 Gy
GYN/STS
Cervix Endometrial Sarcoma
45 Gy WPRT+ 6 Gy x 3 BT boost + 50 Gy / 25
CD to 54 Gy PMB 45-50.4 WPRT
+/– CD 60 to LN Vag 45 Gy + 6x3
+ 6 Gy x 5 BT (RTOG 0418; GOG 279)
RT alone:
5x5 + 45 Gy
PTV HR-CTV = 85 Gy
Point A = 85 Gy
Point B = 60 Gy
Kidney (1 of 2)
Kidney (1 of 1)
Small/large bowel Dmax 60 Gy V55 < 5 cc
potential space V45 < 200 cc
SB V40 < 30%
Bladder (-CTV) D2cc £ 90 Gy D2cc £ 90 Gy
(4 Gy/fx in 5 fx) V45 < 35%
Bladder pt £ 70 Gy V40 < 50%
Rectum D2cc £ 70 Gy (4.5 D2cc £ 70 Gy
Gy/fx in 5 fx) V40 < 80%
Rectal point £ 70 Gy V30 < 60%
Sigmoid D2cc £ 70 Gy
Vagina Surface < 140% pt A Upper 1/3: 120 Gy Anus/vulva:
dose Mid 1/3: 90 Gy V30 < 50%
Low 1/3: 70 Gy
VSD = ~150% rx
Ovary Sterilize: 2 Gy;
Fail: 5-10 Gy
Testis Sterilize: 2 Gy V3 < 5%
Vagina upper third: 120 Gy upper third: 120 Gy
middle third: 80–90 Gy middle third: 80–90 Gy
lower third: 60–70 Gy lower third: 60–70 Gy
Femoral head V30 < 15% V60 < 50%
V44 < 5%
Wt bearing bone V50 < 50%
Epiphysis max 20
Skin strip V20 < 50%
26 N. G. Zaorsky et al.
Lymphoma
Constraint
Rule of thumb use mean 5 Gy on every OAR.
If can't meet this, then use constraints below.
Lung V20 < 20%
Mean < 12 Gy
Heart Mean < 15 Gy
Bone Mean < 8 Gy
Breast < 5–10 Gy
Thyroid Mean < 15 Gy
Kidney Mean < 20 Gy
Liver V25 < 50%
Testes 2 Gy – sterility
0.5 Gy – acute azospermia
Ovary 5-10 Gy failure
2 Gy – sterility
Peds
Organ Constraint
Lacrimal gland Max 41.4 Gy (RMS)
Lens Max 14.4 Gy
Globes Max 35 Gy (Ependymoma)
Optic N Max 56 Gy (Ependymoma)
Max 45–54 Gy (RMS)
Brainstem Max < 63 Gy (Ependymoma)
D90% < 44 Gy (Ependymoma)
Cord Max < 56 Gy
D90% < 3 Gy
(Ependymoma)
Chiasm Max 46.8–54 Gy (RMS)
Heart Max 30.6 Gy (RMS, Wilms)
Lungs (<1/2 of 15 Gy/1.5 Gy max (RMS)
combined lung V15 < 33% (NB)
volume in PTV)
Kidney V14.4 Gy < 100% (NB/Wilms)
100% kidney < 14.4 Gy (NB/Wilms)
Ipsi V19.8 Gy < 50% (NB/Wilms)
Contra kidney V12 < 20% (NB/Wilms)
Max 19.8Gy (RMS)
Liver V9 < 50% (NB/Wilms)
V18 < 25% (NB/Wilms)
V30 < 15% (NB/Wilms)
Max 23.4 Gy (RMS, Wilms)
Bladder Max 45 Gy (Wilms)
Vertebral body If in close proximity, treat whole VB to 18 Gy.
Small bowel V45 < 50% (RMS)
Rectum Max 45 Gy (Wilms)
General Principles of Radiation Oncology 27
combined modality therapy and pathologic response. Radiation Oncology Group Trial (TROG 01.04). Ann
J Clin Oncol. 2006;24:3953–8. Surg. 2017;265:882–8.
29. Gerard JP, Azria D, Gourgou-Bourgade S, et al.
33. Mak RH, Hunt D, Shipley WU, et al. Long-term out-
Clinical outcome of the ACCORD 12/0405 PRODIGE comes in patients with muscle-invasive bladder cancer
2 randomized trial in rectal cancer. J Clin Oncol. after selective bladder-preserving combined-modality
2012;30:4558–65. therapy: a pooled analysis of Radiation Therapy
30. Rodel C, Graeven U, Fietkau R, et al. Oxaliplatin Oncology Group protocols 8802, 8903, 9506, 9706,
added to fluorouracil-based preoperative chemo- 9906, and 0233. J Clin Oncol. 2014;32:3801–9.
radiotherapy and postoperative chemotherapy of 34. Moore DH, Ali S, Koh WJ, et al. A phase II trial of
locally advanced rectal cancer (the German CAO/ radiation therapy and weekly cisplatin chemotherapy
ARO/AIO-04 study): final results of the multicentre, for the treatment of locally-advanced squamous cell
open-label, randomised, phase 3 trial. Lancet Oncol. carcinoma of the vulva: a gynecologic oncology
2015;16:979–89. group study. Gynecol Oncol. 2012;124:529–33.
31. Bujko K, Wyrwicz L, Rutkowski A, et al. Long-course 35. Moore DH, Thomas GM, Montana GS, et al.
oxaliplatin-based preoperative chemoradiation versus Preoperative chemoradiation for advanced vul-
5 x 5 Gy and consolidation chemotherapy for cT4 or var cancer: a phase II study of the Gynecologic
fixed cT3 rectal cancer: results of a randomized phase Oncology Group. Int J Radiat Oncol Biol Phys.
III study. Ann Oncol. 2016;27:834–42. 1998;42:79–85.
32. Ansari N, Solomon MJ, Fisher RJ, et al. Acute
36. Keys HM, Bundy BN, Stehman FB, et al. Cisplatin,
adverse events and postoperative complications in a radiation, and adjuvant hysterectomy compared
randomized trial of preoperative short-course radio- with radiation and adjuvant hysterectomy for
therapy versus long-course chemoradiotherapy for bulky stage IB cervical carcinoma. N Engl J Med.
T3 adenocarcinoma of the rectum: Trans-Tasman 1999;340:1154–61.
Pediatrics Cancers
2
Nicholas G. Zaorsky, Daniel M. Trifiletti,
and Christine E. Hill-Kayser
Abstract • ATRT
This chapter discusses the general manage- • Mets
ment of patients with various pediatric solid
tumors, with special focus on principles that
Posterior Fossa Syndrome
guide radiotherapy management. Several key
• Mnemonic: SAME
components of pediatric cancer care are
• Swallowing dysfunction
discussed.
• Ataxia
• Mutism
• Emotional lability
• Occurs postop after resection
Pediatrics Pearls [1]
N. G. Zaorsky (*)
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
D. M. Trifiletti
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
C. E. Hill-Kayser
University of Pennsylvania, Philadelphia, PA, USA
unfav subsites emb15– FKR FKR (bad) plan w adapted IR: 4w Group II: pre-chemo GTV + 1 cm
19 alv t(2:13) surgery Group I tage 1–3: HR: 20w R1: 36 Gy; 41.4 Gy PTV = CTV + 0.5; or
t(1:13) VAC, no RT CNS: 0w pLN+ to entire LN PTV = preop,
NF1, Everyone else: chain pre-chemo GTV +
Li-Fraumeni, VAC and RT Group III: 1.8–2 cm
Beckwith- 45 Gy orbit; 50.4 Gy
Wiedemann others
SRBCT
NRSTS Bone pain Synovial Yes Y Surg, then PORT N All LG → observe GTV = tumor on MRI
sarcoma, for R1 HG, ≤ 5 cm R0 → post contrast
t(X;18) HGs + chemo if observe CTV = 1.5 cm radially
MPNST >5 cm HGs HG, ≤ 5 cm R1 → and longitudinally
55.8 Gy PTV = CTV + 0.5 cm
HG, > 5 cm → w/ daily KV imaging
55.8 Gy + Ifos/Doxo x 5
Unresected → NA CRT
(45 Gy)
Langer- HSC: skull 2 Birbeck LSD, Yes Y/N If multiorgan 14d for DI N DI: 15 Gy to pituitary/
hans cell lesions, DI, granules multisys- involvement: HD hypothalamus
histiocy- exophthalmos Hand- tem pred + vinblastine Bone: 5–10 Gy (e.g.,
tosis LSD: hepato- Schuller- involve- If single bone, 3 Gy x 3 fractions with
(LCH) splenomegaly, Christian dz ment then RT small margin)
LAD, lung Letterer-Siwe Adult: 15–24 Gy/2 Gy
lesions, bone disease fraction
lesions
33
34
Location and
classic Bad
Tumor presentation Med age Buzzwords prognostics Bx tumor? BM bx?
Tx paradigm Start RT by CSI Dose Volumes
Ewing Soft tissue (e.g., 14, t(11:22), Yes Y
Neoadj chemo w chemo, N R1/pN+/definitive VB: GTV1 = pre-chemo dz
sarcoma femur), “awakens skewed t(21:22), VDC-IE x 12w. restage, then 50.4 Gy/25 fxs in tissue + bone
from sleep,” right EWS Restage at week week 13 R2/definitive elsewhere: CTV1 = GTV1 +
“growing pains” SRBCT 12 45 Gy to pre-chemo 1–1.5 cm
“moth eaten,” MRI + PET. Then preop +1.5 cm, boost GTV2 = pre-chemo dz
“sunburst,” surg or RT. PORT GTV + 1 cm to in the bone and
“onion skin,” if indicated. Hold 55.8 Gy/31 fxs post-chemo dz in tissue
“laminated,” adria and All w concurrent chemo CTV2 = GTV2 + 1 cm
Codman’s actinomycin (VC/IE), but hold adria
triangle, during RT. Adj M+: after all chemo,
sclerosis chemo for all 48 WLI to 15 Gy. Boost
w. residual to 43 Gy
PORT if R+
(<5 mm), spill,
>10% viable
Nephro- Intrarenal, 4y WT1: WAGR Del ch22q, No Y, for Surg for all. Then POD 8 N Stages I–II, FH: no RT usually AP/PA,
blastoma/ circumscribed unilat2.5y (del 11p13, LOH 1p, (becomes rhab- risk adapted (UH), max Stage III or UH: Preop GTV + 1–2 cm
Wilms Can spill bilat WT1) LOH 16q, Stage III), doid Chemo for low 14 for FH 10.8 Gy to flank based on CT, US, MRI,
(WT) Abdominal mass 90% are Denys-Drash gain 1q, unless and CC risk Diffuse anaplasia or IVP
Wilms = well. <5yo (WT1 mut) blastemal Stage V Chemo, then RT rhabdoid: 19.8 Gy to
Rarely crosses WT2: histo, suspected for std risk flank
midline. Won’t Beckwith- anaplasia, Avoid R2: boost another
move w Wiedemann CCSK, vigorous 10.8 Gy (21.6 Gy total)
inspiration. (11p) Rhabdoid exam Spill or rupture:
Macroglossia, No calcs 10.5 Gy WART + boost
aniridia, GU Stage residual by 10.5 Gy/7
hypospadias or III = BSS- (21 Gy total)
cryptorchidism. LURPP All w concurrent vinc
CCSK: the bone, SRBCT Liver or unresected LN:
brain mets; 19.8 Gy
Rhaboid: brain Whole lung
mets. Lung mets 12/8 + boost residual to
> bone (vs NB). 19.5 or resect
WBRT: 21.6 Gy/12
Focal liver: 19.8 Gy/11
fxs
N. G. Zaorsky et al.
Neuro- Extrarenal, 1.5y #1 extracra- SANDS: Yes, after Y Surg for all. 4–6 weeks N No RT for low or int preop, post-chemo
blastoma ill-defined nial solid ↑stage, ruling out Chemo for low after ASCT risk; 24 Gy if cord tumor vol + 1.5 cm
(NB) margins. 65% tumor in peds ↑age, Wilms risk+. RT rarely with CAPE compression, residual Boost postop
have abd mass Homer- n-myc, +BM bx needed for low or High risk: M2: CTV = 1 cm to
(adrenal or Wright diploid +MIBG std risk. For high PTV = GTV + 1.5 cm. 55.8 Gy
paraspinal). “Not rosette (hypodip- risk (Stage 21.6/1.8 Gy +/− M3
Pediatrics Cancersr
well.” Cross Calcified loid good), 2–4 N-myc+; 14.4 Gy for residual dz Diffuse: CSI 39.6
midline. HTN, Blueberry Shimada Stage 4 N-myc+), (> 1 cc or MIBG-avid) GTV above vs below
renal art muffin. 1p/11q loss induct chemo, Met site: 21.6 Gy, no conus: 45 vs 50.4 Gy
compression. Raccoon eyes surg resection, boost
Opsoclonus. SRBCT myeloblat chemo, If cord compression:
Calcified. HMA/ SCT, EBRT, surg, then chemo
VMA+. Bone immunotherapy Cord compression: 9 Gy
mets > lung (vs if <3. 21.6 if >3yo
WT) After RT, cis-retinoic
acid
Massive HSM:
4.5 Gy/1.5 Gy
Retino- Retina. Opthalm infant RB1 on ON No bx will N Chemo w vinc/ N EBRT to 40 Gy Pre-chemo and
blastoma sx: leukocoria, ch13, G1/S involve- seed. EUA carbo.etop x 6c. I125 plaque BT 40 Gy presurgical disease
strabismus checkpoint ment, by Then local to apex +5 mm = CTV
Flexner- extraocular opthal- therapy (cryo, ARET0321: Induction PTV = CTV + 5 mm
Wintersteiner m mologist surgery, or RT) chemo, consolidation w
rosette involve- w retinal Bilat are txd as SCT (stage IVA/IVB
“trilateral” ment. Thus, mapping separate only), and response-
SRBCT need bone primaries. adapted EBRT:
scan, BM II–III: if residual, dose
bx, CSF. is 45 Gy
IVA: <5 mm residual or
CR: no RT
IVA: > 5 mm residual,
36 Gy
IVB: 36 Gy CSI, 45 Gy
to cranium, 50.4 Gy to
pineal gland
(continued)
35
36
Location and
classic Bad
Tumor presentation Med age Buzzwords prognostics Bx tumor? BM bx? Tx paradigm Start RT by CSI Dose Volumes
Medul- CNS. 4th vent. 7-9yo + Chang Myc No bx. N Resection. If MRI brain Y <3yo: Resect, chemo, CTV = preop tumor +
loblas- Neuro sx 25yo staging Group 3 Surg for <3yo, chemo. If 24–48 h some centers radiate postop cavity +1.5 cm
toma Child = mid COG: avg or all. No LP >3yo, Then RT w postop.MRI tumor bed only PTV = CTV + 5 mm
(and Adult = lat High risk if hi ICP vincristine.RT spine Consider CSI at 3yo.
supratent Blocks Aqueduct WNT (good) dose depends on 10–14d Std risk: CSI 23.4.
PNET) of Sylvius Group 4 risk group. Avg postop Cavity boost to 54 Gy.
(intermed) risk is >3 yo, < Then CSI by High risk: CSI to
MYC (bad) 1.5 cm2, M0, not POD31 36 Gy. Cavity boost to
Group 3 (v PNET. 54–55.8 Gy
bad) Brain mets: 55.8 Gy
Post-fossa Spine mets: 45 Gy
syndrome
Homer-
Wright
rosette
Perivascular
pseudorosette
SRBCT
ATRT CNS. 4th vent. <3 INI-1/ No bx. Y Like high risk MRI brain Y >3yo: 36 Gy CSI; boost CTV = preop tumor +
Neuro sx SMARCB1 Surg for medullo 24–48 h to 54–55.8 Gy. If M+, postop cavity +1 cm
loss all. No LP Surg for all. postop, MRI boost spine dz to 45 Gy PTV = CTV + 5 mm
Vimentin, if hi ICP Chemo investiga- spine < 3yo: 50.4–54 Gy to
EPA, SMA tional. PORT 10–14d primary alone
postop
Then CSI by
POD31
Pineo- CNS Homer- Surg for Like high risk Y If >3yo, CSI to 36,
blastoma Wright all. No LP medullo, but boost to 54 Gy. If <3yo,
rosette if hi ICP lower boost. Surg 50.4–54 Gy to primary
Flexner- for all. Adj RT site
Wintersteiner
rosette
Pineocy- Surg for N Like LGG N If R0, obs. If R+, 54 Gy
toma all
N. G. Zaorsky et al.
Ependy- CNS. 4th vent. 5yo + WHO G1–4 Surg for N Surg for all. MRI MRI spine N, Resid tumor + bed to Pre- and postop
moma 70% infratent, 35yo Ependymal all. No LP brain >10d postop and CSF usly 54, G3 or R1, boost to CTV = GTV + 1 cm
30% supratent. rosettes if hi ICP if wasn’t done cytology 59.4 respecting
Neuro sx, Perivasc preop 10–14d brainstem
Calcified pseudoro- RT if R+ or G2+. postop CSI only if M+ in CSF,
settes Role of chemo to 36 Gy, boost to 45 Gy
Pediatrics Cancersr
Treatment
ALL Chemo
–– Very commonly used. Causes neuropathic
pain in 35%.
–– For CNS-directed therapy, IC MTX is usually
used.
Pediatrics Cancersr 41
Undiff 5% 40%
Fusion-positive 25% FOXO1+; Trunk or abd 15–19 54%
(formerly t(2;13):
Alveolar) PAX3/FKHR
t(1;13):
PAX7/FKHR
–– IRS V: EFS for low risk ~87%, intermed risk • Stage 1, Group II: surgery→chemo(VA) +
73%, high risk 32%. Intermed pts rando to RT at week 13 (36Gy for N0, 41.4Gy for
VAC vs VTC + 2nd look surg. VAC alone N1)
won. High-risk pts VCR + I vs I alone. • Stage 1, Group III: surgery→chemo
VCR + I won. (VA) + RT (50.4Gy except orbit = 45Gy)
–– IRS-VI: • Stage 2 Group II: surgery→VAC → RT at
–– COG D9803: intermed risk→ VAC vs VAC alt wk 13 (36Gy)
with VTC. No difference in DFS or LF. • Stage 3, Group II: surgery→VAC → RT at
–– Mandell 1990: retrospective of group II pts. wk 13 (36Gy for N0, 41.4 for N1)
No LC difference between <40 and >40Gy. –– Intermed Risk
–– ARST0331: goal to decrease Cytoxan dose to • Surgery→chemo→(repeat surgery if
decrease sterility. RCT initially allowed omis- possible)→RT(50.4Gy).
sion of RT for group II/III females w vaginal –– High Risk
RMS w excellent response to chemo but • Chemo (VCPT→VAC). RT to primary and
amended to mandate RT for them. metastatic sites (45–50.4Gy). RT may be
initiated week 0 for symptomatic intracra-
nial extension or cord compression. Dose is
Management 50.4 for Group 3 tumors, unless orbital,
–– GTR. RPLND for paratesticular, axillary or where dose is 45 Gy.
inguinal LND for extremity, and if positive, • For M+ PAX/FOXO1 negative, usually no
then they are M+. RT to mets.
–– Bladder/prostate → pelvic LN sampling (if + • For M+ PAX/FOXO1 negative, RT to mets.
→ LND). NPX has high LN involvement, but • High-risk non-CR/R0 mets:
no LND unless cN+. • Try to tx at week 20 when tx primary, if
• →VAC not possible give at end
• →RT • R0 resection or CR after chemo = No RT
• →chemo • R1 resection = 36 Gy
–– Start RT based on risk classification. • R2 resection or no surgery = 50.4 Gy
–– Dose of RT based on grouping.
Lung Mets
No Surgery for • Always treat lung mets even if CR. Give WLI
–– Parameningeal at the end of all tx
–– Bladder/prostate (except if residual dz) • WLI 15 Gy/1.5 (12 Gy if <6yo),
–– Vagina/cervix/uterine • then boost limited gross lung disease to
–– Orbit 50.4 Gy if feasible
• boost limited lung disease to 54 Gy (give at
No RT Given Only if end of all chemo).
–– Vagina/cervix/uterus AND embryonal with
CR after induction chemo Technique
–– Bx→VAC –– Orbit: biopsy only→45 Gy to tumor+1 cm
–– Group I extremity s/p amputation (5 mm CTV and 5 mm PTV; per IRS-VI),
–– Group I paratesticular s/p orchiectomy crop anatomic boundaries (pre-chemo
–– Metastatic site R0 scans).
–– Otherwise CTV = GTV + 1–1.5 cm. Note
General Guidelines 1 cm used on IRS-VI. Definitive non-orbit
–– Low Risk dose is 50.4 Gy.
• Stage 1–3, Group I: surgery→chemo (VA –– Contours should be preop and pre-chemo
or VAC). No RT volume.
Pediatrics Cancersr 45
–– PTV = CTV + 0.5 cm, i.e., • Never deliver actinomycin or adria w RT.
PTV = GTV + 1.8–2 cm • Cytoxan + RT can cause mucositis and
–– Include the entire LN chain if N+ consider dose reduction.
–– Site-specific recs: • Lung metastases are not common, and
• Confine orbital CTV to orbit unless tumor management is not well studied, but whole-
extent beyond. lung RT (15 Gy in 10 fractions) may be
• Boys 10+ with paratesticular tumors need considered in setting of multiple lung
aggressive LN sampling. Boys <10yo metastases.
should have thin-cut CT (5 mm slices) or
retroperitoneum and pelvis, w further Timing
workup for suspicious LNs. Boys all ages • For FH: usually w 13 unless reason to delay
need LND if cLN+. (e.g., marrow preservation).
• Radiate only regionally involved LN • For UH: w4.
basins. • If symptomatic intracranial extent can con-
• Gen no ENI in orbit, HN, and female GU. sider RT at w0.
• Can move and replant gonads. • Timing (week 4 vs 13) probably is not impor-
• LND required for paratesticular, pelvic, tant (Spalding, IJROBP, 2013).
and extremities (>20% LN+ rate). • WLI, if indicated, may be deferred until com-
• CR after chemo in bladder/vagina/uterus pletion of planned chemotherapy.
doesn’t need RT.
Group (post-op)
Descripti I II III IV
Sites TNM
on (Ro) (IIA: R1 / IIB: pLN+) (R2 / GTV) (M+)
Fav site, Fav Any T,
M0 site Any N Orbit:
UH (w13): 45 Gy
1
36 Gy Low High
w13
Stage (pre-op)
Ewing Sarcoma [4–8, 16–21] –– Chest imaging necessary to rule out RMS,
Wilms
Overview –– LDH
–– Three hundred cases/yr. Median age 14, –– Bx: open is best. Longitudinal cut
skewed right. –– BMBx
–– Caucasian. –– Cytogenetics bc t(11;22) found in 85%
–– Can arise in bone or soft tissue. –– Fertility preservation
–– Tends to be radiosensitive. • Male (infertility = 2 Gy) → sperm bank +
–– Ewing sarcoma is generally staged as local vs clam shell
metastatic but refers to STS chapter for refer- • Female (ovarian failure = 8 Gy) → ovarian
ence for a formal staging system transposition
–– DDx (“EGMODE”)
Natural History • Epiphysis = Giant cell
–– Location • Soap bubble appearance
• 25% in axial skeleton (CW, spine, skull) • Metaphysis = osteosarcoma
• 20% in pelvis • Sunburst, Codman’s triangle
• 65% in lower skeleton (30% in femur, 10% • Diaphysis = Ewing
in tibia) • Moth-eaten, onion skin
• 15% in upper skeleton • MRI findings: T1 hypointense, T2
–– 80% localized but 25% have macromets and heterogeneous
nearly all have micromets • Other: met, trauma, osteomyelitis, lipoma,
–– Strong rationale for local therapy to DM, Legg-Calve-Perthes, osteosarcoma, RMS,
being investigated on open COG study sarcoma, lymphoma, neuroblastoma
currently –– DMs are principle concern
–– DM sites: the lungs, bone, marrow –– Plain film: “moth eaten,” “sunburst,” “onion
skin,” “laminated,” Codman’s triangle, sclero-
Histology sis, permeative. Usually lytic at diaphysis
–– SRBCT
–– Ewing family: Ewing sarcoma (87%), Poor prognostics: central lesions, >8 cm
Extraosseous Ewing (8%), PNET (5%), (200 cc), M, high LDH, soft tissue extension,
Askin’s tumor (a Ewing of the chest wall) poor chemo response
–– EWS: RNA-binding protein
–– EWS and FLI work as chimeric TF Trials
–– t(11:22): involves the FLI1 on ch11 and EWS –– IESS-1: nonmetastatic dz → VAC + D vs VAC
gene on ch22 vs VAC + prophylactic whole-lung
–– t(21:22): 5–10%. ERG and EWS RT. VAC + D won. 5 yr. RFS (60 → 24 → 44%).
–– ↑c-myc activity (↑n-myc in neuroblastoma) –– IESS-2: VAC + D high dose vs continuous.
–– CD99+, vimentin+, NSE- (PNETs are CD99+, High dose improved RFS but not OS.
vimentin+, NSE+) –– IESS-3: VACD +/− IE. More chemo won. 5 yr
OS 61 → 72%. Did not improve OS for M1
Disease 5y OS % disease.
Localized 70 –– CESS 86: chemo→ surgery vs surgery+PORT
Single met 50
vs RT alone. No difference in 5 yr OS
Diffuse mets 30
(69%). LC worse without surgery
(100 → 95 → 86%).
Workup –– POG 8346, Donaldson 1998: chemo→surgery
–– H&P, labs, plain film or RT. RT was randomized whole bone
–– CT/MRI, bone scan or PET-CT, CT chest 39.6 + boost 16 Gy vs 4 cm margin to
Pediatrics Cancersr 49
55.6 Gy/1.8 Gy fx. Central path and RT • Surg margin: 2–5 cm is preferable for bone
review. No difference in LC or RFS. LC was but accepts >1 cm, >0.5 cm for soft tissue
53% in both RT arms. Thus, do not treat whole (fat, muscle, medullary bone), >0.2 cm for
bone. LC was 80% in extremity, 69% proxi- fascia, periosteum, and septae.
mal, 82% central, 44% pelvis. Protocol devia- • Unresectable bones (more likely axial) are
tion → poor outcomes. maxilla, BOS, vertebrae, and periacetabu-
–– EICESS analysis: any patients with lung mets lar pelvis. Also soft tissue. These typically
benefited from WLI with improved EFS. receive RT.
–– Schuck 2002: Askin tumors. 7 yr EFS • If giving RT, adriamycin and actinomycin-
improved with hemithorax RT and boost to D are held during RT.
primary. –– (3) PORT if necessary. Indications: R+ (<5–
–– No RCTs to guide surgery, RT, surg + 10 mm) and poor response from chemo (<90%
RT. Selection bias confounds data; central tumor necrosis)
tumors likey to get RT, while peripheral likely –– (4) Adjuvant chemo, up to 48w
to get surgery. –– LC w RT alone is >70%. LC is lowest in pel-
–– AWES0031: chemotherapy Q2 weeks vis is 70–80% and 90–95% in the
improved EFS compared to Q3 weeks. extremities
–– AEWS 1221: current COG M+ ES protocol –– Doses >60 Gy have high-risk bone
examining anti IGF-1R mAb ganitumab. All malignancy
pts have local therapy to primary + DMs. RT –– Doses <40 Gy have high LR rates
to metastatic sites is a study question. SBRT
recommended for DMs in bone <5 cm in max
dimension. NCCN
–– Preop RT (Rare to do this!)
Chemo • 36 Gy.
–– VDC/IE: vincristine, adriamycin, cyclophos- –– Definitive RT
phamide, ifosfamide, and etoposide. The stan- • MRI essential, inc T2/FLAIR.
dard in the USA. • GTV1 = pre-chemo dz in tissue + bone.
–– VTC/VDC: vincristine, topotecan, and cyclo- • CTV1 = GTV1 + 1–1.5 cm.
phosphamide/VDC. This is on AEWS1031 • GTV2 = pre-chemo dz in bone and post-
–– VIDE: vincristine, ifosfamide, doxorubicin, chemo dz in tissue. If dz is pushing on an
and etoposide. The standard in Europe. organ but not invading, and has response,
do not pre-chemo volume.
Ewing Treatment Paradigm • CTV2 = GTV2 + 1 cm.
–– (1) NAC w VDC, alt with IE x 12 weeks. Note • PTV = CTV + ~0.5 cm.
that on AEWS1031, randomization to VDC/ • PTV1: 45 Gy/25 fxs. Exception is extraos-
IE vs VTC/VDC. seous ESFT with CR to chemo, then dose
Preop RT indications: Expect R+. Preop dose is 50.4/28 fx and then no PTV2.
36–45 Gy. • PTV2: boost 10.8 Gy/6 fxs, total to 55.8 Gy
–– (2) Then at week 12, local treatment: surgery (unless at cord tolerance, then stop at 45–
or RT (try to avoid both). Surgery is preferred, 50.4 Gy). COG and IESS both have defini-
unless unresectable. tive dose of 55.8 Gy.
• “Expendable bones” are ribs, fibula, clavi- • All w concurrent chemo hold adria and
cle (lateral 4/5ths), and ilium. actinomycin.
• “Borderline resectable” bones are long • Consider boosting to 59.4 for chemo
bones and mandible. With wide resection response <50%.
and good response, 1% LR rate. If poor
response, 12% LR.
50 N. G. Zaorsky et al.
Very low risk Stage I, <2 yo, tumor <550g, no LOH Obs (on AREN 0532; note these are the risk factors on trial) N/A
FH (1p, 16q), no gain of 1p, FH
Low risk FH Stage I-II, 2 yo, tumor 550g, no VA (vinc, actinomycin) chemo x 18w. No RT. N/A
LOH (1p, 16q), no gain of 1p, FH
Standard risk Stage III, FH Flank 10.8 Gy/6 fxs at1.8 Gy w concurrent vinc 8–10
Stage III, FH, local spillage + 10.8 Gy for gross residual (total 21.6 Gy)
Stage I–II, FH, but LOH 1p, 16q Then chemo usually VAD x 24w
Stage I–II, UH (e.g. focal anaplasia),
<16 yo
FH: Whole abdomen, 10.5 Gy/7 fxs at 1.5 Gy w concurrent vinc 8–10
Stage III with SPAR (spillage in
High Risk + Boost residual by 10.5 Gy/7 fxs at1.5 Gy to 21 Gy total.
abdomen, peritoneal seeding,
(ruptured) UH: Whole abdomen, 19.8 Gy/11 fx at 1.8 Gy
ascites, rupture)
Then VAD chemo x 24w
Flank 19.8 Gy/1.8 w concurrent vinc 8–10
Stage III, UH, > 16 yo
High Risk GTV/R2: + 10.8 Gy (total 30.6 Gy)
Stage I–III Rhabdoid > 12 mo
(non-ruptured) Shield kidney after to limit dose to < 14.4 Gy
Stage I–III, diffuse anaplasia
Then VAD/C/E chemo x 24w for UH.
Special unresected LNs GTV, 19.8 Gy/11 fxs or 21 Gy/14 fxs. 8–10
scenario Lung mets Whole lung: >12 mo: 10.5 Gy/7 vs. <12 mo: 12 Gy/8 fxs. Boost With flank or
residual to 19.5 Gy or resect w concurrent vinc. after 6w chemo
If rapid CR with chemo, omit RT(on ARST0533)
Histology affects WLI dose and chemo type
Liver mets (diffuse) Whole liver 19.8 Gy/1.8. 8–10
Brain mets Per AREN 0532/0533
>16yo: 30.6/1.8 Gy WBRT, no boost
<16yo: 21.6 Gy WBRT in 12 fxs + 10.8 Gy boost (total 32.4 Gy)
Historical dose: 25.2 Gy + 10 Gy
Bone mets <16 yo: 25.2 Gy
>16 yo: 30.6 Gy
52 N. G. Zaorsky et al.
Overview Presentation
– – 450 cases/yr usually 3–4 year olds. 2.5y –– Smooth, nontender abdominal mass or swell-
for V ing, in absence of other sx
–– Fourth most common childhood cancer –– Wilms = “well”; Neuroblastoma = “not
–– Intrarenal well”
–– Favorable histo (FH, 90% of cases): –– Bilat dz 7%
Blastemal, stromal, and epithelial. Mixed his- –– Multifocal dz 12%
tology is most common (~40%) –– Renal v invasion 10%
–– Unfavorable histology (UH): anaplastic, sar- –– LN+ 20%
comatous, clear cell sarcoma of kidney –– M+ 10%
(CCSK), and rhabdoid (RTK, technically not
Wilms) DDx: Wilms, neuroblastoma, nephroma, and
–– Diffuse anaplasia (DA): nonlocalized, RCC
localized w severe dysplasia elsewhere,
anaplasia outside capsule or M+, and ana- Workup
plasia in random bx –– H&P, US abd is the first test of choice: CT/
–– SRBCT MRI, CT chest (not CXR), urine catechol-
–– Most common abdominal tumor of children amines, LFTs, CMP, ECG, and echo
–– Very radiosensitive. Typical doses –– Will be intrarenal
10.5–21 Gy • CT assesses mass size, contra involvement,
–– 90% will be resectable capsule rupture, and ascites (most likely
–– AA > white > Asian from rupture). CT for Wilms has calcs in
10% vs 70–90% of neuroblastomas have
Genetics calcs.
–– WT1 tumor suppressor gene on 11p13. –– Do not do: Bx, unless unresectable or bilat,
Associated syndromes vigorous abdominal exams. Auto-upgrade to
• WAGR syndrome (del 11p13, WT1): stage III and PORT recommended bc of
Wilms, aniridia, GU anomalies, and seeding
retardation –– Bx for: unresectable, bilateral dz. If possible,
• Denys-Drash syndrome (WT1 mutation. use post approach
WT1 is a Zn finger protein): renal dz, male –– After surgery:
pseudohermaphroditism, and Wilms • Rhabdoid: add MRI brain (15% have syn-
–– WT2 tumor suppressor gene on 11p15. chronous brain tumor, 2nd primary ATRT),
Associated syndromes BMBx
• Beckwith-Wiedemann syn (WT2 mut, • Clear cell: add MRI brain, BMBx, bone
11p15.5): IGF-2 overactivity, no active scan (25% have bone mets)
copy of CDKN1C, macrosomia, macro- –– Prognosis: DA < rhabdoid, < clear cell <
glossia, omphalocele, prominent facial fea- others
tures (earlobe pits, creases), large kidneys, –– OS5:
and hemihypertrophy –– FH > 90% (stage IV 80%)
–– 2% of Wilms is familial. –– FA/DA 80%
–– 10% of Wilms is assoc with congenital abn. –– CCSK 75%
–– Del ch22q, LOH 1p and LOH 16q, gain 1q –– RTK 30%
have poorer RFS and OS.
–– Clear cell and Rhabdoid are not actually Treatment Paradigms
Wilms tumors and are treated differently. –– Biopsy is avoided because it may cause tumor
–– FH: fathers who are welders (RR 5.3); moth- spill.
ers who use hair dyes (RR 3.6). –– NWTS: The USA. Surgery, then adjuvant tx.
Pediatrics Cancersr 53
• Note: USA staging typically postop must omit WLI. Slow responders get WLI and dif-
assess regional LNs, contra kidney, perito- ferent chemo.
neum, liver, and renal vein/IVC
involvement Chemo Types
–– SIOP: Europe. Preop chemo, and then resect. –– EE4A (VA): vincristine and dactinomycin.
And then assess adjuvant tx. Consider if stages Used in FH
IV and V or if would likely be stage III (R+/ –– DD4A (VAD): EE4A + doxorubicin. Used
spill). Gain of 1q has worse 5y EFS 88% vs when inc risk (e.g., FH w lung mets, UH)
75% and OS, 94% vs 88%. –– M: DD4A + cytoxan+etoposide
–– UKW3: RCT of NWTS vs SIOP approaches. –– CAVE for CCSK: cytoxan, adriamycin, vin-
SIOP has imp stage dist, 20% reduced use of cristine, and etoposide
RT, doxorubicin. Similar EFS and OS. –– CEC for RTK: carbo, etoposide, and
cytoxan
What Do You Want to Know from Surgery?
–– Extent of resection RT General Rules
–– Nodal status –– All patients get an overall stage and a local
–– Extent outside kidney stage.
–– Spillage (focal or diffuse), cytology, and –– All get postop RT except FH I/II and CCSK I
implants (review by central pathology).
–– Pathology: FH versus FA/DA/CCSK/RTK –– RT to primary site: for all pts w unfavorable
–– 1p, 16q, del ch22q status histology (e.g., focal anaplasia).
–– Review by central pathology –– RT should be initiated by day 10 concurrent
with start of chemo. Hold adria during RT.
Trials –– Treat abdominal stage when pt has stage IV dz
–– NWTS 1: showed no RT needed for group 1, (i.e., if stage IV by pulm mets, but abdomen is
<2 yo if given chemo. RT starts <9d after sur- stage II, and FH do not tx abdomen).
gery. FH 2y CSM 7% vs 44% for UH. V + A –– When to WART: (“SPAR”) tumor spill, perito-
better than V or A alone. WAI is not needed if neal mets, Ascites/gross dz, and preop rupture.
local spill; just use flank. Note that Bx not always = spill.
–– NWTS 2: showed RT not needed for all group –– When to use >10 Gy: any residual dz (e.g.,
1. Adding Adriamycin can improve OS for unresected, mets).
group 2–4. –– Always RT for anaplasia. 10.8 Gy for focal
–– NWTS 3: showed RT is not needed for stage II anaplasia and 19.8 Gy for stage III–IV diffuse
if chemo given. 10 Gy for stage III if anaplasia.
Adriamycin used. Must start RT w/in 9 days, –– RTK stage I–IV all need RT, 19.8 Gy; some
at latest 14d. centers use higher doses.
–– NWTS 4: showed pulse-intensive chemo (6 m –– Only stage III–IV w FH require RT.
vs 15 m) less toxic than standard
–– NWTS 5: stage I, FH and <550 g tumor can be Bilateral Wilms’ Approach
observed after surgery (2 yr DFS 87%, OS –– Bx and stage each site.
100%). LOH 1p or 16q assoc with relapse and –– Ex-lap w bilateral renal bx.
death. For UH, etoposide improved OS. Vinc/ –– Preop chemo by worst histo.
Adr/cyclophos/etoposide improved outcomes –– Then, either bilatal partial nephrectomy or
in II-IV w DA. additional chemo if unresectable. Once it is
–– AREN 0533: higher risk FH WT. Resection, resectable, do bilat partial nephrectomy and
local RT, chemo, RT to non-lung mets. If rapid then adjuvant chemo and RT by most aggres-
responder (CR after 2 cycles DD4A), and then sive dz. If it never becomes unresectable,
preop RT 12–16 Gy reassess.
54 N. G. Zaorsky et al.
Lung volume
T11
Remaining
kidney
Pre-op
Remaining GTV
kidney
L4 L1
Remaining
kidney
INRGSS clinical staging (preop). New! –– Suprarenal (vs intrarenal for Wilms).
Stage L1 Localized tumor –– Homer-Wright rosettes.
Stage L2 Locally invasive by defined criteria –– Stains NSE+, synaptophysin+, and
Stage M Metastatic disease except MS neurofilament+.
Stage Metastatic to only the skin/liver/marrow –– Shimada classification: based on stroma, age,
MS and <18 m
diff, mitoses, and nodular/diffuse (SAD
INSS OLD staging (still used for risk groups)
MiNd).
I GTR (R0/1); Can include adherent LN –– Poorer prognosis: ↑stage, ↑age, n-myc amp,
II STR (R2) or ipsi LN diploid, and Shimada (SANDS).
III Unresectable or crosses midline (dz or LNs) –– Also poorer prognosis: LOH 1p or
IV Distant mets: distant LN or the bone, liver skin 11q,↑telomerase.
IVS < 1 yo, stage I–II, w/ BM < 10%, the liver, or skin –– Screening: Tumors can spontaneously
regress, so screening is not helpful (Japan,
Overview Canada, Austria trial); only recommend if
–– 650 cases/yr, the most common extracranial familial.
childhood tumor and most common cancer for –– Location: adrenal (35–40%), post-
children <18 m. mediastinum (20%), paraspinal ganglion (25–
–– Median dx is 17–22 m (Wilms is 3–4 yo). 32%), and the pelvis (5%). Thus, ~65% are
–– Represents ~8% of childhood cancers. abdominal.
–– Primitive neural crest cells (usually calcified; –– Associations: NF1, Hirschsprung, central
Wilms isn’t). hypovent, familial ALK mutations, Turner,
–– SRBCT. and material opiates.
Langerhans Cell Histiocytosis (LCH) lesions, DI, and hemangiomas; lytic bone
lesions (thus, negative BS). Good prognosis. >
Risk group Definition 2 yo
Low 1 lesion in bone, LN, GI, CNS
High <2 yo w organ dysfunction; liver, spleen, BM
• Litterer-Siwe disease: Hepatosplenomegaly,
lymphadenopathy, pulmonary lesions, and
bone lesions. Widespread seborrheic rash.
Background
<2yo
• 1200 cases/y
• Manifestation: the bones in children and the
• 1–3 yo
lungs in adults. In children, usually bone pain,
• M > F
soft tissue mass, the lung, or oral mucous
• Dysregulation of monocular cell line
membrane involvement
• Previously called eosinophilic granuloma and
• Workup: HP, labs, skeletal survey (appears
histiocytosis X
“punched out”), NOT BS, biopsy
• LCs are SPCs to lymphocytes in the skin,
• Histology: Birbeck granules on EM. CD1a,
mucosa, and spleen.
S100, and CD207 (Langerin)
• Hand-Schuller-Christian dz: proliferation
of histiocytes causing exophthalmos, skull
Treatment
Planning
DI: 15 Gy to pituitary/hypothalamus within 14 d
Bone: 5–10 Gy (e.g. 3 Gy x 3 fractions w small margin) or 1.5 Gy x 8 fractions
Adult: 15–24 Gy/2 Gy fraction
Pediatrics Cancersr 63
–– The entire tx takes ~1y. ACNS 0331 RT takes –– Adult medullo. Adults cannot tolerate chemo
6w. Concurrent chemo for 6w w vcr, then 9c like peds patients. Further, since adult is fully
of chemo q6w. Total = 55w tx grown, there is less concern about long-term
toxicity. Some argue tx like peds, others advo-
Dose Constraints cate RT alone.
–– Cochlea: V30 < 50%, max 35 Gy –– Standard risk cannot get chemo: treat like
high risk, 36 Gy CSI + 19.8 Gy boost.
Special Considerations (Mostly High Risk)
–– M2/Brain/thecal mets 54–55.8 Gy. Follow-up
–– Spinal mets 45 Gy if above terminus, 50.4 Gy –– Second malignancy rate is 0.4% per year
if below terminus. per ACNS 0332. (from ACNS 9961).
–– Diffuse spinal disease 39.6 Gy.
–– Pineoblastoma: Treat like a high-risk medullo
(CSI to 36, boost to 54 Gy).
66 N. G. Zaorsky et al.
COG Approach
COG approach
Risk group RT Trials
Age Surgery Chemo CSI Boost SJMB12 (approximate groups;
(y) note supratentorial PNET
excluded):
Low >3 MSR 15 Gy Tumor bed Stratum W1 4 cycles cyclo, cis,
to 54 Gy (WNT): 15 Gy VCR.
CSI, 51 Gy boost
Avg/Std Risk >3 MSR 23.4 Gy Tumor bed ACNS 0331 reported: RT to Stratum S1 4 cycles cyclo, cis,
/13 fx to 54 Gy tumor bed > posterior fossa. Still (SHH) 23.4 Gy VCR. Then
CTV = need 23.4 Gy CSI CSI, 54 Gy maintenance
GTV + 1.5 boost. vismodegib x 12
If chemo
cm. months
not
PTV =
given,
RT with vincristine 1.5 mg/m2, start 4 weeks after surg, then qw x 7c. Then adjuvant cis.
boost.
Stratum N2 AABAABB (7
Note: concurrent chemo not used on SJMB12
Gy boost.
High Risk >3 MSR. If 36 Gy / Posterior 3-21 yo. Rando to (1) concurrent Stratum S2 (SHH 4 cycles cyclo, cis,
unresectable, 20 fx fossa to VCR + RT vs (2) Concurrent + MYC): 36-39.6 VCR. Then
proceed w 54-55.8 VCR + carbo + RT. Then 2nd Gy CSI, 54 Gy maintenance
CRT rando for both for chemo alone vs boost. vismodegib x 12
chemo + isotretinoin months
Stratum N3 AABAABB (7
(Non-SHH, Non- cycles)
WNT, MYC A: cyclo, cis, VCR
amp; high risk): B: pemetrexed,
36–39.6 Gy CSI, gemcitabine
54 Gy boost.
Pineoblastoma Pineocytoma
–– Treat like a high-risk medulloblastoma patient. –– Treat like LGG.
–– CSI to 36 Gy with boost to 54 Gy, with CVP –– If GTR: observe.
chemo. –– If < GTR: RT to 54 Gy.
Pediatrics Cancersr 67
• If STR then push for induction chemo, 7w –– If Spinal met: RT for incomplete resection or
of vcr, carbo cpm OR vcr, carbo, etop. anaplastic histology
Then re-resection so there is a GTR or (a) Two vert bodies above/below to 45 Gy.
NTR. On ACNS 0831, pts with STR do not (b) Boost to 50.4–59.4 Gy if no cord in field.
receive RT. –– Ependymoblastoma
• If GTR and high-risk features (e.g., (c) Treat like high-risk medulloblastoma
G2/3; adult patient), then adjuvant RT. Note
that RT usually local. Simulation/Planning
(i) > 3 yo: Resection and adj RT. R1 or G3 1. Local therapy (volumes from ACNS0831)
above the cord to 54, then comes (a) CTV1 = residual & cavity +1 cm
down (off brainstem, chiasm, cord) CTV2 = residual tumor and resection bed
to 59.4 Gy. above the cord
(ii) < 3 yo: Need to delay RT. Resection PTV1 = CTV1 + 0.3–0.5 cm
and chemo (cisplatin-cyclophos- PTV2 = CTV2 + 0.3–0.5 cm
etoposide) Can give 2 cycles then re- (b) Dose: Treat PTV1 to 54 Gy/1.8 Gy fx.
resection vs RT. If used, RT should be Boost PTV2 to 59.4 Gy if >18 m,
to 54 Gy. However, if pt < 18 m w GTR, then no
• Indications for CSI (ideally after 3 yo): focal boost.
(iii) M+ (e.g. +CSF, MRI positive) (c) No RCT for 54 vs 59.4. However, brain-
(iv) Grade IV (treat like medullo) stem toxicity can be fatal. Consider treat-
• ACNS0831. Supratent ependymoma. ing to only 54 Gy if close proximity to
Phase III study of: brainstem.
(1) Obs after GTR, differentiated supratent 2. If delivering CSI
ependymoma (a) Dose is 36 Gy
(2) After STR, induction chemo cinv carbo (b) If gross spine dz, would boost to 45 Gy
cyclosphos, and etopo, second-look
surg, then RT, then chemo Follow-Up
(3) All others get RT, then rando +/− 1. For >10 yrs, late recurrences happen.
chemo. Supratent ependymomas get 2. Craniospinal MRI Q3-6 m then Q1yr.
59.4/1.8. GTV is any residual
tumor+bed. Pre- and postop imaging Risk of brainstem toxicity: chemo, age < 3
used to define GTV. CTV = GTV + yo, ATRT, and multiple surgeries
0.5 cm. PTV = CTV + 3–5 mm (3 if
IGRT)
70 N. G. Zaorsky et al.
Hemangioblastoma Treatment
–– Surgery w MSR preferred
Background • No need to remove entire cyst if R0.
–– 20–50yos • 15% M/M
–– Arises in cerebellum • 50–80% LC
–– VHL association –– SRS is alternative, typically reserved for LR:
–– WHO I • 15–21 Gy to 50% IDL
–– Made of endothelial stem cells, “foamy cells” • 90% LC at 2y, 75% at 5y
that make “clear cell” morphology –– EBRT is for multiple tumors, after STR for
–– Single lesion = sporadic, older recurrence, large (>3 cm), lesions in eloquent
–– Multiple lesions = familiar, younger brain regions
• 50–55 Gy/2
VHL Hemangioblastomas
• Young age, mean 29, 50% in cord, 40% cere-
bellum, 10% brainstem
• PCV bc of EPO production by tumor
• Have better prognosis after EBRT
Workup
–– MRI: eccentric, peripheral cystic mass, and
70%. Intensely enhancing
Pediatrics Cancersr 75
Chemo Options
–– Carbo 175 mg/m2 + vinc 1.5 mg/m2 induc-
tion, then maintenance x 12c
–– 6-thioguanine, procarbazine, dibromodulcitol,
CCNU, and vinc
–– Avastin + irino: recurrence
Pediatrics Cancersr 77
Chemo
–– No consensus on what is best.
–– PEI =cisplatin, etoposide, ifosfamide.
–– PEV = cisplatin, etoposide, vincristine.
–– VCR = vincristine.
78 N. G. Zaorsky et al.
Abstract
This chapter discusses the general manage-
ment of patients with central nervous system
tumors, with special focus on principles that
guide radiotherapy management. Several key
components of radiotherapy and radiosurgery
care are discussed.
@NicholasZaorsky
@DanTrifMD
Brain pathology
Nuclear Atypia GBM
Mitotic index
Endothelial proliferation
Necrosis
Rosenthal fibers JPA
Psammoma bodies, whorls Meningioma
Verocay body Schwannoma
Schiller-Duval bodies Yolk sac
Fried egg Oligodendroglioma
Pseudorosettes Ependymoma: cystic, cerebellar
Homer-Wright rosettes Medulloblastoma, neuroblastoma, PNET, +/−pineoblastoma
Flexner-Wintersteiner rosettes Retinoblastoma, +/−pineoblastoma
common are nerve sheath tumors (schwanno- with different dose gradients, for example,
mas, neurofibromas) and meningiomas. where isocenters have been inappropriately
–– Intradural intramedullary spinal cord centered on the edge of the target volume.
tumors are usually primary tumors. –– Conformity index (CI): (Rx isodose vol-
Subtypes: ependymoma (90%), astrocytoma, ume)/(PTV).
hemangioblastoma, subependymoma, and • Preferred <1.2 in lung SBRT
ganglioglioma. • Controversial use in CNS
–– Gradient index (GI): (volume of half the pre- –– Central hypodensity, ring enhancement, high
scription isodose)/(volume of the prescription edema to enhancing tumor ratio, T1/T2 mis-
isodose). match, low PET avidity, ↓ choline peak, ↑ lac-
• Preferred to be ~1.0 in CNS. tate peak, and ↑ lipid peak occur at >6 m post
• For example, for a plan normalized to the RT. Necrosis may have decreased blood flow
50% IDL, it is 25% isodose volume/50% on perfusion series (e.g., arterial spin labeling
isodose volume. GI will differentiate [ASL]), while some tumors (e.g., GBM,
between plans of similar conformity but metastases) will have increased blood flow.
90 S. K. Nath et al.
TERT mutations common ATRX loss common MGMT promoter meth variable Correlate with clinical, imaging, histological, and
CIC and FUBP1 mutations variable P53 mutations common TERT mutations common molecular features.
MGMT promoter meth common MGMT promoter meth common Chromosome 10 loss common
PTEN mutation variable H3 K27M mutations (diffuse midline glioma)
EGFR amplification variable BRAF mutations (some PXAs, gangliogliomas,
pylocytic astros; also seen in epithelioid GBM)
The classification of gliomas has evolved signifi- mutated oligodendrogliomas and astrocytomas
cantly in the past decade. Historically, WHO will typically retain their characteristic molecular
grades I and II gliomas were broadly classified driver mutations after transformation to higher-
and treated as “low-grade” tumors, and WHO grade tumors, suggesting that grade II and III oli-
grades III and IV tumors were classified and godendrogliomas represent different points in the
treated as “high-grade” tumors based on light phenotypic evolution of tumors from a common
microscopy features. In the 2016 edition of the molecular lineage and the same could be said of
WHO classification of tumors of the CNS, glio- IDH-mutated astrocytomas of grades II, III, and
mas are now classified according to both histo- IV (i.e., secondary GBMs).
pathologic and defining molecular features. Primary GBMs, which are more common
Some of the most important updates are related (~90%) than secondary GBMs (~10%) and tend
to the recognition of IDH mutations as early, to occur in older adults, typically do not have
stable driver mutations in the majority of grades IDH mutations and are instead driven by other
II and III gliomas, as well as a characteristic fea- characteristic molecular features, including but
ture of most secondary GBMs (i.e., those GBMs not limited to TERT mutations, chromosome 10
that arise from lower-grade tumors). IDH- loss, PTEN mutations, and EGFR amplification.
mutated tumors that subsequently develop down- IDH wild-type astrocytomas often exhibit molec-
stream codeletion of chromosomal arms 1p and ular profiles and clinical behaviors that are more
19q are now classified as oligodendroglial similar to primary GBMs than to IDH-mutated
tumors by definition, whereas IDH-mutated astrocytomas and are sometimes referred to as
tumors without 1p/19q codeletion are classified “molecular GBMs.”
as astrocytic tumors and commonly have down- In 2018, the majority of the available clinical
stream ATRX loss and p53 mutations (Eckel- data is based on older WHO classification sys-
Passow 2005, Cancer Genome Atlas Research tems, and guideline-endorsed treatment para-
Atlas 2015) [3, 4]. digms are driven more by histologic grade than
The assignment of WHO grades II, III, and IV molecular profile (see below). However, it is
is still dependent on histopathologic features clear that future risk stratification and treatment
including atypia, mitoses, microvascular prolif- algorithms will be increasingly based on genomic
eration, and necrosis. However, both IDH- alterations.
Central Nervous System Cancers 91
–– EORTC 22844 Believers (dose-esc) trial PFS and OS in Major LGG Randomized
(Karim, IJROBP 1996). 379 patients with Trials
LGG randomized to 45 Gy/25 Fx vs
PFS OS
59.4 Gy/33 Fx. No difference in OS.
Trial Treatment (years) (years)
–– Intergroup (dose-esc) trial (Shaw, JCO Non- Obs (delayed RT) 3.4 7.2
2002): 203 patients randomized to 50.4 Gy/28 Believers
Fx vs 64.8 Gy/36 Fx. No difference in OS. RT (54 Gy) 5.3 7.4
High-grade neurotox worse with high-dose Believers RT (45 or 59.4 Gy) 4 to 5 6 to 7
RT. Ninety-two percent of failures were Intergroup RT (50.4 or 64.8 Gy) 5.5 9.3
EORTC RT (50.4 Gy) 3.8 –
infield. OS better with age <40, oligo histo,
22033
and smaller size. TMZ 3.2 –
RTOG 9802 RT (54 Gy) 4.0 7.8
RT +/– Chemotherapy for LGGs Combined RT (54 Gy) 10.4 13.3
–– RTOG 9802 (Buckner NEJM 2016). High- and then PCV x6c
risk LGG (defined as age ≥ 40 or STR), 251
NEW NCCN high risk OLD high risk (SATANIC)
patients randomized to 54 Gy/30 Fx +/− PCV
>40 yo or STR Size ≥6 cm
chemo x 6 cycles. 43% oligodendro. With
Age >40yo
12-yr follow-up, PCV dramatically improved Tumor crossing midline
OS (median 13.3 vs 7.8 yrs) and PFS (10.4 vs Astrocytoma (pure)
4.0 yrs). Curves separate at 4y. Neuro deficit preop
–– RTOG 0424 (Fisher 2015): Single-arm phase IDH1/2 non-mutated as high
II. 129 patients with high-risk LGG, with 3 or risk
1p/19q non-Codeleted
more risk factors (age ≥ 40, astro, bi-
hemispherical tumor, ≥6 cm, or preop neuro
function status of >1), were treated with RT Treatment Paradigms in 2018
(54 Gy/ 30 Fx) plus concurrent and adjuvant –– Cranial LGGs need max safe resection first.
TMZ. Three-year OS of 73% improved vs his- Per 2017 NCCN, if low risk (GTR and age <40
torical control of 54%. per RTOG 9802), may follow with close
–– EORTC 22033-26033 (Baumert, Lancet observation.
Oncol 2016): Randomized single-modality –– If high-risk (STR or age >40y), then adjuvant
comparison of RT alone (50.4 Gy) vs TMZ RT + chemo. PCV is category-1 based on
alone (max 12 cycles) in high-risk supratent RTOG 9802, but many centers use TMZ.
LGG, with at least one of these features: –– The contemporary NCCN stratification of high
age >40, progressive dz, size >5 cm, cross vs low risk provides a useful backbone for
midline, and neuro sx. No diff in median LGG management; however, we would caution
PFS, 46 months for RT vs 39 months for the conceptual overreliance on the high- vs
TMZ. OS results still maturing and not low-risk inclusion criteria from a single trial
reported in 2016 publication. Results should (i.e., RTOG 9802). Additional clinical risk fac-
be taken in context of the RTOG 98-02 tors have been identified and used in other trials
results showing best outcomes with (e.g., tumor size, histology, symptoms, crossing
RT + chemotherapy. midline, progressive, etc.). Molecular-based
94 S. K. Nath et al.
stratifications (IDH, 1p/19q) are also likely to –– Two dedicated randomized RT dose-esc trials
gain increasing importance in future trials. from the pre-chemo era (EORTC 22844 and
IDH-negative astros, in particular, may exhibit Intergroup) do not support an advantage of RT
natural histories similar to primary GBMs doses above 45–50.4 Gy. The RTOG 9802
(Cancer Genome Atlas Research Network, used 54 Gy/30 fx. EORTC used 50.4 Gy/28
NEJM 2015). fx. Consequently, 2017 NCCN acknowledges
–– For spinal LGGs, the adj RT paradigm is simi- doses from 45–54 Gy.
lar, although the role of chemotherapy is cur- –– New CODEL trial incorporates boost for GRD
rently undefined. Surgery with max safe to 59.4 Gy.
resection first. If GTR, role of adj RT is con-
troversial and may be individualized. If STR Simulation/Planning – Spine
or less, adj RT is usually advocated because –– Supine, vacloc, aquaplast mask if above T4.
failure is typically local. –– MRI should be used to define the GTV.
–– Classically, fields targeted 2 VBs above and
Simulation/Planning: Brain below the myelogram-defined tumor. With
–– Supine, aquaplast, and fuse post-op MRI. Fusion modern MRI-based planning, a CTV of
of preop MRI may be useful for reference. GTV + 0.5–1.0 cm is reasonable.
–– GTV = post-op cavity + any T1 post-contrast –– PTV = 3–5 mm with daily IGRT.
and T2 FLAIR abnormality. Most LGGs will –– Dose: 45–50.4 Gy are reasonable for LGG;
not have T1 post-contrast enhancement. extrapolated from cranial disease; higher
–– CTV = 0.5–1.0 cm exp, cropped at anatomic doses can be considered based on size, loca-
boundaries (e.g., falx, tentorium, bone, cis- tion, tumor grade, and risks of radiation-
terns). CTVs should be allowed to cross white induced myelopathy vs myelopathy from
matter tracts commonly including corpus cal- tumor regrowth.
losum but where appropriate consideration
should also be given to cerebral/cerebellar Follow-Up
peduncles and ant/post commissures. –– NCCN advocates MRI Q6m for 5 years and
–– PTV = 2–5 mm with daily IGRT. *PTVs should then annually.
be based on institutional technical standards. –– Lifelong surveillance recommended.
Central Nervous System Cancers 95
(>65 yrs). Resection associated with improved Strategies that Have not Improved on
OS (5.7 vs 2.8 mo, p = 0.035). 60 Gy/30 Fx
–– Brown 2016: Meta of >41,000 pts; improved –– SRS boost (e.g., RTOG 9305, RTOG 0023)
OS with GTR >STR and STR >biopsy alone. –– Hyper Fx (e.g., RTOG 9006, RTOG 8302)
–– Lacroix 2001: n = 416. Ninety-eight EOR –– Brachytherapy (e.g., BTCG NIH 8701)
was cut off and contributed to all or none sur-
gical philosophy. Elderly: RT vs Best Supportive Care
–– Sanai 2011: n = 500. EOR as low as 78% ben- –– Keime-Guibert 2007: 81 patients >70 yrs.
eficial. Stepwise improvement in OS for EOR RCT of 50.4 Gy vs best supportive care. No
thresholds of 95%, 98%, and 100%. chemo. Trial stopped at first interim analysis
–– Chaichana 2014: n = 259. EOR at >70% with superior OS with RT, 7.3 vs 4.3 mo.
beneficial.
Elderly/Poor KPS: Hypofractionation,
BCNU/Carmustine (Gliadel) Wafers in the No TMZ
Surgical Cavity –– Roa 2004 (“Roa 1”): 100 elderly (>60 yo)
–– Westphal 2006: RCT (N = 240) of newly GBM patients. Surgery, then randomize
diagnosed HGG. Improved OS with wafers, 60 Gy/30 Fx vs hypofractionated RT 40 Gy/15
13.8 vs 11.6 mo (p = 0.017). Fx. No chemo. No difference in median OS:
–– Valtonen 1997: RCT (N = 32) of newly diag- 5.1 vs 5.6 mo. No change in average KPS score
nosed HGG. Improved OS with wafers, 53 vs or KPS over time.
40 weeks (p = 0.008) for GBM. –– Roa 2015 (“Roa 2”): 98 with GBM. Three
–– Brem 1995: RCT (N = 222) of recurrent patient types included: frail = age 50+ and KPS
HGG. Improved OS with wafers, 31 vs 50–70, elderly = age 65+ and KPS 80–100, and
23 weeks (p = 0.006). elderly and frail. Note that 40/15 is 70% BED
–– 2017 NCCN acknowledges wafers in cavity at of 60 Gy/2. Randomized to 25 Gy/5 Fx vs
resection as an option for both newly diagnosed 40 Gy/15 Fx. No chemo. No diff in OS, PFS, or
and recurrent HGGs, but note that this can be QOL. Dose constraint is the max dose (40 Gy).
an exclusion criterion for some clinical trials.
Elderly/Poor KPS: Hypofractionation
Surgery +/– RT in the Pre-TMZ Era and + TMZ
–– Laperriere 2002. Systematic review and pooled –– Nordic trial (Malmstrom 2012): Single-
analysis of six RCTs from the pre- TMZ era modality comparisons – TMZ alone vs hypo-
(Shapiro 1976, Walker 1978, Anderson 1978, frac RT 34/10 alone vs standard RT 60/30
Walker 1980, Kristiansen 1981, Sandberg- alone for GBM in >60yo. OS outcomes with
Wollheim 1991). OS favored RT (HR 0.81, TMZ alone (8.3 mo) similar to 34/10 (7.5 mo)
p < 0.0001). These historic trials used doses from and superior to 60/30 (6 mo). Standard RT
45 to 60 Gy and most involved whole-brain RT. 60/30 patients were less likely to complete
treatment. MGMT+ patients did best with
WBRT vs Limited-Field RT TMZ alone. In MGMT-negative, RT alone
–– Shapiro 1989: Trial included a randomization was superior to TMZ alone. Supports use of
of WBRT 60 Gy vs WBRT 43 Gy and then 34/10 over 60/30 for elderly.
cone down to 60 Gy. No differences in OS. –– NCI Canada/EORTC/TROG (Perry 2017):
Elderly pts ≥70 yrs, randomized to hypofrac-
RT Dose tionated RT (40 Gy/15 Fx) +/− conc/adj
–– RTOG 74-01 (Nelson 1988): No benefit to TMZ. Improved OS (9.3 vs 7.6 mo) with TMZ
70 Gy over 60 Gy for GBM overall and in MGMT methy (p < 0.001) and
–– MRC (Bleehen 1991): 474 pts. 45/20 vs unmeth patients (p = 0.055). This trial effec-
60/30. 60 Gy improved OS (12 vs 9 mo) tively ended the preceding debate about the
Central Nervous System Cancers 97
–– Call progression within 3 months of RT only –– RTOG 1205 (ongoing): RCT of bevacizumab
if new enhancement is beyond 80% IDL alone vs bevacizumab plus reRT. RT is 35
–– Unequivocal pathological evidence of viable Gy/10 Fx to cavity and T1 post-contrast
tumor abnormality with tight margins.
General Workup
–– 4% of primary CNS tumors. –– H&P, CBC, CMP, LDH, HIV
–– In immunocompetent pts, most dx between 45 –– Stereotactic brain biopsy; hold steroids until
and 65 years of age. path bc of 40% response rate
–– The primary risk factor is immunodeficiency: –– CSF cytology if safe
HIV, iatrogenic immune deficiency, –– Slit lamp exam to rule out ocular involvement
autoimmune disorders, and genetic immune –– MRI +/−spine if spinal canal involvement
deficiency syndromes. suspected. MRI has classic “cotton” wool
–– EBV+ in 60% of immunocompetent cases. appearance with fuzzy borders
–– Gain of chr12 → ↑MDM2 → ↓p53. –– CT CAP or PET-CT to rule out systemic NHL
–– Most (>95%) have a negative systemic lym- –– Testicular ultrasound for men >60 years
phoma workup. If lymphoma found outside –– Consider BM biopsy
CNS, it is NHL with CNS involvement. –– SPECT if immunocompromised
–– The majority of PCNLS are DLBCL. –– Vitrectomy for diagnosis if ocular lymphoma
–– There are normally no B cells in the CNS. suspected
–– Clinical prognostic groups (Ferreri JCO 2003)
based on age, PS, serum LDH, CSF protein Chemo +/– RT
concentration, and deep brain involvement: –– G-PCNSL-SG-1 (Thiel 2010): Phase 3 non-
age >60, ECOG >1, elevated LDH, elevated inferiority trial. 551 enrolled (318 treated per
CSF protein, deep brain involvement. protocol), immunocompetent patients ran-
–– “ABCDE” domized to high-dose MTX (with or without
–– Abrey/MSKCC, 2006 nomogram: ifosfamide) +/− WBRT (45 Gy/1.5 Gy Fx
• RPA1: Age <50. MST 8.5 y daily). No diff in OS (but failed to reach non-
• RPA2: Age >50, KPS >70: MST 3.2 y inferiority margin). WBRT assoc with
• RPA3: Age >50, KPS <70: MST 1.1 y improved PFS and more neurotox.
–– RTOG 1114 (ongoing): R-MVP and consoli-
dative Ara-C +/− low-dose WBRT (23.4 Gy)
Imaging
–– T1p homogenously enhancing mass, pre-con Chemo + Low-Dose WBRT
T1 hypointense, T2 iso to hyperintense. –– Morris 2013. Multicenter phase II, 52 patients.
–– Majority (60–70%) are solitary lesions in R-MVP (MTX, procarbazine, VCR,
immunocompetent pts, with higher rates of ritux) + reduced dose RT to 23.4 Gy in the case
multifocality in the immunocompromised. of CR or 45 Gy for PR, then consolidation cyta-
–– Lesions tend to be nonhemorrhagic, periven- rabine. 2-year PFS 77%, median PFS 7.7 years.
tricular masses, in deep white matter. 3-year OS 87%. Median OS not reached.
Central Nervous System Cancers 101
General
History
–– 34% of all primary tumors.
–– Many are asymptomatic: HA, personality
–– From arachnoid cap cells in arachnoid villi.
changes, CN deficits, paresis, paresthesias,
–– Approximately 2–3% of the population may
and seizures.
have meningiomas.
–– If suspecting NF-2, check for café au lait
–– Second most common primary brain tumor (at
spots.
15–30%), and the incidence increases with
age.
Workup
–– Two percent of the population may have
–– Make the diagnosis radiographically. If not
meningiomas (based on autopsy series),
possible, then consider biopsy or octreotide
though 90% are benign.
scan.
–– Median age 65, rare in children (increased in
–– CT head thin cuts w/ and w/o contrast.
NF2 or after early RT exposure).
–– MRI brain w/ and w/o contrast.
–– Female:male ratio 2–3:1 for grade I s but less
pronounced in grades II–III s.
MRI
–– Risk factors: RT, hormone therapy, NF2,
–– Extra-axial, dural-based mass.
schwannomatosis, and MEN1 syndrome.
–– Classically pre-contrast T1 isointense or
–– Somatic NF2 mutations may be present in up
hypointense, T2 isointense or hyperintense.
to 1/2 of sporadic meningiomas.
–– Strong, homogenous T1 post-contrast
–– Psammoma bodies and calcifications and ker-
enhancing.
atin whorls (Starry Night by Van Gogh sign).
–– Frequently have adjacent dural thickening that
–– Factors predicting slow progression: calcifica-
tapers peripherally (“dural tail”).
tions, hypointense on T1 MRI, old age, and no
–– Features which may suggest a grade II–III
growth on serial scans.
meningioma: brain invasion, brain edema,
intratumoral cystic change, extension through
WHO Rate Name
grade
Mitosis Cellularity
bone, hyperostosis or adjacent bone destruc-
I >80% Benign,
Angioblastic,
<4 /10 HPF tion, and elevated blood volume.
All others –– No imaging feature is pathognomonic for
II 7–15% Atypical ≥4 /10 HPF Increased cellularity
Clear cell Brain invasion Small cells, high N:C high-grade meningiomas in the absence of
Chordoid or ≥3 of the Prominent nucleoli
OR benign w/ following: Patternless or sheetlike growth pathology.
brain Foci necrosis
invasion
III 4% Anaplastic ≥20 /10 HPF
Rhabdoid And/or:
Resemble carcinoma, sarcoma,
melanoma
CT Head Thin Cuts w/ and w/o Contrast
Papillary Loss of usual patterns –– Majority contrast enhancing, peritumoral
Infiltration of brain
Mitoses + atypical forms edema in 60%, bony changes (hyperostosis) in
Multifocal foci necrosis
15–20%; 15% have calcification.
104 S. K. Nath et al.
–– Reports of SRS for grade II s are primarily in –– Growth after surgery:
the setting of STR or recurrence, with widely (a) RT and/or re-resection depending on
ranging control rates from 0 to 90% at 2 yrs, scenario
with most between 50 and 80% (Rogers 2015). –– RT technique:
–– Symptomatic edema is reported more com- (a) SRS generally preferred for suitable lesions
monly after single-fraction SRS for larger due to similar efficacy to conventional RT,
meningiomas and those in parasagittal/para- favorable toxicity, and convenience.
falcine or convexity locations (Sheehan 2015; (b) SRS dose is typically 12–16 Gy for grade I.
Patil 2008; Girvigian 2008). Some authors (c) Conventionally RT is typically 45–55 Gy
have advocated fractionated SRS or for grade I.
conventional RT for such cases.
–– Multi-fraction SRS (frequently in the range Grade II
20–25 Gy delivered in 3–5 fx) has been associ- –– Max safe resection:
ated with encouraging LC and may represent (a) GTR: Role of adj RT is controversial. Either
an attractive option for tumors located near RT or close observation may be acceptable:
critical structures such the optic nerves/chiasm (i) Ongoing NRG BN003 and ROAM/
or brainstem (Marchetti 2016; Navarria 2015). EORTC 1308: Grade II s post-GTR
Overall, the available follow-up durations in randomized to RT (59.4 or 60 Gy) vs
series of multi-fraction SRS are shorter than close observation
the single-fraction SRS literature. (b) STR: Post-op RT is standard.
–– RT technique:
(a) Conventionally fractionated RT is typi-
Treatment Paradigm
cally standard, 54–60 Gy.
Grade I
(b) SRS can be considered in select cases.
–– Small (typically <2–3 cm but no strict size cri-
teria), asymptomatic:
Grade III
(a) Up-front observation preferred. Growth
–– Max safe resection - >adj RT (59.4–60 Gy) is
rate = 4 mm/y
standard.
–– Symptomatic, large (>~3 cm), or growing:
(a) Max safe resection preferred: Pediatric Meningioma
(i) GTR: Observation preferred –– Very rare.
(ii) STR: Observation generally preferred –– Manage primarily w/ surgery.
for grade I. If close to critical struc- –– Consider adjuvant RT if anaplastic, R+, brain
tures, consider RT invasion, and benign meningioma at a site
(b) Unresectable: Radiation alone. FSRT pre- where risks of recurrence outweigh risks of
ferred. SRS cat 2b RT, progressive, or unresectable.
Low GTR G1 (Simpson I–III) observe 92/93/98 86/87/98 RTOG 05-39: MRI
STR G1(Simpson IV–V) q6m for 3y, then q12m
for 10y
Intermed GTR G2 GTV=tumor bed and enhancement 94/96/96 84/86/96 RTOG 05-39: MRI at 3,
Recurrent G1 CTV54=GTV + 1cm 6, 12 months, then
PTV54=CTV54 + 3-5mm every 6-12m for 5y,
then MRI q1-3y.
High GTR G3 CTV54=GTV + 2cm 59/69/79 – RTOG 05-39: MRI at 3
Recurrent G2 PTV54=CTV54+ 3–5mm m, then every q6m for
STR G2 Then followed by 6 Gy boost 3y,then q12m for 10 y.
CTV60 = GTV + 1cm
PTV60 = CTV54 + 3–5mm
106 S. K. Nath et al.
Optic Nerve Sheath Meningioma (b) CTV = 3–5 mm expansion on GTV for
grade I–II meningiomas:
Literature (i) CTVs can be shaved off anatomic bar-
–– Paulsen 2012: 109 patients treated with riers of spread and can be minimized
54 Gy. Radiographic tumor control 98% at at brain parenchymal interface in the
5 yrs. Visual acuity preserved in 91% at 5 yrs. absence of pathologic brain invasion.
–– Milker-Zabel 2009: 32 patients. Median RT Brain parenchyma should be included
dose 54.9 Gy. Median follow-up was 4.5 yrs. in the setting of brain invasion.
LC 100%. (ii) In the setting of suspected or con-
–– Saeed 2010: Br J Opthalmol, 2010. 34 firmed bone involvement, CTVs
patients treated with fractionated RT. At should not be shaved off bone.
median of 58 months, 41% showed improved (iii) The RTOG 0539 used larger expan-
visual acuity, vision stabilized in 50%, dete- sions (1 cm for int risk and 1–2 cm for
riorated in 9%. high risk); however, due to the pre-
dominately infield pattern of failure
RT Technique and low rates of marginal failures, the
–– Simulate supine, aquaplast mask. follow-up NRG BN003 (grade II
–– At minimum, fuse with T1 post-contrast MRIs. meningiomas s/p GTR) uses smaller
T2 series are frequently helpful in distinguish- margins with 5 mm CTV expansions
ing tumor from normal tissues. Neuroradiology which can be shaved down to 3 mm
review recommended for complex cases. around critical structures.
–– Conventionally fractionated RT: (c) CTVs for grade III s may be 0.5–2 cm
(a) GTV = gross tumor, typically defined on depending on institutional practice and
T1 post-contrast MRI: generally include brain parenchyma
(i) Surgical cavity typically included in (d) PTV: Typically 2–5 mm based on
adjuvant RT cases. institution- specific technical and IGRT
(ii) The dural tail is generally not included standards
in protocols. –– Stereotactic radiosurgery
(a) GTV = gross tumor, typically defined on
T1 post-contrast MRI
(b) CTV/PTV expansions: dependent of insti-
tutional practice: often no expansion or
1–2 mm.
Central Nervous System Cancers 107
Brain Metastases [11–15, 128–161] –– Spinal axis MRI and CSF analysis may be
appropriate when concerned for spinal metas-
General tases and/or leptomeningeal spread.
–– Brain metastases are the most common adult
brain tumor. Symptomatic Brain Metastases
–– Occur in approximately 20–40% of patients
with advanced cancers. –– For patients with symptomatic brain metasta-
–– Lung, breast, melanoma, and renal cell carci- ses, corticosteroids are usually appropriate.
nomas are the most common. –– Corticosteroids are generally not indicated in
–– Hemorrhagic metastases are more common in the absence of symptoms; anticonvulsants are
melanoma and renal cell. generally not indicated in the absence of
–– The blood-brain barrier (BBB) can signifi- seizures.
cantly reduce the concentrations of many sys- –– For patients with large and/or symptomatic
temic agents, which can make the CNS a brain metastases associated with significant
pharmacologic sanctuary for metastatic vasogenic edema, midline shift, or risk of her-
progression. niation, urgent neurosurgical consult is
–– Approximately 80% occur in cerebral hemi- indicated.
spheres, 15% in the cerebellum, 5% in the –– Neurosurgical resection for large and/or
brainstem. symptomatic lesions is often preferred for
–– Nomenclature: “solitary” brain metasta- single brain lesions.
sis = only metastatic lesion in the entire body; –– Resection may also be appropriate in select
“single” brain metastasis = only metastatic cases of multiple metastases when surgery is
lesion in the brain. considered the best approach to relieve mass
effect or prevent herniation from a larger
Workup lesion.
–– MRI brain with and without gadolinium. –– If resection is not appropriate or not offered,
–– Brain metastases are typically T1 post-contrast corticosteroids and evaluation for potential
enhancing. radiation therapy should be pursued.
–– Hemorrhage is intrinsically T1 hyperintense
on non-contrast images. General Management Paradigms for
–– Brain metastases may be single or multiple, Asymptomatic Brain Metastases in 2018
typically have circumscribed margins, often –– Single brain lesions:
occur preferentially at the gray-white matter –– SRS alone
interface, may have areas of central necrosis/ –– Neurosurgical resection +/− postoperative
hemorrhage/or cyst, and may be associated SRS to the resection cavity:
with large amounts of vasogenic edema or –– Resection often preferred for large,
mass effect in relation to the lesion size. symptomatic, or threatening lesions.
–– Differential may include metastases, primary –– Limited brain metastases:
brain tumors (e.g., GBM, high-grade glio- –– SRS alone preferred
mas), primary CNS lymphoma, and infection. –– More extensive brain metastases:
Multiple lesions raise suspicion for metasta- –– WBRT
ses, though not pathognomonic. –– SRS to multiple lesions with close MRI
–– Brain biopsy is often not necessary when the surveillance:
probability of metastases is considered high –– SRS candidacy is evolving. Historically,
based on the overall clinical picture (e.g., can- SRS was offered for 1–3 or 4 lesions
cer history, prior brain metastases, multiple based on the inclusion criteria of the
brain lesions, metastases to other organs, ris- randomized SRS +/− WBRT trials
ing tumor markers, etc.). (Aoyama 2006; Chang 2009; Kocher
108 S. K. Nath et al.
2011; Brown 2016); however, there is a –– Vecht 1993. 63 Patients with single met,
growing literature supporting SRS for received WBRT 40 Gy/20 Fx BID randomized
4–10+ lesions (Yamamoto 2014), sug- to +/− up-front Sx. Sx improved median OS
gesting that SRS candidacy should be (10 vs 6 mo).
increasingly individualized. –– Mintz 1996. 84 patients with single met,
received WBRT 30 Gy/10 Fx randomized to
Evolution of Prognostic Models +/− up-front Sx. No difference in OS.
–– Older trials incorporated the RTOG recursive
partitioning analysis (RPA) (Gaspar 1997), Stereotactic Radiosurgery (SRS)
stratifying Class 1 (KPS ≥70, age <65, con-
trolled primary, and no extracranial metasta- WBRT +/− SRS
ses: median OS 7.1 months), Class 3 (KPS –– RTOG 9508 (Andrews 2004): 331 patients
<70; median OS 2.3 months), and Class 2 (not with 1–3 mets, received WBRT 37.5 Gy/15 Fx
Class 1 or 3: median OS 4.2 months). Original randomized to +/− SRS. SRS assoc with
RPA underestimates OS in the modern era, but improved OS for single met (6.5 vs 4.9 mo),
familiarity remains useful for historical pur- LC, KPS, and steroid independence at 6 mo.
poses and interpreting the literature. No diff in OS for all patients with 1–3 mets,
–– Graded prognostic assessment (GPA) neurologic death, or time to any intracranial
(Sperduto 2008) adds # of mets. failure.
–– Diagnosis-specific GPA (DS-GPA) (Sperduto –– Kondziolka 1999. 27 patients with 2–4 mets
2012) modified GPA by histo. randomized to WBRT +/− SRS. Stopped early
–– Lung molecular GPA (lung-molGPA) due to LC benefit with SRS on interim
(Sperduto 2017) adds EGFR and ALK to analysis. 1-yr LC 100% vs 8%. Time to local
GPA. Includes 2186 patients, 2006–2014 with failure 36 vs 6 months.
NSCLC (1521 adenocarcinoma and 665 non-
adenocarcinoma). Median OS 12 months in SRS +/− WBRT
the entire cohort (better than any group in the –– JROSG 99-1 (Aoyama 2006): 132 patients
original 1997 RPA). Patients with high lung- with 1–4 mets, randomized to SRS +/− WBRT
molGPA scores had prolonged median OS of 30 Gy/10 Fx. No difference in OS or neuro-
nearly 4 yrs after brain mets. logic death. WBRT assoc with improved LC,
distant brain control.
–– MDACC (Chang 2009): 58 patients with 1–3
Clinical Data mets randomized to SRS +/− WBRT 30 Gy/12
Fx. Stopped early due to worse cognitive out-
Whole-Brain Radiation (WBRT) comes in WBRT arm. SRS alone assoc with
WBRT vs Supportive Care improved 4-month Hopkins verbal learning test
–– MRC “QUARTZ” trial (Mulvenna 2016). and improved median OS (15 vs 6 mo). WBRT
538 patients with NSCLC, randomized to sup- assoc with improved LC and CNS DFS.
portive care +/− WBRT 20 Gy/5 Fx. No sig- –– EORTC 22952 (Kocher 2011): Local therapy
nificant difference in OS or quality of life. (surgery or SRS) randomized to +/−
WBRT. No difference in OS or time to KPS
WBRT +/− Up-Front Surgery (Sx) for Single deterioration with WBRT. WBRT reduced LR,
Met distant recurrence, and decreased neurologic
–– Patchell 1990. 48 patients with single met, deaths. Soffietti 2013 Patient-reported QOL
received WBRT 36 Gy/12 Fx randomized to analysis of the EORTC 22952. WBRT assoc
+/− up-front Sx. Median OS improved with with decline in global health at 9 mo, cogni-
Sx (40 vs 15 weeks). Sx reduced rates of neu- tive function at 12 mo, physical function and
rologic death (20% vs 52%). fatigue at 2 mo.
Central Nervous System Cancers 109
–– Brown 2016. 213 patients with 1–3 mets, ran- >2 cm. Multi-fraction assoc with better 1-yr
domized to SRS +/− WBRT 30 Gy/12 Fx. LC (91% vs 77%) and lower rates radionecro-
SRS alone assoc with less cognitive decline at sis (11% vs 20%).
3 mo (and 12 mo in longer-term survivors). –– Eaton 2015. Cohort of 76 resected brain
QOL improved with SRS alone. WBRT assoc metastases cavities ≥3 cm treated with single-
with better CNS control. No diff in OS or fraction SRS (N = 40) or three-to-five fraction
functional independence. SRS (N = 36). LF was equivalent at 1-yr (27
vs 25%). Radionecrosis was lower with multi-
SRS Alone for Multiple Metastases fraction at 1-yr (10 vs 19%).
–– LGK0901 (Yamamoto 2014): Prospective
observational study. 1194 patients with 1–10 Postoperative Radiation
brain metastases treated with single-fraction
SRS alone. Median OS: best for single lesion Surgery +/− Postoperative WBRT
(13.9 mo) but no diff in OS between 2–4 –– Patchell 1998. 95 Patients, status post com-
lesions (10.8 mo) vs 5–10 lesions (10.8 mo). plete resection of single met randomized to
No diff in toxicity. Neurologic mortality rates +/− WBRT 50.4 Gy/28 Fx. No diff in OS or
were low overall with SRS alone (8%), with length of functional independence. WBRT
no diff between groups. reduced local and distant brain recurrence,
decreased neurologic deaths.
SRS Alone for Multiple Metastases and –– EORTC 22952 (Kocher 2011). Local therapy
Targetable Mutations (surgery or SRS) randomized to +/−
–– Colorado (Robin 2017): Cohort of ALK and WBRT. No diff in OS or time to KPS deterio-
EGFR NSCLC, SRS alone to ≥4 mets in a ration with WBRT. WBRT reduced LR,
single session (range 4–26). Median OS in yrs distant recurrence, and decreased neurologic
from brain mets was 4.2 (ALK) and 2.4 deaths.
(EGFR). Five-year freedom from WBRT was
97% and from neurologic death was 84%. For Surgery +/− Postoperative SRS to the Cavity
patients treated to >10 mets in single SRS ses- –– MDACC (Mahajan 2017): RCT of 132
sion, mean whole-brain and hippocampal patients status post complete resection of 1–3
doses were only 1.2 Gy and 0.8 Gy, mets <4 cm randomized to +/− single-fraction
respectively. SRS to the cavity (volume-based dosing,
12–16 Gy). SRS assoc with improved LC at 1
SRS Dose year (72% vs 43%). No diff in OS.
–– RTOG 90-05 (Shaw 1996, 2000): Prospective
SRS dose-escalation study. 156 patients who Surgery + Post-op Fractionated SRS to the
received prior conventionally fractionated Cavity
radiation. One-third had recurrent brain –– Stanford (Soltys ASTRO 2015). Single-arm
tumors and two-thirds had recurrent brain phase I/II dose-escalation from 24 to 33 Gy in
mets. Trial established safe single-fraction 3 Fx. 50 patients. At 12 mo: local failures 7%,
SRS doses of 24 Gy for ≤2 cm, 18 Gy for distant 50%, leptomeningeal disease 15%.
2–3 cm, and 15 Gy for 3–4 cm. Neurotoxicity Authors recommend 27–30 Gy/3 Fx for cavi-
associated with tumor size, SRS dose, and ties 2–4 cm.
KPS. –– MSKCC (Brennan 2014). Single-arm phase
II trial. Forty-nine patients treated with
Multi-fraction SRS for Larger Targets 15–22 Gy (median 18 Gy). At 12 mo, local
–– Minniti 2016. Cohort of 289 patients treated failures 22%, distant 44%. Tumors ≥3 cm
with single-fraction vs multi-fraction with superficial dural/pial involvement had
(27 Gy/3 Fx) SRS for unresected lesions the highest risk of local failure.
110 S. K. Nath et al.
Surgery + Post-op SRS vs Post-op WBRT –– Dose: commonly 30 Gy/10 Fx for better
–– NCCTG N107C (Brown 2017). 194 patients KPS, 20 Gy/5 Fx reasonable for poor KPS.
status post resection of one brain metastasis –– For SRS, fuse with thin-slice T1 post-contrast
randomized to WBRT or single-fraction SRS MRI images.
(volume-based dosing, 12–20 Gy). Improved –– For post-op cases, fuse post-op T1 post images
cog deterioration-free survival with post-op at a minimum. Preop MRIs may also be
SRS over WBRT. SRS assoc with shorter time useful.
to any intracranial tumor progression. No diff –– Target definition:
in OS. –– For intact (unresected) targets = gross
tumor.
Clinical Data Summary Notes –– For post-op cases: surgical cavity + any
–– For single brain metastases, tumor ablation residual tumor at a minimum. In some
(surgery or SRS) has been associated with case, more generous volumes at postopera-
improved OS in RCTs. tive dural areas may be appropriate in an
–– Compared to supportive care, surgery alone, effort to lower risk of dural/leptomeningeal
and SRS alone, the addition of WBRT recurrence.
improves CNS disease control, but does not –– A postoperative SRS contouring consen-
improve OS. sus paper (Soliman 2018) recommends
–– WBRT is associated with measurable declines inclusion of the surgical tract, a CTV
in cognitive function and QOL. expansion of 5 to 10 mm along the dura in
–– SRS alone was historically offered for one to cases of preoperative dural contact, and a
three or four lesions based on the inclusion margin of ≤5 mm into the adjacent venous
criteria of SRS +/− WBRT trials. Growing sinus when preoperative venous sinus con-
data suggests that SRS may be appropriate for tact was present. The recommended inclu-
patients with ≥4 lesions in the setting of close sion of the entire surgical tract and
MRI surveillance. relatively large expansions remain
–– Postoperative SRS to the surgical cavity controversial.
improves LC but not OS (Mahajan 2017). –– Expansions:
–– Multi-fraction SRS has been associated with –– For intact cases, typically 0–1 mm.
encouraging LC and toxicity rates and may be –– For postoperative cases, typically at least
particularly useful for larger intact or postop- 1–2 mm around the resection cavity and
erative targets. possibly larger expansions in the case of
–– As emerging systemic agents demonstrate preoperative dural or venous sinus contact
improved CNS penetration and activity (Peters (Soliman 2018).
2017; Mok, 2017; Davies 2017), brain metas- –– In general, expansions for SRS are indi-
tases paradigms are likely to evolve to incor- vidualized based on institutional practices,
porate multidisciplinary approaches involving physician comfort level, and IGRT/immo-
local and systemic therapy. bilization standards.
–– Typical SRS dose fractionation
Planning –– Single fraction:
–– Sim supine, head neutral, and aquaplast mask. • 20–24 Gy for <2 cm,
–– For WBRT: • 18 Gy 2–3 cm,
–– Opposed lateral beams, block orbits, cover • and 15 Gy for 3–4 cm are common.
below foramen magnum, and flash skin. –– Multi-fraction, 24–30 Gy/3 fraction, and
–– Confirm adequate coverage of intracranial 25–30 Gy/5 fractions are common.
contents including temporal fossa and –– SRS often prescribed to the 50–80% isodose
cribriform plate on lateral fields and 3D lines (corresponding to central hot spots of
dose eval. 125–200% of the prescription dose).
Central Nervous System Cancers 111
Prolactinoma 30 Galactorrhea, 2–25 DA agonists e.g. Stop bromocriptine 2-4 50-54 Gy if 20 Gy/1 fx 25–50
DA secreted by amenorrhea, bromocriptine are first line mos prior to RT GTV
hypothalamus, infertility, and work in 85% of pts.
suppressing PL vaginal 30% cannot tolerate bromo Optic nerve max
production in dryness, bc of N/V, HA, fatigue. 8 Gy, need 3–4
pituitary. alibido, ED Surgery if medical failure mm distance
Compression of fails or visual sx. from tumor to
pituitary stalk chiasm
inhibits DA
suppression,leading
to overproduction
of PL.
Common to have
calcifications,
amyloid
GH 25 Acromegaly, <10 Somatostatin (octreotide), Hold for 2–4 months. 80–100
HA, cardiac lancreotide. DA agonists are Patients on octreotide
dz, bone ineffective in acromegaly, reach normal GH and
changes but can be used in 2nd line. IGF -1 level after
significant longer
interval
(radioprotective?).
ACTH 15 Cushings, 15 Ketoconazole (the best), Pegvisomant and 50–80
Nelson syndrome: HTN, DM, cyproheptadine (inhibits ketoconazole can be
ACTH adenoma hirsutism, ACTH), RU-486 given during RT
develops after skin Consider adrenalectomy
adrenalectomy changes,
Males, elderly osteoporosis
TSH <1% Hyperthyroid <1 Somatostatin (octreotide) Hold for 2–4 months.
–– Headache
General
–– Pituitary apoplexy (sudden hemorrhage)
–– 75% functional, 25% nonfunctional.
–– Prolactinomas: galactorrhea, amenorrhea,
–– Associated with MEN-1.
infertility, or hypogonadism
–– Macroadenoma (≥1 cm) (more common in
–– GH-secreting adenomas: acromegaly
nonsecretory).
–– ACTH-secreting adenomas: Cushings
–– Microadenoma (<1 cm).
–– Prolactinomas are most common (30%).
Differential Diagnosis
DDx Sellar Mass –– Craniopharyngioma, meningioma, cyst, GCT,
–– Craniopharyngioma chordoma
–– Glioma –– Metastases to the pituitary: most likely from
–– Pituitary adenoma or carcinoma breast and lung cancer
–– Germ cell tumors –– Benign pituitary hyperplasia due to preg-
–– Ependymomas nancy, lactation, long-standing primary hypo-
–– Meningioma thyroidism or hypogonadism, cirrhosis
–– Metastasis
–– Benign: cyst, abscess, cavernous sinus AVM Workup
–– H&P, labs: serum prolactin, IGF-1, 24-h
Presentation urine-free cortisol, LH/FSH, and free/total T4/
–– Symptoms due to mass effect or ↑hormone TSH.
secretion –– Thin-cut MRI is only imaging needed; normal
–– Bitemporal hemianopsia or other patterns of pituitary and adenomas are isointense to
vision impairment slightly hyperintense on T1 and may have T2
Central Nervous System Cancers 113
–– Local tumor control in RT series is ~95%. If no hormone normalization after full surgi-
–– Key series: Jagannathan 2007; Minniti 2007; cal + RT treatment and med management:
Sheehan 2013.
–– Cushings → adrenalectomy
Somatotroph Adenomas (Growth Hormone) –– TSH → thyroidectomy
–– Radiation is indicated for patients who fail to
obtain endocrine remission or tumor control Outcomes
from surgical or medical management. –– Non-functioning adenomas: LC is >90%.
–– Endocrine remission defined as normalization –– Functioning adenomas (rates of hormone
of serum GH and IGF-1 levels; some studies normalization):
use oral glucose tolerance test results to define • 50% with RT alone vs >80% with
remission. RT + medical therapy
–– Endocrine remission rates vary across series • >2 years to hormone normalization
but average ~50%. • PRL: 50% with RT alone vs >80% with
–– Mean time to hormone remission is RT + medical therapy.
~24 months but can take up to 15 years. • Time to normalization is 2–8 years.
–– Local tumor control in RT series is ~95%. • ACTH: 50% with RT alone vs >80% with
–– Key studies: Losa 2008; Lee 2014. RT + medical therapy:
• Time to normalization is 1–4 years.
RT indications • GH: 50% w/ RT alone at 10 years, with
–– Inoperable medical therapy is 70%:
–– STR with persistent hypersecretion or residual • Time to normalization is 3–10 years.
near critical structures
–– Recurrence/progression after surgery Toxicity/Follow-up
–– TSH secreting (always give post-op RT) –– 100% will get some deficiency.
–– Panhypopituitarism is most common.
Radiation Technique Hypopituitarism occurs in about 20% at
–– SRS or fractionated RT can be considered. The 5 years but may rise up to 80% at 10 years
literature suggests that both are equally effective (Sheehan 2011; Minniti 2007):
in local control, although non-randomized com- • GH deficiency, 100% at 5y
parisons suggest a shorter interval to hormone • GnRH 60% at 10y
normalization with SRS. However, these are ret- • ACTH <60% at 10y
rospective studies, and SRS may have been • TSH <30% at 10y
preferentially used on smaller tumors. • HyperPRL 20–50%
–– SRS is appropriate for tumors <3 cm and at –– Sensitivity of hormones to RT (“G FLAT”):
least 3–5 mm from the optic pathway. (1) GH, (2) FSH/LH, (3) TSH/ACTH. Because
–– The majority of SRS series have used GK, but prolactin should not be elevated in a
linac-based approaches are reasonable. nonpregnant adult, it is not reported as
–– Standard doses for nonfunctional adenomas suppressed after RT to the pituitary.
are 14–16 Gy in 1 fraction or 45–50.4 Gy in Vasopressin and oxytocin are made in
1.8 Gy fractions. posterior pituitary and are resistant to
–– Standard doses for functional adenomas are RT. Thus, RT is not linked to DI.
16–25 Gy in 1 fraction or 50.4–54 Gy in –– Low risk for optic pathway injury or second
1.8 Gy fractions. malignancy.
–– Constraints: –– MRI and labs q6–12 months.
• Chiasm = 54 Gy –– Endocrinology evaluation and follow-up
• SRS = 8 Gy (need 3–4 mm distance from recommended.
tumor to chiasm)
Central Nervous System Cancers 115
Trigeminal Neuralgia (TN) [208–214] Pain relief was equivalent between two arms,
but two-isocenter arm had more toxicity.
Background –– Maesawa 2001: n = 220. GKRS. Median age
–– Increases with age; most occur after age 50. 70. Sixty-one percent previous surgery.
–– More common in women than men (approx. Complete or partial pain relief in 86% at 1 y.
1.5:1). Complete pain relief 65% at 6 m, 70% at 1 yr
–– Classical TN is either related to vascular CN5 and 75% at 33 m. Only 10% of pts developed
compression or idiopathic. new of increased facial numbness. Facial numb-
–– Secondary TN, or trigeminal neuropathy, may ness was a predictor of long-lasting pain relief.
arise from herpes zoster, postherpetic neural- –– Lopez 2004: Systematic review. Seventy-five
gia, MS, CN5 trauma, and nonvascular CN5 percent of pts have CR after SRS at 1 yr. Only
compression. Sensory loss, bilateral involve- 50% maintain this at 3 yrs.
ment, and younger age are associated with a –– Patients with classic TGN features felt to have
higher risk of secondary TN. better responses to therapy (Taich 2016).
–– Classical TN pain is paroxysmal, unilateral,
lancinating, and provocable (e.g., by talking, Treatment
brushing teeth, chewing). –– Medical therapy: first-line therapy.
–– Classic TN has the following features: Carbamazepine (Tegretol) or other anticon-
(a) Paroxysmal attacks lasting 1 s–2 min, vulsants. Side effects may be intolerable in
affecting any branch of CN5. some patients.
(b) Pain has at least one feature: sharp,
–– Microvascular decompression (MVD) is
intense, stabbing, or superficial. preferred if compressing vessel is found
(c) Attacks are stereotyped in the patient. –– Other surgical procedures: RFA, glycerol
(d) Not attributed to another disorder. injection, balloon compression.
–– Pretreatment MRI to rule out structural causes. –– Gamma Knife SRS: reserved for medically
–– CISS and 3D FIESTA sequences can offer refractory cases. A single 4 mm collimator
high-resolution views of the TGN. gamma knife “shot” is placed on the dorsal
root entry zone where CN5 exits from the
BNI Grade pons, with the 50% IDL placed at the interface
–– (I) No pain, taking no med of the entry zone and the brainstem. 80–85 Gy
–– (II) Occasional pain, but taking no med max point dose prescribed to the 100%
–– (III) Some pain, controlled w/ meds IDL. Brainstem max 45 Gy.
–– (IIIa) No pain, continued med
–– (IIIb) Persistent pain, controlled w/ med Follow-Up
–– (IV) Some pain, not controlled w/ meds –– Median time to some pain relief is 1–2 mo.
–– (V) Severe pain or no relief –– 60–80% some pain relief at 6 mo.
–– 50% complete relief.
Studies –– TN pain can recur during follow-up. Repeat
–– Flickinger 2001: RCT of 87 pts comparing SRS may be an option for carefully selected
one vs two isocenters. Two isocenters patients (Herman 2004, Pollock 2005,
increased length of trigeminal nerve being Hasegawa 2002).
treated. Sixty-eight percent of pts w/ response. –– Check for loss of corneal reflex.
Central Nervous System Cancers 119
glioblastoma multiforme: a prospective randomized radiotherapy boost, external radiation therapy, and
clinical trial. J Clin Oncol. 2004;22:1583–8. carmustine. Neurosurgery. 2002;51:343–55; discus-
30. Stupp R, Hegi ME, Mason WP, et al. Effects of sion 355–7.
radiotherapy with concomitant and adjuvant temo- 42. Laperriere NJ, Leung PM, McKenzie S, et al.
zolomide versus radiotherapy alone on survival in Randomized study of brachytherapy in the initial
glioblastoma in a randomised phase III study: 5-year management of patients with malignant astrocytoma.
analysis of the EORTC-NCIC trial. Lancet Oncol. Int J Radiat Oncol Biol Phys. 1998;41:1005–11.
2009;10:459–66. 43. Souhami L, Seiferheld W, Brachman D, et al.
31. van den Bent MJ, Carpentier AF, Brandes AA, Randomized comparison of stereotactic radiosurgery
et al. Adjuvant procarbazine, lomustine, and vin- followed by conventional radiotherapy with carmus-
cristine improves progression-free survival but not tine to conventional radiotherapy with carmustine
overall survival in newly diagnosed anaplastic oli- for patients with glioblastoma multiforme: report of
godendrogliomas and oligoastrocytomas: a ran- Radiation Therapy Oncology Group 93-05 protocol.
domized European Organisation for Research and Int J Radiat Oncol Biol Phys. 2004;60:853–60.
Treatment of Cancer phase III trial. J Clin Oncol. 44. Tsien C, Moughan J, Michalski JM, et al. Phase I
2006;24:2715–22. three-dimensional conformal radiation dose esca-
32. Walker MD, Alexander E Jr, Hunt WE, et al. lation study in newly diagnosed glioblastoma:
Evaluation of BCNU and/or radiotherapy in the Radiation Therapy Oncology Group Trial 98-03. Int
treatment of anaplastic gliomas. A cooperative clini- J Radiat Oncol Biol Phys. 2009;73:699–708.
cal trial. J Neurosurg. 1978;49:333–43. 45. Stewart LA. Chemotherapy in adult high-grade
33. Wick W, Hartmann C, Engel C, et al. NOA-04 glioma: a systematic review and meta-analysis of
randomized phase III trial of sequential radioche- individual patient data from 12 randomised trials.
motherapy of anaplastic glioma with procarbazine, Lancet. 2002;359:1011–8.
lomustine, and vincristine or temozolomide. J Clin 46. Stupp R, Taillibert S, Kanner AA, et al. Maintenance
Oncol. 2009;27:5874–80. therapy with tumor-treating fields plus temozolo-
34. Wick W, Platten M, Meisner C, et al. Temozolomide mide vs temozolomide alone for glioblastoma: a ran-
chemotherapy alone versus radiotherapy alone domized clinical trial. JAMA. 2015;314:2535–43.
for malignant astrocytoma in the elderly: the 47. Hildebrand J, Gorlia T, Kros JM, et al. Adjuvant
NOA-08 randomised, phase 3 trial. Lancet Oncol. dibromodulcitol and BCNU chemotherapy in
2012;13:707–15. anaplastic astrocytoma: results of a randomised
35. Roa W, Kepka L, Kumar N, et al. International European Organisation for Research and Treatment
Atomic Energy Agency randomized phase III Study of Cancer phase III study (EORTC study 26882).
of radiation therapy in elderly and/or frail patients Eur J Cancer. 2008;44:1210–6.
with newly diagnosed glioblastoma multiforme. J 48. Niyazi M, Brada M, Chalmers AJ, et al. ESTRO-
Clin Oncol. 2015;33:4145–50. ACROP guideline "target delineation of glioblasto-
36. Brandes AA, Franceschi E, Tosoni A, et al. MGMT mas". Radiother Oncol. 2016;118:35–42.
promoter methylation status can predict the incidence 49. Chang S, Zhang P, Cairncross JG, et al. Phase III
and outcome of pseudoprogression after concomi- randomized study of radiation and temozolomide
tant radiochemotherapy in newly diagnosed glio- versus radiation and nitrosourea therapy for
blastoma patients. J Clin Oncol. 2008;26:2192–7. anaplastic astrocytoma: results of NRG Oncology
37. Ruben JD, Dally M, Bailey M, et al. Cerebral RTOG 9813. Neuro-Oncology. 2017;19:252–8.
radiation necrosis: incidence, outcomes, and risk 50. Cabrera AR, Kirkpatrick JP, Fiveash JB, et al.
factors with emphasis on radiation parameters Radiation therapy for glioblastoma: executive sum-
and chemotherapy. Int J Radiat Oncol Biol Phys. mary of an American Society for Radiation Oncology
2006;65:499–508. Evidence-Based Clinical Practice Guideline. Pract
38. Hochberg FH, Pruitt A. Assumptions in the radiother- Radiat Oncol. 2016;6:217–25.
apy of glioblastoma. Neurology. 1980;30:907–11. 51. Lacroix M, Abi-Said D, Fourney DR, et al. A mul-
39. Walker MD, Green SB, Byar DP, et al. Randomized tivariate analysis of 416 patients with glioblastoma
comparisons of radiotherapy and nitrosoureas for the multiforme: prognosis, extent of resection, and sur-
treatment of malignant glioma after surgery. N Engl vival. J Neurosurg. 2001;95:190–8.
J Med. 1980;303:1323–9. 52. Sanai N, Polley MY, McDermott MW, et al. An
40. Shapiro WR, Green SB, Burger PC, et al. extent of resection threshold for newly diagnosed
Randomized trial of three chemotherapy regimens glioblastomas. J Neurosurg. 2011;115:3–8.
and two radiotherapy regimens and two radiotherapy 53. Chaichana KL, Jusue-Torres I, Navarro-Ramirez R,
regimens in postoperative treatment of malignant et al. Establishing percent resection and residual vol-
glioma. Brain Tumor Cooperative Group Trial 8001. ume thresholds affecting survival and recurrence for
J Neurosurg. 1989;71:1–9. patients with newly diagnosed intracranial glioblas-
41. Selker RG, Shapiro WR, Burger P, et al. The Brain toma. Neuro-Oncology. 2014;16:113–22.
Tumor Cooperative Group NIH Trial 87-01: a ran- 54. Brown TJ, Brennan MC, Li M, et al. Association
domized comparison of surgery, external radio- of the Extent of Resection With Survival in
therapy, and carmustine versus surgery, interstitial
Central Nervous System Cancers 125
Glioblastoma: A Systematic Review and Meta- 66. Malmstrom A, Gronberg BH, Marosi C, et al.
analysis. JAMA Oncol. 2016;2:1460–9. Temozolomide versus standard 6-week radio-
55. Valtonen S, Timonen U, Toivanen P, et al. Interstitial therapy versus hypofractionated radiotherapy in
chemotherapy with carmustine-loaded polymers patients older than 60 years with glioblastoma: the
for high-grade gliomas: a randomized double-blind Nordic randomised, phase 3 trial. Lancet Oncol.
study. Neurosurgery. 1997;41:44–8; discussion 2012;13:916–26.
48–9. 67. Perry JR, Laperriere N, O’Callaghan CJ, et al.
56. Westphal M, Ram Z, Riddle V, et al. Gliadel wafer Short-Course Radiation plus Temozolomide in
in initial surgery for malignant glioma: long-term Elderly Patients with Glioblastoma. N Engl J Med.
follow-up of a multicenter controlled trial. Acta 2017;376:1027–37.
Neurochir (Wien). 2006;148:269–75; discussion 68. van den Bent MJ, Baumert B, Erridge SC, et al.
275. Interim results from the CATNON trial (EORTC
57. Garside R, Pitt M, Anderson R, et al. The effective- study 26053-22054) of treatment with concurrent and
ness and cost-effectiveness of carmustine implants adjuvant temozolomide for 1p/19q non-co-deleted
and temozolomide for the treatment of newly anaplastic glioma: a phase 3, randomised, open-label
diagnosed high-grade glioma: a systematic review intergroup study. Lancet. 2017;390:1645–53.
and economic evaluation. Health Technol Assess. 69. Gilbert MR, Wang M, Aldape KD, et al. Dose-dense
2007;11:iii-iv–x-221. temozolomide for newly diagnosed glioblastoma:
58. Kristiansen K, Hagen S, Kollevold T, et al. Combined a randomized phase III clinical trial. J Clin Oncol.
modality therapy of operated astrocytomas grade III 2013;31:4085–91.
and IV. Confirmation of the value of postoperative 70. Rusthoven CG, Koshy M, Sher DJ, et al. Combined-
irradiation and lack of potentiation of bleomycin on modality therapy with radiation and chemotherapy
survival time: a prospective multicenter trial of the for elderly patients with glioblastoma in the temo-
Scandinavian Glioblastoma Study Group. Cancer. zolomide era: a national cancer database analysis.
1981;47:649–52. JAMA Neurol. 2016;73:821–8.
59. Bleehen NM, Stenning SP. A Medical Research 71. Gilbert MR, Dignam JJ, Armstrong TS, et al. A ran-
Council trial of two radiotherapy doses in the treat- domized trial of bevacizumab for newly diagnosed
ment of grades 3 and 4 astrocytoma. The Medical glioblastoma. N Engl J Med. 2014;370:699–708.
Research Council Brain Tumour Working Party. Br J 72. Chinot OL, Wick W, Mason W, et al. Bevacizumab
Cancer. 1991;64:769–74. plus radiotherapy-temozolomide for newly diagnosed
60. Cardinale R, Won M, Choucair A, et al. A phase II glioblastoma. N Engl J Med. 2014;370:709–22.
trial of accelerated radiotherapy using weekly ste- 73. McDonald MW, Shu HK, Curran WJ Jr, et al. Pattern
reotactic conformal boost for supratentorial glioblas- of failure after limited margin radiotherapy and
toma multiforme: RTOG 0023. Int J Radiat Oncol temozolomide for glioblastoma. Int J Radiat Oncol
Biol Phys. 2006;65:1422–8. Biol Phys. 2011;79:130–6.
61. Nelson DF, Diener-West M, Horton J, et al. 74. Minniti G, Amelio D, Amichetti M, et al. Patterns
Combined modality approach to treatment of of failure and comparison of different target vol-
malignant gliomas--re-evaluation of RTOG 7401/ ume delineations in patients with glioblastoma
ECOG 1374 with long-term follow-up: a joint study treated with conformal radiotherapy plus concomi-
of the Radiation Therapy Oncology Group and the tant and adjuvant temozolomide. Radiother Oncol.
Eastern Cooperative Oncology Group. NCI Monogr. 2010;97:377–81.
1988;6:279–84. 75. Gebhardt BJ, Dobelbower MC, Ennis WH, et al.
62. Nelson DF, Curran WJ Jr, Scott C, et al. Patterns of failure for glioblastoma multiforme fol-
Hyperfractionated radiation therapy and bis- lowing limited-margin radiation and concurrent
chlorethyl nitrosourea in the treatment of malignant temozolomide. Radiat Oncol. 2014;9:130.
glioma--possible advantage observed at 72.0 Gy 76. Paulsson AK, McMullen KP, Peiffer AM, et al.
in 1.2 Gy B.I.D. fractions: report of the Radiation Limited margins using modern radiotherapy tech-
Therapy Oncology Group Protocol 8302. Int J niques does not increase marginal failure rate of
Radiat Oncol Biol Phys. 1993;25:193–207. glioblastoma. Am J Clin Oncol. 2014;37:177–81.
63. Werner-Wasik M, Scott CB, Nelson DF, et al. Final 77. Chang EL, Akyurek S, Avalos T, et al. Evaluation of
report of a phase I/II trial of hyperfractionated and peritumoral edema in the delineation of radiotherapy
accelerated hyperfractionated radiation therapy with clinical target volumes for glioblastoma. Int J Radiat
carmustine for adults with supratentorial malignant Oncol Biol Phys. 2007;68:144–50.
gliomas. Radiation Therapy Oncology Group Study 78. Combs SE, Thilmann C, Edler L, et al. Efficacy of
83-02. Cancer. 1996;77:1535–43. fractionated stereotactic reirradiation in recurrent
64. Keime-Guibert F, Chinot O, Taillandier L, et al. gliomas: long-term results in 172 patients treated in
Radiotherapy for glioblastoma in the elderly. N Engl a single institution. J Clin Oncol. 2005;23:8863–9.
J Med. 2007;356:1527–35. 79. Fogh SE, Andrews DW, Glass J, et al.
65. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene Hypofractionated stereotactic radiation therapy: an
silencing and benefit from temozolomide in glio- effective therapy for recurrent high-grade gliomas. J
blastoma. N Engl J Med. 2005;352:997–1003. Clin Oncol. 2010;28:3048–53.
126 S. K. Nath et al.
80. Stupp R, Wong ET, Kanner AA, et al. NovoTTF- International Extranodal Lymphoma Study Group
100A versus physician's choice chemotherapy in experience. J Clin Oncol. 2003;21:266–72.
recurrent glioblastoma: a randomised phase III 93. Nguyen PL, Chakravarti A, Finkelstein DM, et al.
trial of a novel treatment modality. Eur J Cancer. Results of whole-brain radiation as salvage of
2012;48:2192–202. methotrexate failure for immunocompetent patients
81. Shapiro LQ, Beal K, Goenka A, et al. Patterns of with primary CNS lymphoma. J Clin Oncol.
failure after concurrent bevacizumab and hypofrac- 2005;23:1507–13.
tionated stereotactic radiation therapy for recurrent 94. Murray K, Kun L, Cox J. Primary malignant lym-
high-grade glioma. Int J Radiat Oncol Biol Phys. phoma of the central nervous system. Results of
2013;85:636–42. treatment of 11 cases and review of the literature. J
82. Vuorinen V, Hinkka S, Farkkila M, et al. Debulking Neurosurg. 1986;65:600–7.
or biopsy of malignant glioma in elderly people – a 95. Batchelor T, Carson K, O'Neill A, et al. Treatment
randomised study. Acta Neurochir. 2003;145:5–10. of primary CNS lymphoma with methotrexate and
83. Brem H, Piantadosi S, Burger PC, et al. Placebo- deferred radiotherapy: a report of NABTT 96-07. J
controlled trial of safety and efficacy of intra- Clin Oncol. 2003;21:1044–9.
operative controlled delivery by biodegradable 96. Gavrilovic IT, Hormigo A, Yahalom J, et al. Long-
polymers of chemotherapy for recurrent gliomas. term follow-up of high-dose methotrexate-based
The Polymer-brain Tumor Treatment Group. Lancet. therapy with and without whole brain irradiation for
1995;345:1008–12. newly diagnosed primary CNS lymphoma. J Clin
84. Laperriere N, Zuraw L, Cairncross G, et al. Oncol. 2006;24:4570–4.
Radiotherapy for newly diagnosed malignant glioma 97. Glass J, Won M, Schultz CJ, et al. Phase I and II
in adults: a systematic review. Radiother Oncol. study of induction chemotherapy with methotrexate,
2002;64:259–73. rituximab, and temozolomide, followed by whole-
85. Stupp R, Taillibert S, Kanner A, et al. Effect of brain radiotherapy and postirradiation temozolo-
tumor-treating fields plus maintenance temozolo- mide for primary cns lymphoma: NRG Oncology
mide vs maintenance temozolomide alone on sur- RTOG 0227. J Clin Oncol. 2016;34:1620–5.
vival in patients with glioblastoma: a randomized 98. Fisher B, Seiferheld W, Schultz C, et al. Secondary
clinical trial. JAMA. 2017;318:2306–16. analysis of Radiation Therapy Oncology Group
86. Cairncross JG, Wang M, Jenkins RB, et al. Benefit study (RTOG) 9310: an intergroup phase II com-
from procarbazine, lomustine, and vincristine in oli- bined modality treatment of primary central nervous
godendroglial tumors is associated with mutation of system lymphoma. J Neuro-Oncol. 2005;74:201–5.
IDH. J Clin Oncol. 2014;32:783–90. 99. Hoang-Xuan K, Taillandier L, Chinot O, et al.
87. Abrey LE, Ben-Porat L, Panageas KS, et al. Primary Chemotherapy alone as initial treatment for primary
central nervous system lymphoma: the Memorial CNS lymphoma in patients older than 60 years: a
Sloan-Kettering Cancer Center prognostic model. J multicenter phase II study (26952) of the European
Clin Oncol. 2006;24:5711–5. Organization for Research and Treatment of Cancer
88. DeAngelis LM, Seiferheld W, Schold SC, et al. Brain Tumor Group. J Clin Oncol. 2003;21:2726–31.
Combination chemotherapy and radiotherapy 100. Morris PG, Correa DD, Yahalom J, et al. Rituximab,
for primary central nervous system lymphoma: methotrexate, procarbazine, and vincristine followed
Radiation Therapy Oncology Group Study 93-10. J by consolidation reduced-dose whole-brain radio-
Clin Oncol. 2002;20:4643–8. therapy and cytarabine in newly diagnosed primary
89. Laack NN, Ballman KV, Brown PB, et al. Whole- CNS lymphoma: final results and long-term out-
brain radiotherapy and high-dose methylpred- come. J Clin Oncol. 2013;31:3971–9.
nisolone for elderly patients with primary central 101. Illerhaus G, Marks R, Muller F, et al. High-dose
nervous system lymphoma: Results of North Central methotrexate combined with procarbazine and
Cancer Treatment Group (NCCTG) 96-73-51. Int J CCNU for primary CNS lymphoma in the elderly:
Radiat Oncol Biol Phys. 2006;65:1429–39. results of a prospective pilot and phase II study. Ann
90. Nelson DF, Martz KL, Bonner H, et al. Non- Oncol. 2009;20:319–25.
Hodgkin’s lymphoma of the brain: can high dose, 102. Juergens A, Pels H, Rogowski S, et al. Long-term
large volume radiation therapy improve survival? survival with favorable cognitive outcome after
Report on a prospective trial by the Radiation chemotherapy in primary central nervous system
Therapy Oncology Group (RTOG): RTOG 8315. Int lymphoma. Ann Neurol. 2010;67:182–9.
J Radiat Oncol Biol Phys. 1992;23:9–17. 103. Yahalom J, Illidge T, Specht L, et al. Modern radia-
91. Thiel E, Korfel A, Martus P, et al. High-dose metho- tion therapy for extranodal lymphomas: field and
trexate with or without whole brain radiotherapy for dose guidelines from the International Lymphoma
primary CNS lymphoma (G-PCNSL-SG-1): a phase Radiation Oncology Group. Int J Radiat Oncol Biol
3, randomised, non-inferiority trial. Lancet Oncol. Phys. 2015;92:11–31.
2010;11:1036–47. 104. Goldsmith BJ, Wara WM, Wilson CB, et al.
92. Ferreri AJ, Blay JY, Reni M, et al. Prognostic Postoperative irradiation for subtotally resected
scoring system for primary CNS lymphomas: the meningiomas. A retrospective analysis of 140
Central Nervous System Cancers 127
patients treated from 1967 to 1990. J Neurosurg. outcome in 318 patients. Int J Radiat Oncol Biol
1994;80:195–201. Phys. 2014;89:569–75.
105. Olivero WC, Lister JR, Elwood PW. The natural his- 120. Kondziolka D, Flickinger JC, Perez B. Judicious
tory and growth rate of asymptomatic meningiomas: resection and/or radiosurgery for parasagit-
a review of 60 patients. J Neurosurg. 1995;83:222–4. tal meningiomas: outcomes from a multicenter
106. Attia A, Chan MD, Mott RT, et al. Patterns of review. Gamma Knife Meningioma Study Group.
failure after treatment of atypical meningioma Neurosurgery. 1998;43:405–13; discussion 413–4.
with gamma knife radiosurgery. J Neuro-Oncol. 121. Patil CG, Hoang S, Borchers DJ 3rd, et al. Predictors
2012;108:179–85. of peritumoral edema after stereotactic radiosur-
107. Bos D, Poels MM, Adams HH, et al. Prevalence, gery of supratentorial meningiomas. Neurosurgery.
clinical management, and natural course of inci- 2008;63:435–40; discussion 440–2.
dental findings on brain mr images: the population- 122. Marchetti M, Bianchi S, Pinzi V, et al. Multisession
based Rotterdam Scan Study. Radiology. radiosurgery for sellar and parasellar benign menin-
2016;281:507–15. giomas: long-term tumor growth control and visual
108. Simpson D. The recurrence of intracranial menin- outcome. Neurosurgery. 2016;78:638–46.
giomas after surgical treatment. J Neurol Neurosurg 123. Navarria P, Pessina F, Cozzi L, et al. Hypofractionated
Psychiatry. 1957;20:22–39. stereotactic radiation therapy in skull base meningi-
109. Sughrue ME, Kane AJ, Shangari G, et al. Prevalence omas. J Neuro-Oncol. 2015;124:283–9.
of previous extracranial malignancies in a series 124. Paulsen F, Doerr S, Wilhelm H, et al. Fractionated
of 1228 patients presenting with meningioma. J stereotactic radiotherapy in patients with optic nerve
Neurosurg. 2010;113:1115–21. sheath meningioma. Int J Radiat Oncol Biol Phys.
110. Oya S, Kawai K, Nakatomi H, et al. Significance 2012;82:773–8.
of Simpson grading system in modern meningioma 125. Milker-Zabel S, Huber P, Schlegel W, et al.
surgery: integration of the grade with MIB-1 label- Fractionated stereotactic radiation therapy in the
ing index as a key to predict the recurrence of WHO management of primary optic nerve sheath menin-
Grade I meningiomas. J Neurosurg. 2012;117:121–8. giomas. J Neuro-Oncol. 2009;94:419–24.
111. Rogers L, Barani I, Chamberlain M, et al. 126. Kondziolka D, Mathieu D, Lunsford LD, et al.
Meningiomas: knowledge base, treatment outcomes, Radiosurgery as definitive management of intra-
and uncertainties. A RANO review. J Neurosurg. cranial meningiomas. Neurosurgery. 2008;62:53–8;
2015;122:4–23. discussion 58–60.
112. Sun SQ, Hawasli AH, Huang J, et al. An evidence- 127. Santacroce A, Walier M, Regis J, et al. Long-term
based treatment algorithm for the management of tumor control of benign intracranial meningio-
WHO Grade II and III meningiomas. Neurosurg mas after radiosurgery in a series of 4565 patients.
Focus. 2015;38:E3. Neurosurgery. 2012;70:32–9; discussion 39.
113. Yano S, Kuratsu J, Kumamoto Brain Tumor 128. Andrews DW, Scott CB, Sperduto PW, et al. Whole
Research Group. Indications for surgery in patients brain radiation therapy with or without stereotactic
with asymptomatic meningiomas based on an exten- radiosurgery boost for patients with one to three
sive experience. J Neurosurg. 2006;105:538–43. brain metastases: phase III results of the RTOG 9508
114. Rogers L, Zhang P, Vogelbaum MA, et al. randomised trial. Lancet. 2004;363:1665–72.
Intermediate-risk meningioma: initial outcomes 129. Aoyama H, Shirato H, Tago M, et al. Stereotactic
from NRG Oncology RTOG 0539. J Neurosurg. radiosurgery plus whole-brain radiation therapy vs
2017;1:1–13. stereotactic radiosurgery alone for treatment of brain
115. Tanzler E, Morris CG, Kirwan JM, et al. Outcomes metastases: a randomized controlled trial. JAMA.
of WHO Grade I meningiomas receiving definitive 2006;295:2483–91.
or postoperative radiotherapy. Int J Radiat Oncol 130. Chang EL, Wefel JS, Hess KR, et al. Neurocognition
Biol Phys. 2011;79:508–13. in patients with brain metastases treated with radio-
116. Solda F, Wharram B, De Ieso PB, et al. Long-term surgery or radiosurgery plus whole-brain irradia-
efficacy of fractionated radiotherapy for benign tion: a randomised controlled trial. Lancet Oncol.
meningiomas. Radiother Oncol. 2013;109:330–4. 2009;10:1037–44.
117. Stafford SL, Pollock BE, Foote RL, et al. 131. Gaspar L, Scott C, Rotman M, et al. Recursive par-
Meningioma radiosurgery: tumor control, out- titioning analysis (RPA) of prognostic factors in
comes, and complications among 190 consecutive three Radiation Therapy Oncology Group (RTOG)
patients. Neurosurgery. 2001;49:1029–37; discus- brain metastases trials. Int J Radiat Oncol Biol Phys.
sion 1037–8. 1997;37:745–51.
118. Sheehan JP, Cohen-Inbar O, Ruangkanchanasetr R, 132. Gelber RD, Larson M, Borgelt BB, et al. Equivalence
et al. Post-radiosurgical edema associated with para- of radiation schedules for the palliative treatment of
sagittal and parafalcine meningiomas: a multicenter brain metastases in patients with favorable progno-
study. J Neuro-Oncol. 2015;125:317–24. sis. Cancer. 1981;48:1749–53.
119. Fokas E, Henzel M, Surber G, et al. Stereotactic 133. Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant
radiation therapy for benign meningioma: long-term whole-brain radiotherapy versus observation after
128 S. K. Nath et al.
radiosurgery or surgical resection of one to three for lung cancer using molecular markers (Lung-
cerebral metastases: results of the EORTC 22952- molGPA). JAMA Oncol. 2017;3:827–31.
26001 study. J Clin Oncol. 2011;29:134–41. 147. Horton J, Baxter DH, Olson KB. The management
134. Kurtz JM, Gelber R, Brady LW, et al. The palliation of metastases to the brain by irradiation and corti-
of brain metastases in a favorable patient population: costeroids. Am J Roentgenol Radium Therapy, Nucl
a randomized clinical trial by the Radiation Therapy Med. 1971;111:334–6.
Oncology Group. Int J Radiat Oncol Biol Phys. 148. Mulvenna P, Nankivell M, Barton R, et al.
1981;7:891–5. Dexamethasone and supportive care with or without
135. Patchell RA, Tibbs PA, Regine WF, et al. whole brain radiotherapy in treating patients with
Postoperative radiotherapy in the treatment of single non-small cell lung cancer with brain metastases
metastases to the brain: a randomized trial. JAMA. unsuitable for resection or stereotactic radiotherapy
1998;280:1485–9. (QUARTZ): results from a phase 3, non-inferiority,
136. Patchell RA, Tibbs PA, Walsh JW, et al. A random- randomised trial. Lancet. 2016;388:2004–14.
ized trial of surgery in the treatment of single metas- 149. Vecht CJ, Haaxma-Reiche H, Noordijk EM, et al.
tases to the brain. N Engl J Med. 1990;322:494–500. Treatment of single brain metastasis: radiotherapy
137. Minniti G, Scaringi C, Paolini S, et al. Single- alone or combined with neurosurgery? Ann Neurol.
fraction versus multifraction (3 × 9 Gy) stereotac- 1993;33:583–90.
tic radiosurgery for large (>2 cm) brain metastases: 150. Kondziolka D, Patel A, Lunsford LD, et al.
a comparative analysis of local control and risk of Stereotactic radiosurgery plus whole brain radio-
radiation-induced brain necrosis. Int J Radiat Oncol therapy versus radiotherapy alone for patients with
Biol Phys. 2016;95(4):1142–8. multiple brain metastases. Int J Radiat Oncol Biol
138. Sneed PK, Suh JH, Goetsch SJ, et al. A multi- Phys. 1999;45:427–34.
institutional review of radiosurgery alone vs. radio- 151. Mintz AH, Kestle J, Rathbone MP, et al. A random-
surgery with whole brain radiotherapy as the initial ized trial to assess the efficacy of surgery in addi-
management of brain metastases. Int J Radiat Oncol tion to radiotherapy in patients with a single cerebral
Biol Phys. 2002;53:519–26. metastasis. Cancer. 1996;78:1470–6.
139. Yamamoto M, Serizawa T, Shuto T, et al. Stereotactic 152. Soffietti R, Kocher M, Abacioglu UM, et al. A
radiosurgery for patients with multiple brain metas- European Organisation for Research and Treatment
tases (JLGK0901): a multi-institutional prospective of Cancer phase III trial of adjuvant whole-brain
observational study. Lancet Oncol. 2014;15:387–95. radiotherapy versus observation in patients with one
140. Choi CY, Chang SD, Gibbs IC, et al. Stereotactic to three brain metastases from solid tumors after
radiosurgery of the postoperative resection cavity surgical resection or radiosurgery: quality-of-life
for brain metastases: prospective evaluation of tar- results. J Clin Oncol. 2013;31:65–72.
get margin on tumor control. Int J Radiat Oncol Biol 153. Shaw E, Scott C, Souhami L, et al. Radiosurgery
Phys. 2012;84:336–42. for the treatment of previously irradiated recur-
141. Brown PD, Jaeckle K, Ballman KV, et al. Effect of rent primary brain tumors and brain metastases:
radiosurgery alone vs radiosurgery with whole brain initial report of radiation therapy oncology group
radiation therapy on cognitive function in patients protocol (90- 05). Int J Radiat Oncol Biol Phys.
with 1 to 3 brain metastases: a randomized clinical 1996;34:647–54.
trial. JAMA. 2016;316:401–9. 154. Shaw E, Scott C, Souhami L, et al. Single dose
142. Brown PD, Ahluwalia MS, Khan OH, et al. Whole- radiosurgical treatment of recurrent previously irra-
brain radiotherapy for brain metastases: evolution or diated primary brain tumors and brain metastases:
revolution? J Clin Oncol. 2017: JCO2017759589. final report of RTOG protocol 90-05. Int J Radiat
143. Robin TP, Camidge DR, Stuhr K, et al. Excellent Oncol Biol Phys. 2000;47:291–8.
outcomes with radiosurgery for multiple brain 155. Eaton BR, LaRiviere MJ, Kim S, et al.
metastases in oncogene-addicted non-small-cell Hypofractionated radiosurgery has a better safety
lung cancer. J Thorac Oncol. 2018;13(5):715–20. profile than single fraction radiosurgery for
144. Sperduto PW, Berkey B, Gaspar LE, et al. A new large resected brain metastases. J Neuro-Oncol.
prognostic index and comparison to three other indi- 2015;123:103–11.
ces for patients with brain metastases: an analysis of 156. Mahajan A, Ahmed S, McAleer MF, et al. Post-
1,960 patients in the RTOG database. Int J Radiat operative stereotactic radiosurgery versus observa-
Oncol Biol Phys. 2008;70:510–4. tion for completely resected brain metastases: a
145. Sperduto PW, Kased N, Roberge D, et al. Summary single-centre, randomised, controlled, phase 3 trial.
report on the graded prognostic assessment: an accu- Lancet Oncol. 2017;18:1040–8.
rate and facile diagnosis-specific tool to estimate 157. Mok TS, Wu YL, Ahn MJ, et al. Osimertinib or
survival for patients with brain metastases. J Clin platinum-pemetrexed in EGFR T790M-positive lung
Oncol. 2012;30:419–25. cancer. N Engl J Med. 2017;376:629–40.
146. Sperduto PW, Yang TJ, Beal K, et al. Estimating sur- 158. Peters S, Camidge DR, Shaw AT, et al. Alectinib ver-
vival in patients with lung cancer and brain metasta- sus crizotinib in untreated ALK-positive non-small-
ses: an update of the graded prognostic assessment cell lung cancer. N Engl J Med. 2017;377:829–38.
Central Nervous System Cancers 129
159. Davies MA, Saiag P, Robert C, et al. Dabrafenib 174. Preston-Martin S, Thomas DC, Wright WE, et al.
plus trametinib in patients with BRAF(V600)- Noise trauma in the aetiology of acoustic neuromas
mutant melanoma brain metastases (COMBI-MB): in men in Los Angeles County, 1978–1985. Br J
a multicentre, multicohort, open-label, phase 2 trial. Cancer. 1989;59:783–6.
Lancet Oncol. 2017;18:863–73. 175. Matthies C, Samii M. Direct brainstem record-
160. Brennan C, Yang TJ, Hilden P, et al. A phase 2 trial ing of auditory evoked potentials during vestibular
of stereotactic radiosurgery boost after surgical schwannoma resection: nuclear BAEP recording.
resection for brain metastases. Int J Radiat Oncol Technical note and preliminary results. J Neurosurg.
Biol Phys. 2014;88:130–6. 1997;86:1057–62.
161. Soliman H, Ruschin M, Angelov L, et al. Consensus 176. Baschnagel AM, Chen PY, Bojrab D, et al. Hearing
contouring guidelines for postoperative com- preservation in patients with vestibular schwannoma
pletely resected cavity stereotactic radiosurgery treated with d surgery. J Neurosurg. 2013;118:571–8.
for brain metastases. Int J Radiat Oncol Biol Phys. 177. Plotkin SR, Stemmer-Rachamimov AO, Barker FG
2018;100:436–42. 2nd, et al. Hearing improvement after bevacizumab
162. McCollough WM, Marcus RB Jr, Rhoton AL Jr, in patients with neurofibromatosis type 2. N Engl J
et al. Long-term follow-up of radiotherapy for pitu- Med. 2009;361:358–67.
itary adenoma: the absence of late recurrence after 178. Noren G. Long-term complications following
greater than or equal to 4500 cGy. Int J Radiat Oncol gamma knife radiosurgery of vestibular schwan-
Biol Phys. 1991;21:607–14. nomas. Stereotact Funct Neurosurg. 1998;70(Suppl
163. Lopes MB, Sloan E, Polder J. Mixed gangliocytoma- 1):65–73.
pituitary adenoma: insights on the pathogenesis of a 179. Regis J, Pellet W, Delsanti C, et al. Functional
rare sellar tumor. Am J Surg Pathol. 2017;41:586–95. outcome after gamma knife surgery or microsur-
164. Sheehan JP, Pouratian N, Steiner L, et al. Gamma gery for vestibular schwannomas. J Neurosurg.
knife surgery for pituitary adenomas: factors 2002;97:1091–100.
related to radiological and endocrine outcomes. J 180. Pollock BE. Management of vestibular schwan-
Neurosurg. 2011;114:303–9. nomas that enlarge after stereotactic radiosurgery:
165. Jagannathan J, Sherman JH, Mehta GU, et al. treatment recommendations based on a 15 year
Radiobiology of brain metastasis: applications experience. Neurosurgery. 2006;58:241–8; discus-
in stereotactic radiosurgery. Neurosurg Focus. sion 241–8.
2007;22:E4. 181. Pollock BE, Driscoll CL, Foote RL, et al. Patient out-
166. Minniti G, Osti M, Jaffrain-Rea ML, et al. Long- comes after vestibular schwannoma management:
term follow-up results of postoperative radiation a prospective comparison of microsurgical resec-
therapy for Cushing’s disease. J Neuro-Oncol. tion and stereotactic radiosurgery. Neurosurgery.
2007;84:79–84. 2006;59:77–85; discussion 77–85.
167. Sheehan JP, Xu Z, Salvetti DJ, et al. Results of 182. Maniakas A, Saliba I. Microsurgery versus stereo-
gamma knife surgery for Cushing’s disease. J tactic radiation for small vestibular schwannomas:
Neurosurg. 2013;119:1486–92. a meta-analysis of patients with more than 5 years'
168. Brochier S, Galland F, Kujas M, et al. Factors pre- follow-up. Otol Neurotol. 2012;33:1611–20.
dicting relapse of nonfunctioning pituitary macroad- 183. Lunsford LD, Niranjan A, Flickinger JC, et al.
enomas after neurosurgery: a study of 142 patients. Radiosurgery of vestibular schwannomas: sum-
Eur J Endocrinol. 2010;163:193–200. mary of experience in 829 cases. J Neurosurg.
169. Chang EF, Zada G, Kim S, et al. Long-term recur- 2005;102(Suppl):195–9.
rence and mortality after surgery and adjuvant radio- 184. Meijer OW, Vandertop WP, Baayen JC, et al.
therapy for nonfunctional pituitary adenomas. J Single-fraction vs. fractionated linac-based ste-
Neurosurg. 2008;108:736–45. reotactic radiosurgery for vestibular schwannoma:
170. Pouratian N, Sheehan J, Jagannathan J, et al. a single-institution study. Int J Radiat Oncol Biol
Gamma knife radiosurgery for medically and sur- Phys. 2003;56:1390–6.
gically refractory prolactinomas. Neurosurgery. 185. Andrews DW, Suarez O, Goldman HW, et al.
2006;59:255–66; discussion 255–66. Stereotactic radiosurgery and fractionated
171. Liu X, Kano H, Kondziolka D, et al. Gamma stereotactic radiotherapy for the treatment of acous-
knife stereotactic radiosurgery for drug resistant tic schwannomas: comparative observations of 125
or intolerant invasive prolactinomas. Pituitary. patients treated at one institution. Int J Radiat Oncol
2013;16:68–75. Biol Phys. 2001;50:1265–78.
172. Losa M, Gioia L, Picozzi P, et al. The role of stereo- 186. Andrews DW, Werner-Wasik M, Den RB, et al.
tactic radiotherapy in patients with growth hormone- Toward dose optimization for fractionated stereotac-
secreting pituitary adenoma. J Clin Endocrinol tic radiotherapy for acoustic neuromas: comparison
Metab. 2008;93:2546–52. of two dose cohorts. Int J Radiat Oncol Biol Phys.
173. Lee CC, Vance ML, Xu Z, et al. Stereotactic radio- 2009;74:419–26.
surgery for acromegaly. J Clin Endocrinol Metab. 187. Nakamura H, Jokura H, Takahashi K, et al. Serial
2014;99:1273–81. follow-up MR imaging after gamma knife radio-
130 S. K. Nath et al.
formations: the relationship of size to outcome. J 225. Schiariti M, Goetz P, El-Maghraby H, et al.
Neurosurg. 1995;82:180–9. Hemangiopericytoma: long-term outcome revisited.
217. Spetzler RF, Martin NA. A proposed grading sys- Clinical article. J Neurosurg. 2011;114:747–55.
tem for arteriovenous malformations. J Neurosurg. 226. Rutkowski MJ, Jian BJ, Bloch O, et al. Intracranial
1986;65:476–83. hemangiopericytoma: clinical experience and treat-
218. Mohr JP, Parides MK, Stapf C, et al. Medical man- ment considerations in a modern series of 40 adult
agement with or without interventional therapy patients. Cancer. 2012;118:1628–36.
for unruptured brain arteriovenous malformations 227. Soyuer S, Chang EL, Selek U, et al. Intracranial
(ARUBA): a multicentre, non-blinded, randomised meningeal hemangiopericytoma: the role of radio-
trial. Lancet. 2014;383:614–21. therapy: report of 29 cases and review of the litera-
219. Kano H, Kondziolka D, Flickinger JC, et al. ture. Cancer. 2004;100:1491–7.
Stereotactic radiosurgery for arteriovenous malfor- 228. Gorman CA, Garrity JA, Fatourechi V, et al.
mations, Part 6: multistaged volumetric management A prospective, randomized, double-blind,
of large arteriovenous malformations. J Neurosurg. placebo-controlled study of orbital radiotherapy
2012;116:54–65. for Graves’ ophthalmopathy. Ophthalmology.
220. Kano H, Flickinger JC, Tonetti D, et al. Estimating 2001;108:1523–34.
the risks of adverse radiation effects after gamma 229. Prummel MF, Mourits MP, Blank L, et al.
knife radiosurgery for arteriovenous malformations. Randomized double-blind trial of prednisone versus
Stroke. 2017;48:84–90. radiotherapy in Graves' ophthalmopathy. Lancet.
221. Moosa S, Chen CJ, Ding D, et al. Volume-staged 1993;342:949–54.
versus dose-staged radiosurgery outcomes for large 230. Prummel MF, Terwee CB, Gerding MN, et al. A
intracranial arteriovenous malformations. Neurosurg randomized controlled trial of orbital radiother-
Focus. 2014;37:E18. apy versus sham irradiation in patients with mild
222. Spina A, Boari N, Gagliardi F, et al. The current role Graves’ ophthalmopathy. J Clin Endocrinol Metab.
of gamma knife radiosurgery in the management of 2004;89:15–20.
intracranial haemangiopericytoma. Acta Neurochir. 231. Bartalena L, Baldeschi L, Boboridis K, et al. The
2016;158:635–42. 2016 European Thyroid Association/European
223. Cohen-Inbar O, Lee CC, Mousavi SH, et al. Group on Graves’ orbitopathy guidelines for the
Stereotactic radiosurgery for intracranial heman- management of Graves’ orbitopathy. Eur Thyroid J.
giopericytomas: a multicenter study. J Neurosurg. 2016;5:9–26.
2017;126:744–54. 232. Mourits MP, van Kempen-Harteveld ML, Garcia
224. Staples JJ, Robinson RA, Wen BC, et al. MB, et al. Radiotherapy for Graves’ orbitopa-
Hemangiopericytoma--the role of radiotherapy. Int J thy: randomised placebo-controlled study. Lancet.
Radiat Oncol Biol Phys. 1990;19:445–51. 2000;355:1505–9.
Head and Neck Cancers
4
Daniel M. Trifiletti, Nicholas G. Zaorsky,
and Henry S. Park
Abstract
This chapter discusses the general manage-
ment of patients with head and neck cancers,
with special focus on principles that guide
radiotherapy management. Several key com-
ponents of trimodality care and radiation field
design are discussed.
D. M. Trifiletti (*)
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
N. G. Zaorsky
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
H. S. Park
Yale School of Medicine, New Haven, CT, USA
Dissecon
Dissectionname
name Lymph nodes
Lymph nodesMuscles, vessels,
Muscles, vessels,
removed
removed nerves removed
nerves or or
removed
preserved
preserved
Radical neck I-V Removes SCM, IJV,
dissection (RND) CN XI
Modified radical neck I-V Preservation below
dissection (MRND)
transverse body
process
VIII Parotid Zygomatic Angle of Post mandibular Ant of SCM, Styloid
arch, EAC mandible ramus, post belly of process
masseter, digastric
medial pterygoid
musc
IV
Head and Neck Cancers 137
External canal, post-auricular Tympanic n and Sup vagal N/A Sup vagal X
plexus ganglion ganglion
Auricular n (Arnold)
Nasal mucosa Vidian n Geniculate Internal N/A VII
Post ethmoid sinus Greater sup petrosal n ganglion acoustic
Sphenoid sinus meatus
Soft palate
Inf ear, pinna, post-auricular Post auricular n Geniculate Internal N/A VII
ganglion acoustic
meatus
138 D. M. Trifiletti et al.
Head and Neck Cancers 139
PTV Expansion
Flap Coverage –– 0.3–0.5 cm CTV to PTV margin if using daily
KV and/or CBCT
–– In PORT for the head and neck, the entire
post-op bed and flap should be covered in case
of ECE. PET-CT
Mastication Immunotherapy
–– Lateral pterygoid originates from lat ptery- –– Checkmate 141, Ferris, UPMC, 2016. M HN
goid plate and inserts onto the neck of con- SCC w dz. progression 6 m after platinum
dyloid process of the mandible –> opens chemo. Nivo > standard tx, 1-year OS 36% vs
jaw 17%, response rate 13% vs 6%. Response in
–– Masseter and medial pterygoid –> closes jaw both p16+/−. Nivo had stable physical and
social function, which worsened w chemo. If
>1% PD-L1 expression, then favorable OS,
Speech and Swallow HR 0.55. No further benefit if >5% of >10%
expression.
–– Baseline eval for speech and swallowing
dysfunction.
–– Dysphagia/aspiration-related structure Contouring Normal Structures
(DARS): pharyngeal constrictors, supraglot-
tic, and glottic larynx. Brainstem
–– For patients receiving CRT, SLP improves Spinal cord
swallowing at up to 9 months of follow-up. Dorsal vagal complex (DVC), area pos-
trema – Level of inferior cerebellum (on sag),
contour two to three slices of posterior brainstem
PEG or NG Tube going inf (1 above level of hemisphere, 1 at, and
1 below level of hemisphere)
–– Not routinely recommended Cochlea: lucency anterior to IAC on bone
–– Higher G-tube dependence windows. Prefer slices ≤1 mm. Avg volume
–– Higher late esophageal stricture 0.13–0.56 mL. Note cochlea has no known
–– Consider w severe weight loss (5% in 1 month, threshold for sensorineural hearing loss, rec to
10% in 6 months), dehydration, dysphagia, keep dose ALARA.
anorexia, significant comorbidities, aspiration Vestib complex: lucency post to IAC
risk, and large RT fields Pharyngeal constrictors: three sets of con-
tours. Contour 3 mm slice of posterior pharynx.
Sup: when pterygoid plates disappear, superior
Treatment Initiation PC begins. Goes inf to sup hyoid bone.
Middle: spans hyoid bone (sup to inf). Inf:
–– Ideally start definitive therapy within 4 wks start when hyoid no longer visualized and ends at
and post-op therapy with 6 wks the esophagus. Make wider near the inferior
aspect (cricopharyngeus muscle joins pharynx to
Induction Chemo esophagus and is wide)
–– Current indications: Possibly T4b or N3
non-oropharynx cancers, metastatic NPX
cancer to gain control over DMs prior to LR
RT, borderline/unresectable sinonasal neuro-
endocrine tumors, pt unable to lie flat
–– In general, not recommended over concurrent
CRT. PARADIGM and DeCIDE negative for
OS advantage of induction vs concurrent.
Head and Neck Cancers 141
When to
Level Term Sup Inf Ant Post Lat Med Drainage from treat
IA Submental Mandible Hyoid Mandible, Hyoid Ant digastric -- Skin of the chin,
platysma mid-low lip, tip of the
tongue, ant FOM, ant
oral tongue, and
mandibular alveolar
ridge
IB Submandibular Sup edge of Mid body Mandible, Post-edge Mandible, Ant IA, lower nasal cavity,
SMG hyoid platysma SMG platysma digastric hard/soft palate,
maxillary and mandib
alveolar ridges, cheek,
lips, ant tongue, OC, ant
NC, midface, SMG
IIIIA: ant to the Upper 1/3 of IJV Inf edge C1 Inf edge Int carotid, post Post edge Medial SCM Int carotid, “Grand central station”: Standard
post IJVIIB: and CN XI transverse hyoid edge SMG SCM levator face, parotid, SMG, IA, hemi -neck
post to the post process scapulae IB, VIIa, NC, NPX, OC,
IJV OPX, LX, HPX, EAC,
middle ear
IIB: OPX, NPX >> OC,
LX, HPX
III Middle 1/3 IJV Inf edge Inf edge Ant edge SCM Post edge Medial SCM Int carotid, NPX, OC, OPX, LX,
hyoid cricoid SCM paraspinal HPX
M
IV Lower 1/3 IJV Inf edge of Midway b/w Ant edge SCM Post edge Medial SCM Int carotid, NPX, OPX, LX, HPX.
IVa: low and medial sclav cricoid cricoid and SCM paraspinal Infrequently: OC
juguelar clavicle head M
IVb: medial
sclav
V Post triangle Sup edge Trans cervical Post edge SCM Trap Platysma, Paraspinal NPX Post-op,
Va: upper post hyoid vessels skin M node-
triangle positive
Vb: lower post hemi rt neck
triangle
Vc: lat sclav
D. M. Trifiletti et al.
VI Ant compartment Inf edge Sup edge Platysma, skin Ant and lat Thyroid and None or thyroid, LX (subglottic
VIa: ant jugular thyroid cart manubrium trachea SCM midline involvement), extension
VIb: through the thyroid
prelaryngeal, cartilage, HPX
pretracheal,
paratracheal
VII (older) Sup edge Innominate Manubrium Ant and lat Carotids or ---
manubrium vein trachea mediastinum
Head and Neck Cancers
VIIa/RP Retropharyngeal BOS Sup edge Pharyngeal Prevertebral Med edge int Midline Paranasal sinuses If gross
hyoid mucosa Ms carotid positive or
PPW
involved.
Anything
with
“pharynx” in
name of dz
VIIb/RS Retrostyloid BOS Inf edge C1 Retrostyloid M BOS or V Parotid Lat edge int Post-op,
transverse body carotid cLN+
process hemi-neck
VIII Parotid Zygomatic Angle of Post mandibular Ant of SCM, Styloid
arch, EAC mandible ramus, masseter, post belly of process
medial digastric
pterygoid musc
Supraclav Inf border Sup edge SCM and Carotid and Clavicle or SCM,
level 4–5 manubrium clavicle paraspinal fat space carotid,
MS med
clavicle
SMG typically none bc Primary
encapsulated SMG cancer
Parotid
143
144 D. M. Trifiletti et al.
Head and Neck Cancers 145
cancers are skin terminal sulcus, aka aryepiglottic folds, superiorly and G3; adenoid;
cancers, have vallate papillae). esophageal mucosa Clark IV+
different tx inferiorly)
Borders: hyoid bone
or tip of epiglottis
(sup); bottom of
cricoid cart or
cricopharyngeus
muscle (inf)
T1 NPX, OPX, or Limited to 1 subsite*, no £2 cm w/o £2 cm (new in AJCC £2 cm 1 subsite*, normal Normal VC Subglottis One subsite**, £2 cm T1a - £1cm £1mm thick £2 cm £2 cm
nasal cavity maxillary sinus bony invasion extraparenchy- 8: and £5 mm DOI) VCs movement only T1b – 1-2 cm T1a – no ulcer, <
without bone mal extension (T1a one cord, T1b 0.8mm
erosion T1b –ulcer or
both)
0.8-1.0 mm
T2 Parapharyngeal Invading hard 2+ subsites* 2-–4cm w/o 2–4cm or 5-£10 mm 2–4cm 2+ subsites or Supra/subglottis or VCs w 2+ subsites, 2–4cm 2–4 cm, capsule 1.01-2mm thick 2–4 cm 2–5cm
space (AJCC 8: palate, middle (doesn’t matter if extraparenchymal glottis, or region normal intact T2a - no ulcer
+/- med or lat nasal meatus invading hard extension outside supraglottis mobility T2b - ulcer
pterygoid, palate or middle
(BOT [e.g. hot VCs w impaired VCs w
prevertebral nasal meatus)
muscles, carotid potato voice], mobility (previously impaired
space) vallecula, medial T2b) mobility
wall pyriform)
T3 Bony structures, Posterior wall of Maxillary sinus, >4 cm or >4 cm or >10 -£20 >4 cm or lingual Fixed cord T3a - > 4 cm but 2.01–4mm thick >4cm or minor >5cm
skull base, or maxillary sinus, medial orbital extraparenchymal mm DOI surface of limited to thyroid T3a - no ulcer bone erosion,
paranasal sinuses subQ, orbital wall, palate, extension epiglottis Post-cricoid area, >4 cm, esophagus T3b – Gross T3b - ulcer or c/p/rPNI or
wall, pterygoid cribriform plate paraglottic space, Paraglottic space extrathyroidal deep
fossa, ethmoid (“COMP”) pre-epiglottic space extension into
Inner thyroid cartilage strap muscles
T4a Intracranial,any T4a – anterior Anterior orbit, Skin, mandible, Non-lip:>20 mm DOI, (p16- is split a/b, Through thyroid cartilage Cricoid, Thyroid/cricoid T4a – subQ, >4 mm thick Gross cortical Into bone,
CNs, orbit, skin, skin, pterygoid ear canal, CN VII cortical bone, however p16+ is thyroid cartilage, hyoid, larynx, trachea, T4a - no ulcer bone invasion muscle,
infratemporal pterygoid plates, plates, cranial maxillary sinus, deep lumped cartilage central compartment esophagus, fascia,
fossa, infratemporal fossa, muscles of tongue together into T4 invasion recurrent cartilage
hypopharynx, fossa, cribriform sphenoid/frontal (genio, hyo, stylo, alone) larynx, (AJCC 7:any laryngeal, or any
orbit, parotid, plate, sinuses (“POS palatoglossus). Note: deep tongue cartilage was anaplastic
masticator space, sphenoid/frontal CSF”) superficial bone does muscles, medial T4a)
soft tissue sinuses not count pterygoid, hard
beyond lat lip: through cortical palate, Beyond larynx (e.g.esophagus, soft tissues of neck,
pterygoid, bone, inf alveolar N mandible extrinsic tongue muscles, strap muscles, thyroid)
parotid gland, (V3), FOM, skin of *No mention of
masseter muscle face (skin, nose) PNI
T4b Orbital apex, Orbital apex, Skull base, Masticator space, Lateral pterygoid Prevertebral space, carotid, mediastinum T4b – T4b – ulcer BOS
dura, brain, dura, brain, pterygoid plates, pterygoid plates, (e.g. trismus < prevertebral
middle cranial middle cranial carotid skull base, encases 4 cm),pterygoid fascia, carotid,
fossa, CNs other fossa, CNs other carotid plates, lateral mediastinum
than V2, than V2, nasopharynx, (anaplastic out of
nasopharynx, nasopharynx, skull base, thyroid)
clivus clivus carotid
147
––
148
N2 Bilateral, ≤6cm, above inf Contra or bilateral, all ≤ > 4 nodes, any location N2a: Single ipsi, 3–6 cm, no Single ipsi, 3–6 cm, no ECE; or 2–3 nodes In transit mets, In transit mets,
cricoid 6 cm ECE ipsi/contra < 3 cm with ECE+ N2a – clinically occult no LNs no nodes
N2b – at least one clinically apparent
N2b: Multiple ipsi, ≤6cm, no Multiple ipsi, 3-6 cm, no ECE
N2c – 1 clinical node, and in-
ECE
transit/satellite/microsatellite
N2c: Bilateral or contralat, Bilateral or contralat, 3-6 cm,
≤6 cm, no ECE no ECE
N3 >6 cm and/or below inf cricoid Any node > 6 cm N3a: >6 cm, no ECE >6 cm, no ECE ≥4 nodes In transit mets In transit mets
N3b: Any clinically overt ECE Single >3 cm with ECE; or N3a - ≥4 nodes, all occult AND nodes AND nodes
multi nodes with any ECE+ N3b - ≥4 nodes some felt, or matted
nodes
N3c –2+ clinical nodes, and in-
transit/satellite/microsatellite
D. M. Trifiletti et al.
Head and Neck Cancers 149
Overview Trials
–– 2000 cases/yr (1/3 asymptomatic), 4/1 –– COMS 1997: obs small tumors. Five-yr OS
million. 94%, 33% progressed
–– 98% Caucasians. –– COMS 28, 2006: (COMS
–– Ocular melanoma is the #1 eye malignancy of medium)→enucleation vs eye plaque. Same
primary cancers. 12-yr OS (~20%), 13% of plaque pts ended up
–– Usually arises from choroid (85%) > adnexa getting enucleations 2/2 tumor or pain
(10%) > conjunctiva (5%). –– COMS medium, Quivey 1993: retrospective
–– Melanocytes of uveal stroma (neural crest ori- I-125. 13% local failure at 5y, due to tx failure
gin) give rise to ocular melanoma. or pain from BT complications
–– 1–2% present w DM. Usually mets to liver –– COMS large: preop RT does not improve DFS
(90%). Twenty percent will had DM over 5y, or OS between two tx groups.
50% at 15y.
–– BAP1 inactivation found in most ocular Plaque Technique
melanoma. –– I-125 seeds, 0.7–1 Gy/h. Treatment time
4–7 days.
Anatomy –– Ru-106, a beta emitter may be used, because it
–– Layers of globe: outer fibrous layer (sclera), has limited dose penetration vs I-125, so it
middle vascular layer (choroid), inner nerve results in less toxicity and is easier to insert.
layer (retina) –– 85 Gy to apex (if less than 5 mm apex→Rx to
–– Macular important for color vision 5 mm).
–– Optic disk is 2 mm medial to macula, 1.5 mm –– 2 mm around tumor.
diameter –– LC 93%.
Pathology
WHO Old WHO keratin EBV assoc? Differentiated Notes
I Keratinizing N Differentiated Smokers,
radioresistant,
U.S.
II IIA Non-keratinizing Y Differentiated
III IIB (EBV associated) Y Undiff,
lymphoepithelioma
Basaloid Rare
Anatomy Workup
–– Borders –– H/P: hearing loss, otalgia; nasal congestion,
–– Ant: choanae epistaxis
–– Post: clivus, C1, C2 • CN deficits (vision) or trismus.
–– Sup: sphenoid sinus • First nerve affected: CN VI through fora-
–– Inf: soft palate men lacerum, then V1 and V2
–– Lat: eustachian tube, torus tubarius, fossa of • If spread through foramen rotundum: V2
Rosenmuller (deep is parapharyngeal space) and V3
Head and Neck Cancers 151
–– Indications for adjuvant radiation therapy –– Add chemo for +margin, ECE, SNUC
include: –– 60–66 Gy to primary, 50–54 to necks
–– T1N0 w positive margins –– T2+/N + = Sx → PORT
–– Consider for T2N0 –– 66/60/54 (same for all nasal cavity/parana-
–– T3 or T4 sal sinus)
–– Perineural invasion • Bilateral IB-II
–– Adenoid cystic histology –– N+: IB, RSS/II-V
–– For any N+, add neck RT –– *posterior 2/3 involvement: cover RP nodes
–– Radiation therapy alone can be used as a pri- –– *nasal vestibule involvement: cover facial
mary modality for inoperable patients. nodes
–– Unresectable: chemo-RT to 70 Gy, cisplatin
Chemo? –– When to cover elective LNs in N0 dz: (1)
–– Sinus cancers are not included in post-op CRT involvement of NPX bc of high rate of
trials, but indications (i.e., margins, ECE) usu- lymphatic spread; (2) T3/4; (3) PNI, G3–4,
ally extrapolated from SCC histology. olfactory neuroblastoma
–– LN volumes controversial
Historical Treatment –– Consider alt fractionation for tissue sparing
3D-CRT w 4 field: AP photons, wedged opposed –– Accelerated: BID once per week (6 fx/wk)
la photons, and interorbital anterior electron field. –– Concomitant boost: BID last 2 wks
Wedge pairs were also used. –– Hyperfractionation: BID throughout
–– CTV1 = GTV + 1–1.5 cm margin, modi-
Sinus Sim/Planning (Based on Nancy Lee’s fied at natural barriers + ENI (bilateral
Recs) RP + level II-IV)
–– Dentistry, supine, mask, eyes straight, bite –– Phase I (initial volume): 48.4 Gy in 44 fx at
block. 1.1 Gy BID
–– Diagnostic MRI co-registered with planning –– CTV2 = GTV + 0.5–1 cm margin, modi-
CT for contouring. fied by natural barriers.
–– Standard is 1.8–2 Gy daily. • Phase II (small field boost): 24.2 Gy in
–– 50 Gy if neoadjuvant 22 fx @ 1.1 Gy BID
–– 60–66 if post-op • PTV = CTV + 3 mm
–– 70 Gy if definitive • Total PTV dose: 72.6 Gy in 66 fx @
–– Esthesioneuroblastoma can do cist/estopo x2 1.1 Gy BID
cycles, then 50 Gy (with ENI), then • IMRT preferred for normal tissue
resection. sparing
–– Dose limits: • Preop RT or CRT to 50 Gy accepted
–– Brainstem: 54 Gy (60 Gy max)
–– Optics: 54 Gy SNUC
–– Retina 45 Gy, lens 10 Gy –– A, B, C = Surgery → PORT (for low-grade A,
–– Parotid mean < 26Gy can consider RT alone)
–– Pituitary/thyroid (62% develop hormone –– B, C = + bilateral RP, IB-II
deficiencies) –– Consider adj cis/etop bc SRBCT
NCCN
–– Resectable: surgery + RT for T3, T4,+margin,
PNI, ACC, ethmoid (all ethmoid tumors need
adj RT)
156 D. M. Trifiletti et al.
T1 T2 T3 T4a T4b
N0 I II III IVA
N1 III III III IVB
N2 IVA
N3 IVB
M1 IVC
• T1 – ≤2 cm and ≤5 mm DOI (note DOI new (anterior to terminal sulcus, aka vallate
in AJCC8 vs 7) papillae).
• T2 – 2–4 cm or >5 – ≤10 mm DOI • The circumvallate papilla differentiates the
• T3 – >4 cm or >10 mm DOI oral tongue from BOT; on axial CT it is
• T4 separated by uvula.
• T4a (everything but lip) – cortical bone, –– Extrinsic tongue muscles: genioglossus, sty-
maxillary sinus, deep muscles of tongue loglossus, palatoglossus, hyoglossus
(genio, hyo, stylo, palatoglossus). Note:
superficial bone does not count. 5y OS
• T4a (lip) – through the cortical bone, inf Lip 89%
Oral tongue 65%
alveolar N (V3), FOM, skin of face (chin,
FOM 52%
nose)
Gum 59%
• T4b – masticator space, pterygoid plates,
skull base, encases carotid
LN drainage
Single ipsi node Single ipsi node Lip
cN1 ≤3 cm, no ECEpN1 ≤3 cm, no ECE –– Upper (10%, worse prognosis) → Facial, IB, II
cN2 pN2
–– Lower (90%) → IA, IB, II
cN2a Single ipsi, pN2a Single ipsi, 3–6 cm,
3–6 cm, no ECE no ECE; or <3 cm –– Oral commissure → 15% LN risk
with ECE+
cN2b Multiple ipsi, pN2b Multiple ipsi, 3–6 cm, Alveolar Ridge
3–6 cm, no ECE no ECE –– Lower (80%), often involves molars, presents
cN2c Bilateral or pN2c Bilateral or contralat, as ill-fitting dentures
contralat, 3–6 cm, no ECE
3–6 cm, no ECE –– Alveolar ridge w/ PNI need to cover length of
cN3 pN3 IAN
cN3a >6 cm, no ECE pN3a >6 cm, no ECE
cN3b Any clinically pN3b Single >3 cm with RMT
overt ECE ECE, or multi-nodes –– Anterior spread along alveolar ridge, IAN
with any ECE+
–– Pterygomandibular raphe → masticator space
and FOM
Anatomy
Oral Tongue
–– Subsites: mucosal lips (most common), FOM –– LN drainage
(2nd most common), buccal mucosa, superior • Tip of tongue = IA
alveolar ridge (aka upper gingiva), inferior • Anterior tongue = IB and III
alveolar ridge (aka lower gingiva), retromolar • Posterior tongue = IB and II
trigone (RMT), hard palate, ant 2/3 tongue
158 D. M. Trifiletti et al.
–– Anterior and midline = more likely to be –– MACH-NC update, Blanchard, 2011: benefit
bilateral broken down by dz sites:
• 5% bilateral overall • OC: 9%, NSS
• If N+ ipsilateral, 30% bilateral • OPX: 8%, SS
• LX: 5%, SS
Floor of Mouth • HPX: 4%, NSS
–– DOI ≥ 1.5 mm = high risk of LN involvement,
therefore needs neck dissection Post-op RT, CRT
–– LN drainage = IB –– Ang 2001: OC SCC: n = 213. Aim to deter-
–– Often invades oral tongue, alveolar ridge, and mine validity of previously reported path risk
mandible factors. All pts had surgery. RFs were then:
–– Mylohyoid muscle divides sublingual space primary site (OC), +margin, PNI, ECE, num-
from submandibular space ber and location of post LNs. Pts w no risk
factors (low risk) observed. Pts w 1 RF besides
Workup ECE (intermed risk) got 57.6/32. Pts w 2 RFs
–– H&P or ECE (high risk) randomized to either CFRT
–– Tobacco, ETOH, poor hygiene, betel/areca 63/35 in 6.5 w vs AFRT 63/35 in 5 wks. Risk
nuts groups were validated and had ↑failure with-
–– Oral leukoplakia (10% risk) out post-op RT. For high risk, trend for
–– Erythroplakia (30% risk) improved LRC and OS for AFRT vs CFRT.
–– CN XII, motor; CN V, sensory; CN VII, taste –– EORTC 22931: operable stage III/IV H&N
(BOT CN IX taste) SCC of OC, OP, larynx, hypopharynx. pT3–4
–– Otalgia: auriculotemporal nerve (CN V3) any nodal, except T3N0 of LX, w R0, or T1–2
–– palpation, FNL, CN exam, bx, labs w N2–3 M0. Patients w Stage T1–2 N0–1 w
–– MRI + CT, CT chest, +/− PET unfavorable path findings (e.g., ECE, R+,
–– Dentistry PNI, LVI) also eligible. Randomize: post-op
66/33 +/− concurrent cisplatin (100 mg/m2
Studies Q3wks). CRT won: 5-yr DFS 36→47%, OS
Altered Fractionation (No Chemo) 40→53%, LRC 69→82% (DM unchanged
–– RTOG 9003: advanced H&N SCC→ 70/35 ~25%). ↑toxicity (21→41%).
(std) vs 81.6 at 1.2 BID (hyperfrac) vs 67.2 at –– RTOG 9501: operable H&N (≥2 LN, ECE,
1.6 BID split (split) vs 72 w/ last 12fx BID +margin): 66/33 +/− concurrent cisplatin
(conboost). Hyperfrac and Conboost won (100 mg/m2 Q3wks): CRT won: 2 yr DFS
(LRC 54%, DFS 39%, OS 53%). ↑toxicity 43→54%, LRC 72→82%, trend for OS.
–– MARCH meta-analysis: 6515 pts. 3.4% OS ↑toxicity.
benefit at 5 years for altered fractionation, –– Meta-analysis of 9501 and 22,931: CRT
mostly for young pts improved OS, DFS and LRC for ECE
or + margins. Trend for stage III/IV, PNI,
Chemo-RT LVSI, low neck nodes.
–– MACH-NC meta-analysis, Pignon, 2009: –– MDACC: Peters, IJROBP, 1993, RCT to
17,346 pts from 87 RCTs. Median 5.6y fol- determine optimal dose. Pts stratified by
low-up. At 5y, 4.5% OS benefit with any che- adverse path features (e.g., ECE, SMs, 2+
motherapy (neoadjuvant, concurrent, LN+, T stage, PNI, OC primary) into low and
adjuvant), HR 0.88. The greatest benefit with high risk. Subsequently, low-risk patients
concurrent (6.5%). Platinum-monotherapy is observed. Intermediate-risk patients random-
a gold standard, no benefit if age >71. ized to 57.6 Gy/32 or 63 Gy/35; high risk to
Head and Neck Cancers 159
63 Gy/35 or 68.4 Gy /38. <54 Gy had higher though volume should be kept as small as
failure. ECE >63 Gy. # adverse features (0–1; possible
2–3; ECE; 4 or greater; in order or worsening –– CTV60 = preop GTV + entire post-op bed
prognosis) predicted for poorer LRC. 4+ neg- including residual tongue, BOT, FOM, glosso-
ative factors had a similar LRC to ECE. tonsillar sulcus, and anterior tonsillar pillar
(CTV P):
Post-op RT Timing • For R0 resection in pT4 mandible, go 1 cm
–– Daly, Stanford, IJROBP, 2011: 37 pts. Worse into bone
LC if initiating >6w after vs <6w: LC was • For midline OC tumor, always treat contra IB
80% vs 40%. Also noted difference in treat- • If pN+, include ipsilateral hemineck (CTV N)
ment package time of > vs <12 w. –– Note: if using LAN to 50 Gy, can do
10 Gy ipsi boost on involved side
Surgery and Pathology • CTV54–56 = low-risk subclinical disease,
–– Finding PNI raises risk of LR by 20–40%. often contralateral neck, ipsi RSS
PNI likely not sign of neurotropism (like in –– Dose limits:
melanoma or adenoid cystic) but for SM. –– Cord <45 Gy max
–– Finding LVI likely increases risk of LNMs –– Parotid mean <26Gy
(similar to cervical cancer). –– Larynx mean <43.5 Gy
–– Mandible <70 Gy max
Neck Dissection
–– For oral SCC neck dissection needs at least 16 Management
LNs
–– Tata Memorial / D’Cruz, NEJM, 2016: ipsi General Oral Cavity
elective ND vs therapeutic ND (at time of
relapse) for cT1–2N0 OC SCC. n = 500. At 3 T1–2N0
y, END had improved OS 80% vs 68%. Rate –– Surgical resection preferred
of LN positivity in END arm is 30%. –– No commissure involvement: surgery (Mohs
Correlated with DOI: 3 mm = 6%, for or WLE). Indications for adj RT: + margin,
4 mm = 17%. Rate of adverse events 6.6% vs PNI, LVI. Oral Tongue requiring LND: >3 mm
3.6%. Thus, need ND if 3+ mm DOI. Thirty- DOI, grade 3, +LVSI, recurrence
three percent of END group recurred vs 58% –– Adequate margin in OC SCC: 1 cm (1.5 cm
in the therapeutic ND recurred. Of the thera- for tongue)
peutic ND pts who recurred, 53% died of PD, –– Commissure involved: RT 66–70 Gy to pri-
emphasizing limited success of salvage. mary only
–– EBRT and BT generally not done as definitive
Sim/Planning therapy for oral cavity because of morbidity.
–– Dental eval, PEG?, aquaplast, supine, bite –– EBRT + brachy boost (50Gy + 21Gy/7fx HDR
block boost)
–– Sim: supine on head rest, neck extended, –– For HDR, must be >1 cm from mandible
shoulders down, immobilization with thermo-
plastic mask, wire scars/nodes, IV contrast, T3–4N0
Iso typically placed at thyroid cartilage notch. –– Surgical resection +/− neck dissection
Bolus for tumor invading skin/violated neck –– Concurrent CDDP chemo with RT 70 Gy
w/ECE standard frac, treat full neck inc LAN for
T4N0
Volumes –– Altered frac RT 70 Gy alone for T3
–– CTV66 = include areas at very high risk of –– Bone/nerve invasion = Surgery + RT (RT
recurrence such as close/+mgn and ECE, alone = 90% LF)
160 D. M. Trifiletti et al.
p16+ Oropharynx neck staging cN2a Single ipsi, 3–6cm, pN2a Single ipsi, 3–6 cm, no ECE;
no ECE or <3 cm with ECE+
cN1 Ipsi nodes, all ≤6 cm pN1 ≤4 nodes, any cN2b Multiple ipsi, 3–6 cm, pN2b Multiple ipsi, 3–6 cm, no
location no ECE ECE
cN2c Bilateral or pN2c
cN2 Contra or bilateral, pN2 >4 nodes, any Bilateral or contralat, 3–6 cm,
contralat, 3–6 cm, no
all ≤6 cm location ECE
no ECE
Anatomy Anatomy
–– Subsites: soft palate, palatine tonsils, tonsillar
–– Vallecula: part of boundary bw LX and OPX pillars, base of tongue, pharyngeal wall
–– ventricle / Morgangi’s sinus: part of LX bw –– Borders: superior soft palate to superior hyoid
TVC and FVC bone (floor of vallecula)
–– Ear pain?
HPV+ vs HPV– –– Oral tongue: auriculotemporal nerve
–– RTOG 0129, 0522 patients (CN V)
–– Similar rates of DM, but improved OS for –– BOT: Jacobson’s nerve (CN IX) – inner-
HPV+ vates the post 1/3 of the tongue and tonsil-
lar fossae/pillars and the ear (via tympanic
nerve)
–– Larynx/HPX: Arnold’s nerve (CN X)
SS in yellow HPV+ HPV– pval
Median time to 8 7 NS
progression, m
Work-up
patients w DM, % 46 43 NS
–– H&P, palpation, FNA, CN exam, biopsy (p16)
First site of progression 55 Similar
–– MRI + CT, CT chest, +/− PET, dentistry
LRR only, % patterns
First site of progression 39 of Surgery
DM only, % failure –– Need 18+ LNs for LN dissection, diagnostic,
LRR+DM, % 2 and therapeutic (Ebrahimi, Cancer, 2010;
DM 2y after dx 13% 4% SS Divi, Cancer, 2016).
2y OS, % 55 28 SS –– For tonsil primary with cN0 neck, the rate of
MST after progression, 2.6 0.8 SS contra LNs is 10–15%.
y
Preop vs Post-op RT
–– RTOG 7303 HN Surg, 1987, advanced
HPV+ SCC HNSCC, preop RT (50 Gy) vs. post-op RT
–– Younger, nonsmokers (~60% of new (60 Gy) vs. definitive RT alone (65–70 Gy)
cancers) +/− salvage surgery (the third arm was not
–– subtypes 16(80%), 18: ↑nodes, ↑mets offered to patients with cancer of the supra-
–– E6→↓p53; E7→↓Rb→↑p16 glottic larynx or HPX). Post-op RT vs. preop
–– Chin / Wash U St Louis, 2011: p16+ OPX w RT vs. RT alone had marginally improved
ECE or close/R+. 60 vs 66Gy IMRT. No dif- OS (36% vs. 33%, vs. 33%, NS) and LRC
ference in LRC 2 years 99% vs 99% (65% vs. 48% vs. 38%). Thus, preop RT typ-
–– RTOG 0129: CFX + concurrent chemo (cis ically not used for SCC. The benefit of RT is
100 mg/m2 d1,22,43) vs AFX + concurrent likely secondary to preservation of the surgi-
chemo (cis 100 mg/m2 d1,22). Three-year OS cal site. The 60 Gy dose became the “stan-
was 70% vs 64% (NSS). ECOG 0–1. No diff in dard” Fletcher dose for subsequent HN
PFS, patterns of failure, acute/late tox. Subgroup RCTs.
analysis: better 3-yr OS for HPV+ (57→82%)
• RPA: (1) HPV; (2) PYs: 10, 20 cut points; Altered Fractionation (RT Alone, NO
(3) T-Stage; (4) N-stage CHEMO)
• Low risk: HPV+ and < 10 PY; or HPV+ –– IAEA ACC, Overgaard, 2010. 9 centers.
and > 10 PY and N0-2 N = 908. Stage I-IV larynx, pharynx, OC. No
• High risk: HPV- and > 10 PY; or HPV- NPX or stage I glottic. Pts got either 6fx/w for
and < 10 PY and T4 40d vs 5fx/w for 47d. Total dose 66–70/2.
• Three-year OS for 93% for low, 71% for 5-year DSS 50% vs 40%, LRC 42% vs 30%.
intermed, and 46% for high-risk groups OS 35% vs 28%. favoring ACC.
Head and Neck Cancers 163
–– RTOG 00–22: T1–2N01 OPX SCC: 66 Gy 93% vs 85% vs 73%. cCR 27% vs 40% vs
in 30 fx (@2.2). Accelerated hypofrac 49%. Concurrent CRT became SOC for unre-
IMRT. Treatment to primary dz and bilat sectable dz after this trial.
neck. RT was 66/2.2, over 6w. PTVs –– GORTEC 99–02, Bourhis, 2012: Stage III/IV
received 54–60 Gy at 1.8–2 Gy/fx w SIB. OC, OPX, HPX, LX. 66% OPX, 55% T4,
2 yr LRC 91% (N staging clinical only), 25% N2c. n = 840. SCC. Randomize: (1)
DFS 82%, OS 96%. 70/35+ carbo/5FU vs (2) 70/30
–– RTOG 9003: advanced HNSCC→ 70/35 (std) (6w) + carbo/5FU vs (3) very accelerated RT
vs 81.6 @1.2 BID (hyperfrac) vs 67.2 @1.6 (VART), 64.8/18, no chemo. Best outcomes
BID (split) vs 72 w/ last 12fx BID (conboost). and toxicity in conventionally fractionated +
Hyperfrac and Conboost won (LRC 54%, chemo. ↑acute tox, feeding tube dependence
DFS 39%) though ↑toxicity. No difference in in 64.8/18 arm. Pts receiving accel frac +
OS 53%. chemo only received 2c chemo. Conclusion is
–– EORTC 22791: T2/3 oropharynx 70/35 vs not to do altered fx + chemo or VART bc no
80.5/70 @1.15 BID. 1980–87. n = 356. T2–3 change in outcome, but worse tox.
N0–1 < 3 cm, age < 75, KPS >60. No BOT in –– MACH-NC meta-analysis: see OPX section.
this trial. Hyperfrac ↑LRC 40→59%, OS
31→47% at 5y. Greatest benefit in T3N0, RT +/– Cetuximab
T3N1. Acute tox worse; 7.5 vs 4.5% had tx –– Bonner 2006: advanced OP, larynx, hypophar-
interruption. Thus, for pts unfit for chemo, ynx: RT vs CRT (cetuximab). RT was 70/35,
hypofx an option. 72–76.8/1.2 BID, or 72/42 concomitant boost,
–– MARCH meta-analysis, Baujat, 2010: 6515 with 32.4/1.8 followed by 21.6/1.8 in AM and
pts 3.4% OS benefit and 6.4% LC benefit at 18.0/1.5 in PM. RT per tx institution. Cetux
5 yrs for altered frac, mostly in young pts., was initial loading of 400 mg/m2 1w prior to
mostly with the use of hyperfractionated RT RT, then weekly 250 mg/m2 during RT. CRT
(rather than accelerated RT) won, 3-yr LRC 34→47%, OS 45→55%. Rash
–– DAHANCA, 2003: n = 1476. RT in 5 vs. with cetuximab, and those pts. w G2–4 rash
6 weeks to 66–68/2. 5-yr LRC 70% vs 60% for did better: MST 69 vs 26 m, HR 0.5. Delayed
accelerated. DFS: 73% vs 66%. No OS differ- accelerated CB + cetux did the best.
ence. Acute mucositis worse w accelerated:
53% vs 33%, but all pts healed within 3 months CRT +/– Cetuximab
of start. No difference in severe late tox. –– RTOG 0522 (Ang, 2014): T2N2–3M0 or T3–4
any N SCC OPX, HPX, LX. Randomize to (1)
Chemo-RT RT (either 72 Gy/42 over 6 w, BID RT for 12
–– GORTEC 9401, Denis, 2004: stage III/IV oro- d; or 70 Gy/35) + cis 100 mg/m2 vs (2)
pharynx: 70/35 +/− carbo/5-FU. Carbo RT + cis + cetux 400 mg/m2. Three-year OS
70 mg/m2 and 5-FU 600 mg/m2 on d1,22,42. (73% vs 76%), PFS (61% vs 59%), LRF (20%
CRT improved 5-yr LC 25→48%, DFS vs 26%), DMs (13% vs 10%) were similar.
15→27%, OS 16→23%. Cis-cetux-RT had more interruptions (27% vs
–– INT, Adelstein 2003: unresectable stage III/ 15%) and G3 mucositis (43% vs 33%).
IV H&N SCC. Excludes NPX, parotid, para- • pCR is 86% for HPV associated OPX SCC.
nasal sinus. (1) RT 70 /2 (2) + cisplatin –– MSKCC retrospective (Koutcher, 2011):
(100 mg/m2 Q3wks) (3) split course CRT w 3 2 year LRF of cis-RT vs. cetux-RT is 6% vs
courses 4-day CI 5-FU 1 g/m2 + cisplat bolus 40% – 8x the failure! 2-year OS 92% vs 67%.
75 mg/m2 on d1 q4w. RT was 30, then 30–40
Gyk, for total of 60–70 Gy. RT break was Post-op RT +/– Chemo
planned for possible surg. Arm 2/CRT won. –– EORTC 22931, Bernier: operable stage III/IV
3 yr OS 23% vs 37% vs 27%, compliance H&N SCC of OC, OP, larynx, hypopharynx
164 D. M. Trifiletti et al.
(ECE, +margin, PNI, LVSI, level IV/V nodes): 73% induction; 78% CRT)). More adverse
post-op 66/33 +/− concurrent cisplatin events.
(100 mg/m2 Q3wks): CRT won: 5-yr DFS –– Madrid, Hitt 2014: Induction then concurrent
36→47%, OS 40→53%, LRC 69→82% (DM RT vs concurrent CRT. Induction w TPF or
unchanged ~25%). ↑toxicity (21→41%). PF. Chemo-RT was cis 100 mg/m2 q3w. No
–– RTOG 9501, Cooper: operable H&N (≥2 LN, benefit in OS of induction over concurrent.
ECE, +margin): 66/33 +/− concurrent cispla-
tin (100 mg/m2 Q3wks). CRT won: 2-yr DFS Sim/Planning
43→54%, LRC 72→82%, trend for OS. ↑tox- –– Dental eval, PEG?, aquaplast, supine, 35 fx
icity. On unplanned subset analysis, R+ and/or IMRT
ECE had benefit in LRC (33% vs 21%) and –– Bilateral neck unless T1–2N0 tonsil with
DFS (12% vs 18%). Lack of OS benefits likely <1 cm BOT of soft palate invasion (O’Sullivan
bc of high OPX proportion, in comparison to 2001: 3.5% contralateral neck failure, all N+)
EORTC.
–– Meta-analysis of 95–01 and 22,931 (Bernier, Recurrence
2005): CRT improved OS, DFS and LRC for • All pts should be seen at 6 week post-tx clini-
ECE or + margins. Trend for stage III/IV, PNI, cal assessment, and if there is primary pro-
LVSI, low neck nodes gression, then consider surgery. PET is
preferred to post-tx neck dissection. Post-
Induction Chemo chemo-RT PET-CT should be at 3 months. If
–– TAX 323: unresectable H&N SCC: TPF vs PF negative, 0% recurrence. Ninety percent of
induction →RT alone. TPF increased MS equivocal PET CTs will become negative.
16→19 months. • Recurrent p16 positive has MST of 2.6 years
–– TAX 324, Lorch 2011: See LX section. vs. PST negative of 0.8 years MST. Surgery is
Posner: unresectable H&N SCC: taxotere + better than reRT.
cisplatin +5-FU (TPF) vs PF induction →CRT
with carboplatin. Induction CRT won. NCCN
Three-yr OS 48→62%. Five-year OS 52% vs Early Stage
42% MST 71 m vs 35 m. PFS 38 m vs 13 m. T1N0(</= 2 cm)/ T2N0 (>2 cm, </= 4 cm) /
Twenty-five percent of patients never made it Or N1
to RT (progressed, died, withdrew). Concluded Surgery or RT: results are equivalent.
that taxotere needed for induction.
–– DeCIDE trial, Chicago, Cohen, 2014: locally 1. Surgery+/-PORT: Tonsillectomy/hemiglos-
advanced H&N SCC: (1) CRT with docetaxel, sectomy w/elective neck dissection or WLE of
5FU, hydroxyurea, BID fx vs (2) TPF induc- soft palate
tion then same CRT. CRT was 1.5 Gy BID, 2. RT alone:
74–75 Gy to GTV, 54 Gy to high risk, and • High-risk CTV:66–70Gy/30–35fx (boost
39 Gy to low risk. More adverse events w primary to 66 Gy for T1, 70Gy for T2).
induction. No difference in DFS, RFS, and Includes tumor +1 cm margin (min 5 mm)
OS. Underpowered. Fifty-eight percent of and involved LNs.
deaths were from HNSCC; remainder due to • Intermediate-risk CTV(optional):
PNA, PE, drug toxicity, and other cancers. 63Gy/35fx rest of level with involved
–– PARADIGM trial, Harvard (Haddad, 2013): LN/1st echelon LNs
2004–2008. N = 145. Unresectable or unlikely • Low-risk CTV:56 Gy/35fx to elective neck
to be cured surgically. III/IV, N2–3. TPF (levels II-IV, ipsi RP).
induction + CRT with docetaxel or carboplatin • LAN: 50Gy/25 fx; boost 10Gy/5fx if LN+
vs CRT with cisplatin: no difference (3-yr OS hemineck.
Head and Neck Cancers 165
• HPV positive: most commonly use tongue, and if vallecula involved include
50 Gy/25 fractions in 5 weeks, QD; suprahyoid epiglottic larynx (CTV P); as well
64 Gy/40 fractions. 4 w, BID is the second as involved neck levels plus one level above
most common. and below (CTV N).
Well-lateralized tonsil/soft palate: treat • For tonsil:
unilaterally, for N0-1. All N2b + should get High-risk subclinical disease includes CTV70
bilateral neck RT. plus adjacent buccal mucosa, palate, and BOT,
3. Altered fract ≥T2N0 (BID, concomitant as well as first echelon nodes
boost, DAHANCA (Easiest: 2Gy/fx; 6fx/wk, More advanced tonsil include mandibular
add extra fraction on Fridays starting week 2.) bone, ipsi pterygoid muscle, parapharyngeal
**Can boost w 20–30 Gy boost via Ir-192 implant space, and adjacent NPX
• Low-risk CTV (i.e., CTV3):56 Gy/35fx to
Tumor Nodal coverage Primary
elective neck (levels II-V, RP, retrostyloid
coverage space)
Tonsil N-hemineck: II–IV* Ipsilateral soft • LAN: 50Gy/25 fx; boost 10Gy/5fx if LN+
N+ hemineck: RS, RP, palate, BOT,
IB-V, SCV GTS hemineck
BOT N-hemineck: II–IV* GTS, vallecula, • Post-LN-positive hemineck should treat
N+ hemineck: RS, RP, pre-epiglottic
II–IV and RP LNs; and additionally, treat
IB-V, SCV space
Soft palate N-hemineck: RP, II–IV Entire soft level V
N+ hemineck: RS, RP, palate, adj HPV-: lower threshold for neck dissec-
IB-V, SCV NPX, sup tonsil
tion after chemo-RT
2. Altered frac RT (RTOG 9003) if pt. refuses
*Coverage of RP nodes on “involved” node- chemo
negative hemineck is recommended 3
. Surgery (total glossectomy) + post-op RT or
CMT for T4/+margin/ECE
Advanced Stage
Chemoradiation is preferred. Post-op RT Indicated for
Neck
1. Definitive Chemoradiation: 70Gy + concur- • Multiple positive nodes
rent cisplatin (100 mg/m2, q3 weeks or 40 mg/ • >3 cm
m2 weekly); carbo/taxol if poor kidneys; • Extracapsular spread of nodal disease
cetuximab • Close or positive margins
• High-risk CTV (i.e., CTV1): 70Gy/35fx • Prior biopsy/violation of neck/trach prior to
(include gross dz +1–2 cm margin) laryngectomy
• Intermediate-risk CTV (i.e., CTV2;
optional): 63Gy/35fx rest of level with Primary tumor
involved LN/1st echelon LNs • T4
• For BOT: • Extensive subglottic extension
High-risk subclinical disease includes • + Margin/close margins (<5 mm)
CTV70 + 5–10 mm expansion, plus entire • Perineural/vascular invasion
BOT, vallecula, generous portions of oral • High-grade histology
• Concerned surgeon
166 D. M. Trifiletti et al.
p16-Neck staging
cN1 Single ipsi node ≤3 cm, no ECE pN1 Single ipsi node ≤3 cm, no ECE
cN2 pN2
cN2a Single ipsi, 3–6 cm, no ECE pN2a Single ipsi, 3–6 cm, no ECE; or <3 cm with ECE
cN2b Multiple ipsi, 3–6 cm, no ECE pN2b Multiple ipsi, 3–6 cm, no ECE
cN2c Bilateral or contralat, 3–6 cm, no ECE pN2c Bilateral or contralat, 3–6 cm, no ECE
cN3 pN3
cN3a >6 cm, no ECE pN3a >6 cm, no ECE
cN3b Any clinically overt ECE pN3b Single >3 cm with ECE; or multi nodes with any ECE
• Level II = OPX; for Level 2B close to parotid –– HPV: can be detected in FNA specimen.
consider skin met to parotid or salivary gland Should check in all. If positive, tonsillectomy
primary (usually b/l) recommended. TORS results in
• Level V or RP = HPX or NPX 63–100% detection of primary.
• Level IV = often outside H&N. The lung and –– Thyroglobulin, calcitonin, PAX8, or TTF
esophagus (left SCV is Virchow’s node so for anaplastic/undifferentiated tumors
look below diaphragm!)
• Level VI = thyroid Retrospective Reports
–– McQuone 1998: improved diagnostic yield
Work-up with tonsillectomy over biopsy.
–– FNA of LN is preferred. If non-dx, then repeat –– UF 2001: LC 78%, OS 47%.
with US. –– Baker 2005: larynx-sparing RT is just as effec-
–– Open biopsy contraindicated because of: tive, less toxic.
–– Potential tumor spill and disruption of fas- –– Loyola 1997: unilateral neck RT led to 44%
cial planes (which naturally prevent tumor contralateral neck failure and 44% primary
spread) emergence rate.
–– Limitation of future treatments –– Soushtari 2011 (UVA): all IMRT. Five-yr OS
–– Scar formation and increased morbidity 71%, DFS 85%. All nodal failures had bulky
(biopsy wound must be excised or covered disease.
in higher dose volume)
–– Necessitation of second operation if path Treatment Paradigm
staging pursued –– Natural history: Failure usually in ipsi neck >
–– “Neck violation”: incisional/excisional distal. Among surgically treated patients,
biopsy of node mucosal progression is 18% in old series,
–– CT +/− MRI. likely <10% now.
–– PET-CT: detects tumors in 25% of patients. –– Typically, multimodal.
Can show DMs, non-HN primary. FPR: 20%; –– Usually neck dissection + aRT
thus, confirmatory bx necessary. PET scan –– ECEmic in p16+ patients: unknown if aCRT is
PPV 90%, NPV 75%. beneficial. Currently, any ECE is indication
–– Fiberoptic endoscopy. for aCRT.
–– EUA (DL and direct bx): not necessary if pri-
mary found otherwise. For level 2/3 LNs, RT Planning
biopsy of NPX, b/l BOT, b/l pyriform –– Timing of RT does not appear to influence
sinuses, SGL recommended. Tonsil cancers in DFS.
crypts may not be seen. B/l tonsillectomy rec- –– Volumes:
ommended if tissue present. If HPX ca sus- –– Contra neck and mucosal subsites (b/l ton-
pected, then biopsy the pyriform sinus. sil, BOT) controversial, sometimes
–– Pathology: excluded in p16+/EBV- with N1 disease.
• SCC or undifferentiated ➔ HPV (p16 –– NPX is typically included; consider exclud-
staining) and EBV testing ing in non-Asian with p16+/EBV- LNs. NPX
• Adenocarcinoma ➔ TG and calcitonin contour: base of skull to soft palate, posterior
staining. If TG and calcitonin negative (and choana and posterior pharyngeal wall, and
level I-III) likely parotid origin laterally cover fossa of Rosenmuller.
–– Triple endoscopy if levels IV–V. –– LN volumes: bilat RP nodes, bilat IB-IV,
–– EBV: rarely elevated in North American ipsi V. Neck RT will include the RS space
patients w SCCUP. Can check EBV in FNA and RP LNs (level VII) with IMRT. Thus,
specimens. Recommended to direct bx for NPX will receive considerable dose even if
patients from endemic area. not in CTV.
168 D. M. Trifiletti et al.
Neutrons RT Doses
–– RTOG-MRC trial: 32 inoperable salivary –– Post-op:
gland: neutron vs photon/electron: closed –– 60–63 Gy @ 1.8 Gy–2Gy, 66Gy for posi-
early, neutrons won: 10 yr LRC 17→56%, OS tive margin
unchanged (15→25%) –– When to treat BOS: cPNI, rPNI, named
–– 19.2nGy to GTV (1.2nGy x4/wk), 13.2nGy to nerve involved, “extensive” PNI; adenoid
PTV2 cystic
–– PNI considerations:
NCCN –– Parotid:
–– Surgery is the mainstay of treatment. LND –– VII: track to styloid foramen; if VII grossly
indications (supraomohyoid, I-III): HG involved, extend to include facial canal
(except ACC), cN+,4+ cm. through petrous bone
–– Parotidectomy subtypes: –– Submandibular:
• Superficial = superficial –– V3 (lingual): track nerve to foramen ovale,
• Total = superficial + deep, spares VII include Meckel’s cave
• Radical = includes VII and skin/fascia –– XII: track nerve to hypoglossal canal
• Indications for post-op RT: –– VII: track to styloid foramen
1. high grade
2. PNI or LVI Definitive for Unresectable/nonsurgical
3. LN + or ECE 70Gy/35fx for gross dz
4. Recurrent
5. Adenoid cystic histology Treat elective neck to 50–54 Gy in 1.8-2Gy/fx
–– Indications for post-op chemo-RT: Consider neutrons
1. close (<2 mm)/positive margins
2. ECE
–– Indications to treat LNs (N0: IB-III; N+: IB,
RSS/II-V)
1. HG (except ACC)
2. pN+
3. Recurrent
172 D. M. Trifiletti et al.
N3 IVB cN2a Single ipsi, 3–6 cm, no ECE pN2a Single ipsi, 3–6 cm, no ECE; or <3 cm
with ECE
M1 IVC cN2b Multiple ipsi, 3–6 cm, no ECE pN2b Multiple ipsi, 3–6 cm, no ECE
cN2c Bilateral or contralat, 3–6 cm pN2c Bilateral or contralat, 3–6 cm, no ECE
, no ECE6cm, no ECE
cN3 pN3
Risk Factors cN3a >6 cm, no ECE pN3a >6 cm, no ECE
–– Smoking, EtOH, GERD, asbestos, paint cN3b Any clinically overt ECE pN3b Single >3 cm with ECE; or multi
nodes with any ECE
fumes, Ni, soot, tar, vitamin deficiencies (Fe,
B12, C), betel nuts
Head and Neck Cancers 173
–– Larynx innervation: all vagus (X), via recur- –– RTOG 9003, Fu, 2009: compared
rent laryngeal nerve and the superior laryngeal • CFRT (70/35over 7 weeks) vs.
nerve • Hyperfrac RT (81.6/68over 7 weeks, i.e.,
–– Note: thyroid cartilage invasion and extrala- 1.2 Gy BID) vs.
ryngeal spread difficult to determine by CT • Accelerated frac w split course (67.2/42
(PPV, 81%; NPV, 71%; sens, 49%; spec, 92%) fractions over 6 weeks, i.e., 1.6 Gy BID;
Ear pain? with a split course where there was a break
–– Oral tongue: auriculotemporal nerve (CN V) in the middle of treatment) vs.
–– BOT: Jacobson’s nerve (CN IX) • Accelerated frac with delayed concomitant
–– Larynx/HPX: Arnold’s nerve (CN X) boost (1.8 Gy/fx 5d/w w 1.5 Gy/fx as boost
–– Anatomic landmarks: field as second daily tx over the last 12d;
–– Hyoid = C3 total, 72/gy42 fractions). All tumors from
–– Superior border of thyroid = C4 lip to esophageal verge eligible, except
–– Cricoid cartilage = C6 glottic larynx. Parallel opposed field.
• LRF: 29% vs 51% vs 58% vs 52%.
Work-up • OS 30% vs 37% vs 31% vs 34%. Both
–– FNL, biopsy hyperfrac and DCB had improvement in
–– History: SGL = dysphagia/odynophagia, and LRF.
otalgia (Arnold) • OS and LRF improved in the hyperfrac
–– Voice changes (hoarseness), swallowing, aspi- arm. 1% LRC improvement for each day tx
ration, and pulmonary status (can the patient shortened. CFRT had lowest acute tox vs
climb a set of stairs) all others. Worse outcomes in on-tx smok-
–– Physical: palpate larynx (mass at thyroid ers, more so than previous heavy smokers.
notch = preepiglottis space/PES invasion),
gently move (loss of laryngeal click = postcri- LX Cancer – Larynx Preservation
coid area/PCA invasion), palpate tongue and –– VA Larynx Trial (Wolf, 1991): III/IV larynx:
BOT, LN exam (1) surgery+PORT vs (2) Chemoselection:
–– CT +/− MRI, CT chest, +/− PET, dentistry induction cis/5FU x 2c, then re-eval, then 1c
–– PFTs (total 3c), then RT if PR+ or surg/PORT if
<PR. At 4y, LX preservation was 12% vs 64%.
OS ~50% (NSS). TL 88% for surg vs 38%
Studies CRT. Pts alive w larynx 5% vs 31%, favoring
CRT. LC lower for CRT (98→88%). Salvage
RT Alone Fractionation laryngectomy rate 56% in T4, 29% in < T4
–– Yamazaki 2006: phase III, T1 glottis: 2Gy/fx tumors. Note, most T4 is now T3 (bc any thyr
to 60–66 Gy vs 2.25 Gy/fx to 56–63 Gy. ctx invasion was T4, now minor invasion T3)
Hypofrac won. Five-yr LC 77→92%, CSS, –– RTOG 91–11 (Forastiere, 2003; 2013): III/
OS, toxicity unchanged (Bledsoe 2017 showed IV larynx, T2/T3/ “early T4” (now T3) of
possible OS advantage to hypofrac in T1-T2 glottic/supraglottic. Technically some resect-
from NCDB) able. Randomize: (1) RT alone vs (2) induc-
–– RTOG 9512: T2 glottis. N = 250. 70/35 vs tion chemo (cis 100 mg/m2 on d1 and 5-FU
79.2 BID (1.2 Gy). No change, trend for ↑LC 1000 mg/m2 q24h CI, qw, x2c), followed by
w hyperfrac (70→79%, p > 0.11), LRC 67% RT if PR+ or surg/PORT if <PR vs (3) concom
vs 73%, DFS 50% vs 49%, OS 63% vs 72%. CRT (cis 100 mg/m3 q3w × 3c). RT 70/2 and
Hyperfrac better numerically, but not SS, 50/2 to elective neck and sclav. All cN2
likely bc underpowered. Similar late G3+ tox, patients got planned neck dissection after
8%. For cost and pt convenience, consider RT. Endpoint: LX preservation. Concurrent
other fractionation regimens, e.g., 2.25 Gy/d CRT won for LC and LRC (69→55→51%),
174 D. M. Trifiletti et al.
established it as SOC. Concurrent CRT was NR get surg, then PORT. No OS difference.
better for LX preservation vs RT alone, but However, CT + RT +/− surgery had 17% LX
there was no difference bw two chemo groups. preservation rate at 5 years (EORTC 24891
Five-yr larynx preservation 84→71→66%. Lefebvre, JNCI, 1996) and 8.7% at 10 years.
Chemo reduced DMs (13% vs 22%). OS Ten-year DM 36% with surgery and RT, 25%
unchanged. Some argue induction arm is the with chemo, RT, +/− surgery, low OS in both
best (esp if w TPF and not PF). Larynx preser- arms likely due to poor salvage success
vation acceptable for T3
Induction Sim/Planning
–– TAX 324 (Posner, Lorch 2011): unresectable
head and neck (33% larynx/hypopharynx): –– Mask, shoulders down
induction taxotere + cisplatin +5-FU (TPF) vs
induction PF x3 cycles, then 70 Gy CRT. CRT
was 70–74 Gy w concurrent carbo AUC 1.5. General Treatment Recommendation
Induction TPF won. Three-yr OS 48 vs 62%,
5-yr OS 52% vs 42%. MST 71 m vs 35 m. –– Stages I–II: unimodal therapy
LRC 62→70%, DM unchanged. TPF was –– Stages III–IV: multimodal therapy (e.g.,
toxic. chemo-RT)
–– EORTC 24891: same as VA trial but with pyri-
form sinus and required CR. Same OS (~40%).
5-yr functional larynx 35%. Surgery Types
–– GORTEC 2000–01, Janoray, 2015: III-IV LX,
HPX SCC who required TL randomized to –– SGL
TPF vs PF induction. Responders get • Supraglottic laryngectomy (T1, T2, T3 by
RT. Primary endpoint was LX preservation. PES only)
TPF increased LX preservation and larynx • Removes: upper 1/2 thyroid cartilage, preepi-
function. No change in OS, DFS, LRC. TPF glottic space, epiglottis, AE folds, false cords,
also w less G3–4 tox, 17% vs 9%. up to one arytenoid (need to preserve one)
–– Contraindications:
Chemo-RT –– B/L arytenoids
–– See oropharynx (RTOG 9501, EORTC –– Fixed cord/any true cord
22931). –– Inadequate pulmonary reserve/ “cannot
–– For larynx and hypopharynx, the rate of long- walk up two flights” (unable to tolerate
term G-tube placement after tx is 50% (vs. aspiration)
40% for oral cavity and OPX). –– Thyroid/cricoid cartilage
–– For just hypopharynx, the rate is 25%.
–– In the MACH-NC meta-analysis, the benefit GL
of chemo-RT is not higher than that for other –– Mucosal stripping (CIS)
dz sites. –– Transoral laser excision/cordectomy (T1a).
–– If no pCR, the median DFS is 12 months. Contraindicated for AC involvement
–– Vertical partial laryngectomy (T1a, T1b, select
HPX Trials T2)Removes: 1/2 thyroid cartilage + ≤ 1 and
–– Treatment paradigm similar to LX cancer 1/3 TVC
–– EORTC 24891 Lefebvre, 1996, 2012. Rando SGL/GL
(1) surgery, PORT, or (2) induction chemo –– Total laryngectomy (T3, T4a)Removes: hyoid,
(i.e., with cisplatin and 5-FU x3), then (2a) thyroid, cricoid w/ entire larynx +strap
patients with CR get RT, (2b) patients w PR or muscles
Head and Neck Cancers 175
• Patient left with permanent tracheostomy and –– After CO2 laser, do not need adjuvant RT if no
pharynx reconstruction (by suturing to BOT) tumor on ink.
• Most common failure sites = BOT and stoma –– Stripping uses H2O jet to lift the tumor off; it
is not an oncologic procedure and needs adju-
Speech After Laryngectomy vant RT.
–– Failure rate is local only (not LNs), so need
–– Esophageal speech very close follow-up, q 3 m.
–– Tracheoesophageal puncture –– Tis: cord stripping (laser/CO2) or RT.
–– Electrolarynx –– RT:
–– T1: 63 Gy/28 fractions at 2.25 Gy
–– T2: 65.25 Gy/ 29 fractions at 2.25 Gy
Treatment for Supraglottic Larynx –– Historically 5 × 5 field for T1 or 6x6 field for
T2, now more based on anatomic borders.
–– T1N0 Treat in <43 days
–– Supraglottic laryngectomy w/bilateral –– Iso @TVC
MRLND –– 6MV photons, wedges w heels anterior
–– RT alone 66 Gy to primary, 50 Gy to bilat –– Bolus if AC involved
neck –– Hotspot <110%
–– T2N0 –– 3D: CTV covers entire larynx, including glot-
–– (1) Supraglottic laryngectomy with bilat- tis (TVCs, anterior and posterior commissures),
eral modified radical LND part of supraglottis (FVCs, arytenoids) to top of
–– (2) RT alone 70 Gy to primary, 50 Gy to thyroid notch and subglottis inferiorly to cri-
bilat neck (for bulky T2, could consider coid cartilage (can extend to first tracheal ring
altered fractionation schedule but not for T2); paraglottic space and thyroid cartilage
standard) can be included if deemed at risk. CTV should
–– T3-4N+ extend ≥2 cm craniocaudally.
–– CTV70. avoid SIB, esp >2Gy per fraction, –– 2D: superior border is the top of the thyroid
due to risk of late toxicity. cartilage; inferior border is the bottom of the
–– High-risk CTV 63Gy/35fx. Includes cricoid (at C6) or one tracheal ring below cri-
CTV70 + entire larynx (top thyroid notch coid if T2 for subglottic involvement; ante-
to bottom cricoid), paraglottic and preepi- rior border has a 1 cm flash; and posterior
glottic space, pyriform sinuses, vallecula, border is the anterior edge of the vertebral
and up to 1 cm BOT, as well as high-risk body.
first echelon nodes –– Alternative for favorable T2a is 79.2 Gy @ 1.2
–– Elective LN: 56Gy/35fx., e.g., upper level BID, per RTOG 95–12.
II, RPLNs. LAN: 50Gy/25 fx; boost –– Unfavorable T2: “old T2b,” if there is more
10Gy/5fx if LN+ hemineck extensive disease (e.g., cord hypomobility),
then consider chemo-RT and 70 Gy w RNI w
concurrent cis. Argument for this is compar-
Treatment for Glottic Larynx ing CRT arm of 91–11 vs. HFX arm of 9512.
–– T3 or N+: CRT (surgery salvage) or induction
–– Early larynx options: stripping, laser, RT, chemo or laryngectomy.
cordectomy, vertical hemilaryngectomy –– Primary and involved LN: 70Gy in 35 fx.
–– Contraindications to vertical hemilaryn- –– High risk CTV 63Gy/35fx. CTV70 + entire
gectomy: Involves more than one true larynx (top thyroid notch to bottom cricoid),
cord, epiglottic invasion, fixed cord, paraglottic and preepiglottic space, pyriform
bilateral arytenoids involvement, 5 mm
sinuses, vallecula, and up to 1 cm BOT, as
subglottic extension. well as high risk first echelon nodes.
176 D. M. Trifiletti et al.
• If level II/III involvement, do not need to levels III-IV bilaterally, and add level II for
cover RP, level IB, level V. advanced T stage or N+
• If dz. breaks through cartilage, cover level VI. –– CTV50–56* = low-risk elective, including
–– Primary + b/l neck: 70/63/54 in 35 fx. entire larynx (top thyroid notch to bottom cri-
–– cN0: II-IV. coid), paraglottic and preepiglottic space, pyri-
• [+ VI for subglottic extension, HPX form sinuses, vallecula, as well as first echelon
involvement, emergent trach, soft tissue nodes (see above)
extension of primary to neck
• + VII involvement for subglottic extension, Covering level VI indications: emergent
HPX involvement, SCV node] trach, subglottic extension, apex or pyriform
• cN+: IB + RSS/II–V. sinus involvement
–– [+ VI for subglottic extension, HPX involve-
ment, emergent trach, soft tissue extension of
primary to neck
Treatment for Hypopharynx
–– + VII involvement for subglottic extension,
–– T1N0
HPX involvement, SCV node]
–– (1) Partial laryngopharyngectomy w/ bilat-
–– *with any HPX involvement add b/l RP.
eral MRLND
–– CTV70 = primary, gross LN.
–– (2) RT alone 70 Gy to primary, 50 Gy to
–– CTV63 = [CTV_70 + 5 mm], remaining lar-
bilat neck
ynx, piriform sinuses, paraglottic space, PES,
T2N0
vallecula, cN+ neck.
–– (1) Partial or total laryngopharyngectomy
–– CTV56 = cN0 neck.
with bilateral modified radical LND
–– Cis (100 mg/m2) days 1, 22, 43.
–– (2) RT alone: altered fractionation
–– T4: laryngectomy preferred over chemo-RT
T3N+: Chemo-RT if functional larynx and not
because of failure rates on VA larynx.
aspirating
–– PO(C)RT.
–– Primary + b/l neck: 70/60/54 in 35 fx
• Post-op bed + b/l neck: 66/60/54 in 30 fx
–– cN0: RP (stop at C1), II-IV [+ VI for exten-
• Cis (100 mg/m2) days 1, 22, 43.
sion below cricoid or into PS apex]
–– CTV66 = post-op bed if + margin or ECE.
–– cN+: RP (stop at BOS), IB + RSS/II-V [+
–– CTV60 = post-op bed, cN+ neck.
VI for extension below cricoid or into PS
–– CTV54 = stoma, cN0 neck.
apex]
–– CTV70 = primary, gross LN
–– CTV63 = CTV_70 + Entire LX/HPX (hyoid
Treatment for Subglottic Larynx
to cricoid), 3 cm sup/inf if pharyngeal wall
involvement, cN+ neck
T1-2N0 RT alone
–– CTV_56 = cN0 neck
–– RT alone 70 Gy to primary + b/l neck (II-
–– T4a
IV + VI): 70/60/54 in 35 Fx, BID Friday
–– Total laryngectomy +/− post-op RT or CRT
–– For bulky T2, could consider altered fraction-
ation schedule but not standard
I ndications for Treating Stoma
T3-4N+ (“No ESCAPE”)
–– Cisplatin 100 mg/m2 on day 1, 22, 43 w/con- –– Nodes: multiple LNs or positive LN close to
current CTV70 = GTV + 5–10 mm, carving stoma
out bone/air –– Emergency tracheostomy
–– CTV60–63 = high-risk elective, consider N+ –– Subglottic extension >5 mm
neck plus one level above/below. For subglot- –– Surgical scar crosses stoma
tic involvement, always treat level VI and con- –– Anterior soft tissue/skin involvement
sider superior mediastinum. Always cover LN –– Positive/close margins
Head and Neck Cancers 177
PC
––ECE
178 D. M. Trifiletti et al.
Nasopharynx Mandibular
condyle
RP
Tonsil node C1 Spinal canal
C2 Spinous
JD process
node
II
Vallecula C3
Pyriform sinus
C4
Hyoid
Thyroid carlage
C5 III
Arytenoids
True vocal cords
C6
Cricoid carlage
Subglos
Head and Neck Cancers 179
Clinical staging
T1a T1b/T2a T2b/T3a T3b/T4a T4b
N0 IA IB IIA IIB IIC
cN+ III
M1 IV
Pathologic staging
T1a/T1b T2a T2b/T3a T3b/T4a T4b
N0 IA IB IIA IIB IIC
N+
pIIIA T1-T2a and N1a/N2a
pIIIB T1-T2a N1b/c r N2b or T2b-T3a NN1a-N2b
pIIIC T1-T3a N2c/N3 or T3b-4aN+ or T4b N1-
2c
pIIID T4bN3
M1 IV
General Workup
–– Subtypes: superficial spreading (65%), nodu- –– WLE with SLN
lar (25%) lentigo maligna, and acral –– <1 mm: nothing special
lentiginous. –– >1 mm, labs, CXR, consider CT for nodes
–– Wu, 2016: Sunburns are associated w –– No shave biopsy unless index of suspicion low
increased risk of melanoma (vs any other type –– LN+: PET-CT, MRI
of cancer): RR 2.41 in melanoma, 1.48 for –– SLNB if 1+ mm thickness. If pos, then com-
SCC, 1.18 for BCC. plete dissection of all involved LN basins.
–– ABCDE (for melanoma): asymmetry, borders, Twenty percent chance more LNs. Must do
color, diameter >5 mm, enlargement. dissection if cLN+.
182 D. M. Trifiletti et al.
Radiation Therapy
Mucosal Melanoma
Primary site Nodes
Indications (almost never) ECE –– T3 and T4 only
for RT R+ 4+ LNs in –– Sinus/nasal cavity: surgery with ND if N1 or
extensive neurotropism groin
4+ LN in
def RT if T4b
axilla –– PORT to primary site and include neck if N1
2+ cervical –– OC/OPX, LX, HPX: surgery, ND, PORT. If
3+ cm LN T4b, then def RT
Doses 70/35 60/30
50/20 (2.5 Gy) 48/20
35/5 (7 Gy) (2.4 Gy)
30/5
(6 Gy)
184 D. M. Trifiletti et al.
High-Risk Factors for Local Recurrence • Range (<5% dose) is E/2 (cm)
–– Trunk and extremities >2 cm; check/ • Required thickness of lead shielding
forehead/scalp/neck >1 cm; mask areas of the (mm) = E/2
face, genitalia, hands, feet >6 mm
–– Poorly circumscribed
–– Recurrent Standard Skin SCC, NCCN
–– Immune suppression
–– Prior RT or chronic inflammation –– Prefer WLE or Mohs
–– PNI –– PORT for high-risk features
–– Rapid growth –– T3/4
–– G3-4 –– parotid gland
–– adenoid/adenosquamous/desmoplastic –– R+
–– Clark level IV/V or >2–5 mm thick. –– PNI: should be of a named nerve bc nerves
–– For SCC: acantholytic, adenosquamous, des- <0.1 mm tend to not have higher LRs vs no
moplastic, or metaplastic PNI.
–– For BCC: morpheaform, basosquamous, scle- –– Multi high-risk features (>2 mm thick,
rosing, mixed infiltrative, or micronodular clarke ≥IV (invades reticular dermis), pni,
poorly/undiff, ear or nonhair bearing lip)
Orthovoltage Background –– Indication for RT to regional LNs: N2+,
–– Has max dose at surface, less beam constric- ECE, T3/4, PNI
tion at surface and depth, can use smaller –– Topical 5FU/imiquimod for low-risk superfi-
fields, has less penetration through eye shields cial BCC
–– However, has higher exit dose than electrons, –– Definitive RT for
higher absorption in bone/cartilage, not avail- –– high risk or nonsurgical candidates, e.g.,
able in most depths large lesion of scalp, ears, forehead
(>2 cm); central face including >0.5 cm
Electron Background involving the eyelids, ala, nose, lips
–– Similar outcomes to photons –– cartilage, bone, muscle involvement
–– Have lower RBE than orthovoltage X-rays by –– recurrent
15–20% and require commensurate increase –– Neck treatment same as other H&N cancer. If
in dose. Thus, most authors of NCCN will near parotid, treat parotid and levels II–IV on
equate electrons dosed to 90% IDL vs ortho- hemineck
voltage dose to dmax
–– Require bolus, at least 0.5–1 cm RT Contraindications
–– Confirm w OSLDs and phantom –– Poor blood supply (shin, back of hand)
–– Require larger field size to allow sufficient –– Gorlin’s syndrome
buildup and account for lateral constriction, –– ATM
usually 1.0–1.5 cm margin. Wider margins –– Xeroderma pigmentosum
necessary than w orthovoltage X-rays bc of –– Scleroderma
constriction of higher IDLs at depth
–– “Tertiary collimation”: using lead on the skin Sim
to reduce beam penumbra and make tighter –– Based on location. Immobilize and use bolus
margins for OARs. Does not work for superfi- 5 mm
cial X-rays bc of backscatter
–– 4-3-2-2 rule.
–– For an electron beam with energy, E
• Depth to 90% dose (cm) at E/4 cm
• Depth to 80% dose (cm) at E/3 cm
186 D. M. Trifiletti et al.
Contouring/Planning Chemo
–– Electrons –– BCC: only if metastatic (hedgehog inhibitor)
–– Primary/post-op area plus 1.5 cm margin –– SCC: may consider cisplatin for locally
–– 6–12 MeV advanced cases
–– Prescribe to 90% IDL
–– Lead eye shield: use if treating medial can-
thus/eyelid tumors
–– Try to block out lacrimal gland
–– Photons
–– Use IMRT when covering regional LNs
Tumor RT
diam margins Dose
<2 cm 1–1.5 cm 60–64 Gy/30–32 fx @ 2 Gy in
6–6.4 w
55 Gy/20 fx @ 2.75 Gy in 4 w
50 Gy/15 fx in 3 w
30 Gy/5 fx in 2–3 w
12–20 Gy/1 fraction for
symptomatic management of
large bleeding tumors of very ill/
nursing home type patient
>2 cm 1.5–2 cm 66–70 Gy/33–35 fx @ 2 Gy in
6.6–7 w
55 Gy/20 fx @ 2.75 Gy in 4w
Post-op 50 Gy/20 fx @ 2.5 Gy
60 Gy/30 fx
Head and Neck Cancers 187
29. Straatsma BR, Diener-West M, Caldwell R, et al. a randomized controlled clinical trial. Int J Radiat
Mortality after deferral of treatment or no treat- Oncol Biol Phys. 2006;66:981–91.
ment for choroidal melanoma. Am J Ophthalmol. 42. Bristol IJ, Ahamad A, Garden AS, et al. Postoperative
2003;136:47–54. radiotherapy for maxillary sinus cancer: long-term
30. Al-Sarraf M, LeBlanc M, Giri PG, et al. outcomes and toxicities of treatment. Int J Radiat
Chemoradiotherapy versus radiotherapy in patients Oncol Biol Phys. 2007;68:719–30.
with advanced nasopharyngeal cancer: phase III 43. Dulguerov P, Jacobsen MS, Allal AS, et al. Nasal
randomized Intergroup study 0099. J Clin Oncol. and paranasal sinus carcinoma: are we making
1998;16:1310–7. progress? A series of 220 patients and a systematic
31. Baujat B, Audry H, Bourhis J, et al. Chemotherapy review. Cancer. 2001;92:3012–29.
in locally advanced nasopharyngeal carcinoma: an 44. Le QT, Fu KK, Kaplan MJ, et al. Lymph node
individual patient data meta-analysis of eight ran- metastasis in maxillary sinus carcinoma. Int J Radiat
domized trials and 1753 patients. Int J Radiat Oncol Oncol Biol Phys. 2000;46:541–9.
Biol Phys. 2006;64:47–56. 45. Allen MW, Schwartz DL, Rana V, et al. Long-term
32. Chen QY, Wen YF, Guo L, et al. Concurrent chemo- radiotherapy outcomes for nasal cavity and septal can-
radiotherapy vs radiotherapy alone in stage II naso- cers. Int J Radiat Oncol Biol Phys. 2008;71:401–6.
pharyngeal carcinoma: phase III randomized trial. J 46. Cantu G, Bimbi G, Miceli R, et al. Lymph node
Natl Cancer Inst. 2011;103:1761–70. metastases in malignant tumors of the parana-
33. Lee AW, Lau WH, Tung SY, et al. Preliminary sal sinuses: prognostic value and treatment. Arch
results of a randomized study on therapeutic gain by Otolaryngol Head Neck Surg. 2008;134:170–7.
concurrent chemotherapy for regionally-advanced 47. Kim SJ, Kim K, Kim BS, et al. Phase II trial of con-
nasopharyngeal carcinoma: NPC-9901 Trial by the current radiation and weekly cisplatin followed by
Hong Kong Nasopharyngeal Cancer Study Group. J VIPD chemotherapy in newly diagnosed, stage IE to
Clin Oncol. 2005;23:6966–75. IIE, nasal, extranodal NK/T-Cell Lymphoma: con-
34. Lee AW, Tung SY, Chan AT, et al. Preliminary sortium for improving survival of lymphoma study. J
results of a randomized study (NPC-9902 Trial) on Clin Oncol. 2009;27:6027–32.
therapeutic gain by concurrent chemotherapy and/or 48. Isobe K, Uno T, Tamaru J, et al. Extranodal natu-
accelerated fractionation for locally advanced naso- ral killer/T-cell lymphoma, nasal type: the sig-
pharyngeal carcinoma. Int J Radiat Oncol Biol Phys. nificance of radiotherapeutic parameters. Cancer.
2006;66:142–51. 2006;106:609–15.
35. Lee N, Harris J, Garden AS, et al. Intensity- 49. Ang KK, Harris J, Wheeler R, et al. Human papillo-
modulated radiation therapy with or without che- mavirus and survival of patients with oropharyngeal
motherapy for nasopharyngeal carcinoma: radiation cancer. N Engl J Med. 2010;363:24–35.
therapy oncology group phase II trial 0225. J Clin 50. Ang KK, Trotti A, Brown BW, et al. Randomized
Oncol. 2009;27:3684–90. trial addressing risk features and time factors
36. Lee N, Xia P, Quivey JM, et al. Intensity-modulated of surgery plus radiotherapy in advanced head-
radiotherapy in the treatment of nasopharyngeal and-neck cancer. Int J Radiat Oncol Biol Phys.
carcinoma: an update of the UCSF experience. Int J 2001;51:571–8.
Radiat Oncol Biol Phys. 2002;53:12–22. 51. Beitler JJ, Zhang Q, Fu KK, et al. Final results of
37. Sultanem K, Shu HK, Xia P, et al. Three-dimensional local-regional control and late toxicity of RTOG
intensity-modulated radiotherapy in the treatment 9003: a randomized trial of altered fractionation
of nasopharyngeal carcinoma: the University of radiation for locally advanced head and neck cancer.
California-San Francisco experience. Int J Radiat Int J Radiat Oncol Biol Phys. 2014;89:13–20.
Oncol Biol Phys. 2000;48:711–22. 52. Bernier J, Cooper JS, Pajak TF, et al. Defining
38. Lin JC, Wang WY, Liang WM, et al. Long-term risk levels in locally advanced head and neck can-
prognostic effects of plasma epstein-barr virus DNA cers: a comparative analysis of concurrent postop-
by minor groove binder-probe real-time quantitative erative radiation plus chemotherapy trials of the
PCR on nasopharyngeal carcinoma patients receiv- EORTC (#22931) and RTOG (# 9501). Head Neck.
ing concurrent chemoradiotherapy. Int J Radiat 2005;27:843–50.
Oncol Biol Phys. 2007;68:1342–8. 53. Bernier J, Domenge C, Ozsahin M, et al.
39. Wei WI, Sham JS. Nasopharyngeal carcinoma. Postoperative irradiation with or without concomi-
Lancet. 2005;365:2041–54. tant chemotherapy for locally advanced head and
40. Kam MK, Leung SF, Zee B, et al. Prospective neck cancer. N Engl J Med. 2004;350:1945–52.
randomized study of intensity-modulated radio- 54. Calais G, Alfonsi M, Bardet E, et al. Randomized
therapy on salivary gland function in early-stage trial of radiation therapy versus concomitant che-
nasopharyngeal carcinoma patients. J Clin Oncol. motherapy and radiation therapy for advanced-
2007;25:4873–9. stage oropharynx carcinoma. J Natl Cancer Inst.
41. Pow EH, Kwong DL, McMillan AS, et al. Xerostomia 1999;91:2081–6.
and quality of life after intensity-modulated radio- 55. Cooper JS, Pajak TF, Forastiere AA, et al.
therapy vs. conventional radiotherapy for early- Postoperative concurrent radiotherapy and che-
stage nasopharyngeal carcinoma: initial report on motherapy for high-risk squamous-cell car-
Head and Neck Cancers 193
cinoma of the head and neck. N Engl J Med. papillomavirus-related oropharyngeal squamous cell
2004;350:1937–44. carcinoma. JAMA Oncol. 2017;3:358–65.
56. Eisbruch A, Harris J, Garden AS, et al. Multi- 70. Huang SH, Perez-Ordonez B, Weinreb I, et al.
institutional trial of accelerated hypofractionated Natural course of distant metastases following radio-
intensity-modulated radiation therapy for early-stage therapy or chemoradiotherapy in HPV-related oro-
oropharyngeal cancer (RTOG 00-22). Int J Radiat pharyngeal cancer. Oral Oncol. 2013;49:79–85.
Oncol Biol Phys. 2010;76:1333–8. 71. Chin RI, Spencer CR, DeWees T, et al. Reevaluation
57. Grabenbauer GG, Rodel C, Brunner T, et al. of postoperative radiation dose in the management
Interstitial brachytherapy with Ir-192 low-dose-rate of human papillomavirus-positive oropharyngeal
in the treatment of primary and recurrent cancer cancer. Head Neck. 2016;38:1643–9.
of the oral cavity and oropharynx. Review of 318 72. Overgaard J, Mohanti BK, Begum N, et al. Five
patients treated between 1985 and 1997. Strahlenther versus six fractions of radiotherapy per week for
Onkol. 2001;177:338–44. squamous-cell carcinoma of the head and neck
58. Marcial VA, Pajak TF, Kramer S, et al. Radiation (IAEA-ACC study): a randomised, multicentre trial.
Therapy Oncology Group (RTOG) studies in head Lancet Oncol. 2010;11:553–60.
and neck cancer. Semin Oncol. 1988;15:39–60. 73. Koutcher L, Sherman E, Fury M, et al. Concurrent
59. Pignon JP, le Maitre A, Maillard E, et al. Meta- cisplatin and radiation versus cetuximab and radia-
analysis of chemotherapy in head and neck cancer tion for locally advanced head-and-neck cancer. Int J
(MACH-NC): an update on 93 randomised trials and Radiat Oncol Biol Phys. 2011;81:915–22.
17,346 patients. Radiother Oncol. 2009;92:4–14. 74. Chera BS, Amdur RJ, Tepper J, et al. Phase 2 trial of
60. Posner MR, Hershock DM, Blajman CR, de-intensified chemoradiation therapy for favorable-
et al. Cisplatin and fluorouracil alone or with risk human papillomavirus-associated oropharyn-
docetaxel in head and neck cancer. N Engl J Med. geal squamous cell carcinoma. Int J Radiat Oncol
2007;357:1705–15. Biol Phys. 2015;93:976–85.
61. Vermorken JB, Remenar E, van Herpen C, et al. Cisplatin, 75. Fakhry C, Zhang Q, Nguyen-Tan PF, et al. Human
fluorouracil, and docetaxel in unresectable head and papillomavirus and overall survival after progression
neck cancer. N Engl J Med. 2007;357:1695–704. of oropharyngeal squamous cell carcinoma. J Clin
62. D’Cruz AK, Vaish R, Kapre N, et al. Elective ver- Oncol. 2014;32:3365–73.
sus therapeutic neck dissection in node-negative oral 76. O’Sullivan B, Huang SH, Perez-Ordonez B, et al.
cancer. N Engl J Med. 2015;373:521–9. Outcomes of HPV-related oropharyngeal cancer
63. Kuo P, Mehra S, Sosa JA, et al. Proposing prognostic patients treated by radiotherapy alone using altered
thresholds for lymph node yield in clinically lymph fractionation. Radiother Oncol. 2012;103:49–56.
node-negative and lymph node-positive cancers of 77. Porceddu SV, Pryor DI, Burmeister E, et al.
the oral cavity. Cancer. 2016;122:3624–31. Results of a prospective study of positron emission
64. Daly ME, Le QT, Kozak MM, et al. Intensity- tomography-directed management of residual nodal
modulated radiotherapy for oral cavity squamous abnormalities in node-positive head and neck can-
cell carcinoma: patterns of failure and predic- cer after definitive radiotherapy with or without sys-
tors of local control. Int J Radiat Oncol Biol Phys. temic therapy. Head Neck. 2011;33:1675–82.
2011;80:1412–22. 78. Barker CA, Morris CG, Mendenhall WM. Larynx-
65. Peters LJ, Goepfert H, Ang KK, et al. Evaluation sparing radiotherapy for squamous cell carcinoma
of the dose for postoperative radiation therapy of from an unknown head and neck primary site. Am J
head and neck cancer: first report of a prospective Clin Oncol. 2005;28:445–8.
randomized trial. Int J Radiat Oncol Biol Phys. 79. Erkal HS, Mendenhall WM, Amdur RJ, et al.
1993;26:3–11. Squamous cell carcinomas metastatic to cervi-
66. Blanchard P, Baujat B, Holostenco V, et al. Meta- cal lymph nodes from an unknown head-and-neck
analysis of chemotherapy in head and neck cancer mucosal site treated with radiation therapy alone or
(MACH-NC): a comprehensive analysis by tumour in combination with neck dissection. Int J Radiat
site. Radiother Oncol. 2011;100:33–40. Oncol Biol Phys. 2001;50:55–63.
67. Ang KK, Zhang Q, Rosenthal DI, et al. Randomized 80. McQuone SJ, Eisele DW, Lee DJ, et al. Occult
phase III trial of concurrent accelerated radiation tonsillar carcinoma in the unknown primary.
plus cisplatin with or without cetuximab for stage III Laryngoscope. 1998;108:1605–10.
to IV head and neck carcinoma: RTOG 0522. J Clin 81. Reddy SP, Marks JE. Metastatic carcinoma in the
Oncol. 2014;32:2940–50. cervical lymph nodes from an unknown primary site:
68. O’Sullivan B, Huang SH, Su J, et al. Development results of bilateral neck plus mucosal irradiation vs.
and validation of a staging system for HPV- ipsilateral neck irradiation. Int J Radiat Oncol Biol
related oropharyngeal cancer by the International Phys. 1997;37:797–802.
Collaboration on Oropharyngeal cancer Network 82. Shoushtari A, Saylor D, Kerr KL, et al. Outcomes
for Staging (ICON-S): a multicentre cohort study. of patients with head-and-neck cancer of unknown
Lancet Oncol. 2016;17:440–51. primary origin treated with intensity-modulated
69. Husain ZA, Chen T, Corso CD, et al. A comparison radiotherapy. Int J Radiat Oncol Biol Phys.
of prognostic ability of staging systems for human 2011;81:e83–91.
194 D. M. Trifiletti et al.
83. Galloway TJ, Ridge JA. Management of squamous n onsurgical treatment strategies to preserve the lar-
cancer metastatic to cervical nodes with an unknown ynx in patients with locally advanced larynx cancer.
primary site. J Clin Oncol. 2015;33:3328–37. J Clin Oncol. 2013;31:845–52.
84. Jesse RH, Perez CA, Fletcher GH. Cervical lymph 97. Lefebvre JL, Andry G, Chevalier D, et al. Laryngeal
node metastasis: unknown primary cancer. Cancer. preservation with induction chemotherapy for
1973;31:854–9. hypopharyngeal squamous cell carcinoma:
85. Mourad WF, Hu KS, Shasha D, et al. Initial expe- 10-year results of EORTC trial 24891. Ann Oncol.
rience with oropharynx-targeted radiation therapy 2012;23:2708–14.
for metastatic squamous cell carcinoma of unknown 98. Lefebvre JL, Rolland F, Tesselaar M, et al.
primary of the head and neck. Anticancer Res. Phase 3 randomized trial on larynx preserva-
2014;34:243–8. tion comparing sequential vs alternating che-
86. Chen AM, Bucci MK, Quivey JM, et al. Long-term motherapy and radiotherapy. J Natl Cancer Inst.
outcome of patients treated by radiation therapy 2009;101:142–52.
alone for salivary gland carcinomas. Int J Radiat 99. Olsen KD. Reexamining the treatment of advanced
Oncol Biol Phys. 2006;66:1044–50. laryngeal cancer. Head Neck. 2010;32:1–7.
87. Chen AM, Bucci MK, Weinberg V, et al. Adenoid 100. Richard JM, Sancho-Garnier H, Pessey JJ, et al.
cystic carcinoma of the head and neck treated by sur- Randomized trial of induction chemotherapy in lar-
gery with or without postoperative radiation therapy: ynx carcinoma. Oral Oncol. 1998;34:224–8.
prognostic features of recurrence. Int J Radiat Oncol 101. Trotti A 3rd, Zhang Q, Bentzen SM, et al.
Biol Phys. 2006;66:152–9. Randomized trial of hyperfractionation versus con-
88. Chen AM, Garcia J, Lee NY, et al. Patterns of nodal ventional fractionation in T2 squamous cell carci-
relapse after surgery and postoperative radiation noma of the vocal cord (RTOG 9512). Int J Radiat
therapy for carcinomas of the major and minor sali- Oncol Biol Phys. 2014;89:958–63.
vary glands: what is the role of elective neck irradia- 102. Weber RS, Berkey BA, Forastiere A, et al. Outcome
tion? Int J Radiat Oncol Biol Phys. 2007;67:988–94. of salvage total laryngectomy following organ pres-
89. Laramore GE, Krall JM, Griffin TW, et al. Neutron ervation therapy: the Radiation Therapy Oncology
versus photon irradiation for unresectable salivary Group trial 91-11. Arch Otolaryngol Head Neck
gland tumors: final report of an RTOG-MRC ran- Surg. 2003;129:44–9.
domized clinical trial. Radiation Therapy Oncology 103. Yamazaki H, Nishiyama K, Tanaka E, et al.
Group. Medical Research Council. Int J Radiat Radiotherapy for early glottic carcinoma (T1N0M0):
Oncol Biol Phys. 1993;27:235–40. results of prospective randomized study of radiation
90. Mendenhall WM, Morris CG, Amdur RJ, et al. fraction size and overall treatment time. Int J Radiat
Radiotherapy alone or combined with surgery for Oncol Biol Phys. 2006;64:77–82.
adenoid cystic carcinoma of the head and neck. Head 104. Akimoto T, Nonaka T, Kitamoto Y, et al. Radiation
Neck. 2004;26:154–62. therapy for T2N0 laryngeal cancer: a retrospective
91. Mendenhall WM, Morris CG, Amdur RJ, et al. analysis for the impact of concurrent chemother-
Radiotherapy alone or combined with sur- apy on local control. Int J Radiat Oncol Biol Phys.
gery for salivary gland carcinoma. Cancer. 2006;64:995–1001.
2005;103:2544–50. 105. Beitler JJ, Muller S, Grist WJ, et al. Prognostic accu-
92. Terhaard CH, Lubsen H, Rasch CR, et al. The role racy of computed tomography findings for patients
of radiotherapy in the treatment of malignant sali- with laryngeal cancer undergoing laryngectomy. J
vary gland tumors. Int J Radiat Oncol Biol Phys. Clin Oncol. 2010;28:2318–22.
2005;61:103–11. 106. Bhayani MK, Hutcheson KA, Barringer DA, et al.
93. Amini A, Waxweiler TV, Brower JV, et al. Gastrostomy tube placement in patients with hypo-
Association of adjuvant chemoradiotherapy vs pharyngeal cancer treated with radiotherapy or
radiotherapy alone with survival in patients with chemoradiotherapy: factors affecting placement and
resected major salivary gland carcinoma: data from dependence. Head Neck. 2013;35:1641–6.
the National Cancer Data Base. JAMA Otolaryngol 107. Furusaka T, Matsuda A, Saito T, et al. Concurrent
Head Neck Surg. 2016;142:1100–10. chemoradiation therapy with docetaxel (DOC)
94. Induction chemotherapy plus radiation compared for laryngeal preservation in T2N0M0 glot-
with surgery plus radiation in patients with advanced tic squamous cell carcinomas. Acta Otolaryngol.
laryngeal cancer. The Department of Veterans 2013;133:99–112.
Affairs Laryngeal Cancer Study Group. N Engl J 108. Garden AS, Forster K, Wong PF, et al. Results of
Med. 1991;324:1685–90. radiotherapy for T2N0 glottic carcinoma: does the
95. Forastiere AA, Goepfert H, Maor M, et al. Concurrent “2” stand for twice-daily treatment? Int J Radiat
chemotherapy and radiotherapy for organ preserva- Oncol Biol Phys. 2003;55:322–8.
tion in advanced laryngeal cancer. N Engl J Med. 109. Le QT, Fu KK, Kroll S, et al. Influence of fraction
2003;349:2091–8. size, total dose, and overall time on local control of
96. Forastiere AA, Zhang Q, Weber RS, et al. Long- T1-T2 glottic carcinoma. Int J Radiat Oncol Biol
term results of RTOG 91-11: a comparison of three Phys. 1997;39:115–26.
Head and Neck Cancers 195
110. Machtay M, Moughan J, Trotti A, et al. Factors 124. Ballo MT, Strom EA, Zagars GK, et al. Adjuvant
associated with severe late toxicity after c oncurrent irradiation for axillary metastases from malig-
chemoradiation for locally advanced head and nant melanoma. Int J Radiat Oncol Biol Phys.
neck cancer: an RTOG analysis. J Clin Oncol. 2002;52:964–72.
2008;26:3582–9. 125. Beadle BM, Guadagnolo BA, Ballo MT, et al.
111. Janoray G, Pointreau Y, Garaud P, et al. Long- Radiation therapy field extent for adjuvant treatment
term results of a multicenter randomized phase of axillary metastases from malignant melanoma. Int
III trial of induction chemotherapy with Cisplatin, J Radiat Oncol Biol Phys. 2009;73:1376–82.
5-fluorouracil, +/− Docetaxel for Larynx preserva- 126. Bonnen MD, Ballo MT, Myers JN, et al. Elective
tion. J Natl Cancer Inst. 2016;108:1–7. radiotherapy provides regional control for patients
112. Brierley JD, Panzarella T, Tsang RW, et al. A com- with cutaneous melanoma of the head and neck.
parison of different staging systems predictability of Cancer. 2004;100:383–9.
patient outcome. Thyroid carcinoma as an example. 127. Burmeister BH, Mark Smithers B, Burmeister E,
Cancer. 1997;79:2414–23. et al. A prospective phase II study of adjuvant post-
113. Chen J, Tward JD, Shrieve DC, et al. Surgery and operative radiation therapy following nodal surgery
radiotherapy improves survival in patients with ana- in malignant melanoma-Trans Tasman Radiation
plastic thyroid carcinoma: analysis of the surveil- Oncology Group (TROG) Study 96.06. Radiother
lance, epidemiology, and end results 1983-2002. Am Oncol. 2006;81:136–42.
J Clin Oncol. 2008;31:460–4. 128. Chang DT, Amdur RJ, Morris CG, et al. Adjuvant
114. De Crevoisier R, Baudin E, Bachelot A, et al. radiotherapy for cutaneous melanoma: comparing
Combined treatment of anaplastic thyroid carcinoma hypofractionation to conventional fractionation. Int
with surgery, chemotherapy, and hyperfractionated J Radiat Oncol Biol Phys. 2006;66:1051–5.
accelerated external radiotherapy. Int J Radiat Oncol 129. Overgaard J. The role of radiotherapy in recur-
Biol Phys. 2004;60:1137–43. rent and metastatic malignant melanoma: a clinical
115. Haigh PI, Ituarte PH, Wu HS, et al. Completely radiobiological study. Int J Radiat Oncol Biol Phys.
resected anaplastic thyroid carcinoma com- 1986;12:867–72.
bined with adjuvant chemotherapy and irradia- 130. Overgaard J, Gonzalez Gonzalez D, Hulshof MC,
tion is associated with prolonged survival. Cancer. et al. Randomised trial of hyperthermia as adjuvant
2001;91:2335–42. to radiotherapy for recurrent or metastatic malig-
116. Kebebew E, Greenspan FS, Clark OH, et al. nant melanoma. European Society for Hyperthermic
Anaplastic thyroid carcinoma. Treatment outcome Oncology. Lancet. 1995;345:540–3.
and prognostic factors. Cancer. 2005;103:1330–5. 131. Sause WT, Cooper JS, Rush S, et al. Fraction size
117. Robbins RJ, Schlumberger MJ. The evolving role of in external beam radiation therapy in the treat-
(131)I for the treatment of differentiated thyroid car- ment of melanoma. Int J Radiat Oncol Biol Phys.
cinoma. J Nucl Med. 2005;46(Suppl 1):28S–37S. 1991;20:429–32.
118. Rosenbluth BD, Serrano V, Happersett L, et al. 132. Agrawal S, Kane JM 3rd, Guadagnolo BA, et al. The
Intensity-modulated radiation therapy for the treat- benefits of adjuvant radiation therapy after therapeu-
ment of nonanaplastic thyroid cancer. Int J Radiat tic lymphadenectomy for clinically advanced, high-
Oncol Biol Phys. 2005;63:1419–26. risk, lymph node-metastatic melanoma. Cancer.
119. Tennvall J, Lundell G, Hallquist A, et al. Combined 2009;115:5836–44.
doxorubicin, hyperfractionated radiotherapy, and 133. Eggermont AM, Chiarion-Sileni V, Grob JJ, et al.
surgery in anaplastic thyroid carcinoma. Report on Adjuvant ipilimumab versus placebo after complete
two protocols. The Swedish Anaplastic Thyroid resection of high-risk stage III melanoma (EORTC
Cancer Group. Cancer. 1994;74:1348–54. 18071): a randomised, double-blind, phase 3 trial.
120. Tsang RW, Brierley JD, Simpson WJ, et al. The Lancet Oncol. 2015;16:522–30.
effects of surgery, radioiodine, and external radia- 134. Kirkwood JM, Manola J, Ibrahim J, et al. A pooled
tion therapy on the clinical outcome of patients analysis of eastern cooperative oncology group and
with differentiated thyroid carcinoma. Cancer. intergroup trials of adjuvant high-dose interferon for
1998;82:375–88. melanoma. Clin Cancer Res. 2004;10:1670–7.
121. Ang KK, Peters LJ, Weber RS, et al. Postoperative 135. Balch CM, Soong SJ, Smith T, et al. Long-term
radiotherapy for cutaneous melanoma of the head results of a prospective surgical trial comparing
and neck region. Int J Radiat Oncol Biol Phys. 2 cm vs. 4 cm excision margins for 740 patients with
1994;30:795–8. 1-4 mm melanomas. Ann Surg Oncol. 2001;8:101–8.
122. Ballo MT, Bonnen MD, Garden AS, et al. Adjuvant 136. Wu S, Cho E, Li WQ, et al. History of severe sun-
irradiation for cervical lymph node metastases from burn and risk of skin cancer among women and men
melanoma. Cancer. 2003;97:1789–96. in 2 prospective cohort studies. Am J Epidemiol.
123. Ballo MT, Garden AS, Myers JN, et al. Melanoma 2016;183:824–33.
metastatic to cervical lymph nodes: can radiother- 137. Thomas JM, Newton-Bishop J, A’Hern R, et al.
apy replace formal dissection after local excision of Excision margins in high-risk malignant melanoma.
nodal disease? Head Neck. 2005;27:718–21. N Engl J Med. 2004;350:757–66.
196 D. M. Trifiletti et al.
138. Hodi FS, O’Day SJ, McDermott DF, et al. Improved 146. Mojica P, Smith D, Ellenhorn JD. Adjuvant radia-
survival with ipilimumab in patients with metastatic tion therapy is associated with improved survival
melanoma. N Engl J Med. 2010;363:711–23. in Merkel cell carcinoma of the skin. J Clin Oncol.
139. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab 2007;25:1043–7.
plus dacarbazine for previously untreated metastatic 147. Poulsen M, Rischin D, Walpole E, et al. High-risk
melanoma. N Engl J Med. 2011;364:2517–26. Merkel cell carcinoma of the skin treated with syn-
140. Avril MF, Auperin A, Margulis A, et al. Basal
chronous carboplatin/etoposide and radiation: a
cell carcinoma of the face: surgery or radiother- Trans-Tasman Radiation Oncology Group Study--
apy? Results of a randomized study Br J Cancer. TROG 96:07. J Clin Oncol. 2003;21:4371–6.
1997;76:100–6. 148. Lewis KG, Weinstock MA, Weaver AL, et al.
141. Balamucki CJ, Mancuso AA, Amdur RJ, et al. Skin Adjuvant local irradiation for Merkel cell carci-
carcinoma of the head and neck with perineural inva- noma. Arch Dermatol. 2006;142:693–700.
sion. Am J Otolaryngol. 2012;33:447–54. 149. Ghoshal S, Chakraborty S, Moudgil N, et al. Quad
142. Garcia-Serra A, Hinerman RW, Mendenhall WM, shot: a short but effective schedule for palliative
et al. Carcinoma of the skin with perineural invasion. radiation for head and neck carcinoma. Indian J
Head Neck. 2003;25:1027–33. Palliat Care. 2009;15:137–40.
143. Mendenhall WM, Parsons JT, Mendenhall NP, et al. 150. Nguyen NT, Doerwald-Munoz L, Zhang H, et al.
T2-T4 carcinoma of the skin of the head and neck 0-7-21 hypofractionated palliative radiotherapy: an
treated with radical irradiation. Int J Radiat Oncol effective treatment for advanced head and neck can-
Biol Phys. 1987;13:975–81. cers. Br J Radiol. 2015;88:20140646.
144. Rogers HW, Weinstock MA, Harris AR, et al. 151. Petersen IA, Kriss JP, McDougall IR, et al. Prognostic
Incidence estimate of nonmelanoma skin cancer in the factors in the radiotherapy of Graves’ ophthalmopa-
United States, 2006. Arch Dermatol. 2010;146:283–7. thy. Int J Radiat Oncol Biol Phys. 1990;19:259–64.
145. Mendenhall WM, Kirwan JM, Morris CG, et al.
Cutaneous Merkel cell carcinoma. Am J Otolaryngol.
2012;33:88–92.
Lung Cancer
5
Nicholas G. Zaorsky, Daniel M. Trifiletti,
and Henry Wagner Jr
Abstract
This chapter discusses the general manage-
ment of patients with thoracic cancers, with
special focus on principles that guide radio-
therapy management. Several key components
of trimodality care and stereotactic body radi-
ation therapy (SBRT) are discussed.
FVC
Lung Cancer 199
NSCLC: Post-op Therapy [22, 33, 34] PORT not mandatory, given per each center’s
policy. Chemo had improved OS, benefit to
Adjuvant Chemo Stage II/IIIA (pN+), no benefit to Stage
–– CALGB 9633 (Strauss, JCO, 2008): Stage IB I. Chemo and PORT had best outcomes, and
T2 N0 randomized to carbo/taxol vs obs. PORT is better than obs. PORT techniques
Stopped early because of OS benefit, though relatively modern.
not SS. Exploratory analysis: adjuvant chemo –– LCSG 773: T3 or N2 → obs vs PORT (50Gy).
improves outcomes if primary ≥4 cm (contro- PORT improved LR (41 → 3%) but same OS.
versial) or ≥5 cm (higher SS). Thus, while not –– PORT meta-analysis Trialists Group, 1998:
level I evidence, should be discussed w ten trials. PORT→↓OS, mostly in Stages I–
patient. II. Included old techniques (Co-60 in four tri-
als), large fraction sizes, high total doses
Adjuvant BT delivered, mostly early stage. No detriment to
–– Allegheny Gen Hosp (Colonias, 2011): pN2.
Retrospective. Stage I NSCLC get sublobar –– SEER (Lally, 2006): Benefit for pN2 patients,
resection and interop I-125 mesh. 1996–2008. HR of 0.86 for death.
n = 145. Mean dose 117 Gy to 33 cm2. Periop –– No benefit to CRT over PORT alone (INT
mortality 3%. LR 4%, 6% w regional failure, 0115/RTOG 9105/ECOG 3590).
17% w DMs. On MVA, no factors predictive –– RTOG 0214: Evaluated PCI. Stage III NSCLC
of LR s/p definitive thoracic treatment. PCI 30 Gy/15
–– ACOSOG Z4032 (Fernando, 2013). n = 224. reduced brain failure (18% vs 8%), but same
Sublobar resection +/− BT. No improvement OS 76%. No diff in 1 year for QOL, MMSE,
in LR w BT, at 8% or ADLs; however, there were neurocog dif-
ferences at 3 months (major difference
Adjuvant Chemo and RT between eligibility criteria and in positive
–– LACE meta-analysis: 5% for OS at 5 yrs for SCLC trials).
Stages I–II after resection. Most benefit in N1
pts. Subset analysis: no benefit for chemo in
IA/B dz. However, CALGB 9633 data not Technique
included in this trial. –– Need to ask what LN stations were
–– Pre- vs post-op chemo: no difference (EORTC dissected.
08012, CHEST trial, NATCH Spanish trial). –– Lung Adjuvant RT Trial (Lung ART): RCT for
–– Italian study (Trodella, 2002): 104 pts, pN2 to PORT vs obs provides guidelines. In
pN0 → PORT vs obs. 50.4 Gy. PORT all patients, bronchial stump, ipsi hilum,
improved LF (23 → 2%) and 5-yr OS extension to mediastinal pleura facing resec-
(58 → 67%). tion bed always in CTV. Also cover involved
–– ANITA trial + reanalysis (Arriagada, 2004; LN level, LN levels at risk.
Douillard, 2008): NSCLC w surgery, R0. –– For N0–N1 R1, cover region of R1. For N2,
Adjuvant vinorelbine + cis vs observation. cover involved LN region.
210 N. G. Zaorsky et al.
• 1-year LR 15% vs 25%, favoring 60 Gy. –– CALGB 39801 (Vokes, JCO, 2007):
• 2-year LR 31% vs 39%, favoring 60 Gy. Unresectable Stage III. CRT to 66 Gy +/−
• G3+ tox similar bw RT groups. Esophagitis induction chemo (carbo/taxol). Chemo was
worse w 74 Gy, 21% vs 7%. carbo AUC 2 and paclitaxel 50 mg/m2. Same
• +/− cetux had the same OS. OS, around 30%. Toxicity worse during induc-
• G3+ tox worse w cetux, 86% vs 70%. tion, 38% w G3+ neutropenia.
• On MVA, factors predicting OS were RT
dose (60 Gy), max esophagitis grade, PTV,
heart V5, and heart V30. Sequential Chemo Versus Concurrent Chemo
• On MVA, predictors of G3+ pneumonitis –– LAMP trial: three arms – Dillman
were RT technique, Stage IIIA vs IIIB, and (chemo→RT) vs (chemo→CRT) vs
lung V20. (CRT → chemo). Chemo was carbo/taxol.
• RT and chemo compliance, RT technique, CRT → chemo improved MS
not significant for OS. IMRT did reduce (13 → 16.3 → 12.7 m).
pneumonitis. –– Auperin meta-analysis (Auperin, JCO,
–– RTOG 0617 secondary analysis (Chun, 2016): 2010): sequential vs. concurrent CRT for LA
IMRT associated w less G3+ pneumonitis NSCLC – At 5 y, OS was 15.1% vs 10.6%,
than 3D CRT (3% vs 8%). No difference in 4.5% absolute improvement for concurrent.
OS, PFS, LR, DMFS. IMRT assoc w lower Also has decreased LRP (HR 0.77) but higher
heart dose. Heart V40 associated w OS. Lung G3 esophagitis (18% vs 4%).
V5 not assoc w G3+ toxicity, but V20 was.
–– CHART: 54 Gy at 1.5Gy TID (x12 consec Consolidation Chemo
days) vs 60/30. TID improved 3-yr OS by –– SWOG 9504 (Gandara, 2003): path IIIB
10% but ↑tox. Mostly SCC. NSCLC. Concurrent CRT. Chemo is cis
50 mg/m2 d1, 8, 29, 36 and etoposide 50 mg/
Induction Chemo + RT vs RT Alone m2 d 1–5, 29–33. Concurrent TRT to 61 Gy.
–– Induction generally only appropriate if upfront Consolidation docetaxel started 4–6w after
CRT plan exceeding DVH constraints. CRT, at initial dose 75 mg/m2. Did not improve
–– CALGB 8433 (Dillman, 1990, 1996): OS.
IIIA→60/30 vs cis/vinblast induction +60/30 –– HOG: IIIA/B cis/etop x2c with RT 60 Gy + −
(sequential). Cis was 100 mg/mq q28d and consolid docetaxel. No diff OS, more
vinblast was 5 mg/m2 qw. Sequential CRT toxicity.
improved ↑MS (10 → 14 m). 5-yr OS –– SWOG: IIIA/B cis/etop x2c with RT 61 Gy
(7 → 19%). - > docetaxel + − gefitinib (Iressa). Worse OS
–– RTOG 9410 (Curran, 2011): Dillman trial with gefitinib.
worked, so now will concurrent chemo beat
sequential? Three arms→Dillman to 63Gy vs Immunotherapy
concurrent CRT to 63Gy vs 69.6Gy CRT –– Checkmate 057, Borghaei, 2015. Nivolumab
BID. Chemo was cis/vinblast, but BID arm vs docetaxel in M+. Nivo improved 1-y OS
got cis/etop. Conventional CRT improved MS 51% vs 39%, MST 12 vs 9 m.
(14.6 → 17 → 15.6 m). More tox with chemo.
This RCT defined current SOC. (not cis/vbl). Toxicity
–– RTOG 8808 (Sause, 2000): II–IIIB→ 60/30 –– Palma, 2013: 1082 patients received concur-
vs 69.6 BID vs cis/vinblast induction +60/30 rent CRT from 4 continents. V60 was best pre-
(Dillman style). CRT improved MS dictor of esophagitis. Cut points of
(11.4 → 12 → 13.2 m). V60 <0.07%, 0.07–17, and >17%. Thus, keep
V60 <17%.
Lung Cancer 213
Superior Sulcus Tumors CRT to 45 Gy/25 with cis 50 mg/m2 + etop
(Pancoast) [63] 50 mg/m2. Repeat CT during last week of RT
(week 5) to allow surgeon to make decision
Staging regarding resectability. If ineligible, continue
–– By definition at least T3 RT to 61.2/1.8. Then surgery and post-op
chemo. 20% pts unresectable and went on to
Symptoms complete RT to 60 Gy. Of 80% who went to
–– Pancoast triad: (1) shoulder pain, (2) brachial surg, 94% had R0 resection, 29% had pCR,
plexus palsy, (3) Horner’s syndrome (ptosis, and 26% had minimal microscopic residual,
meiosis, enophthalmos, and ipsilateral with a combined rate of 56% for response.
anhidrosis) 5-yr OS 44%. Recurrence: distant (non-brain)
–– Pancoast syndrome: pain in C8/T1, lower bra- only 33%, brain only 33%, local only 17%,
chial plexopathy from invasion of stellate gan- local + distant 12%. Created SOC for these
glion, + chest wall (rib invasion), has to be at tumors.
least T3 –– SWOG 0220 EP concurrent, docetaxel
consolidation.
Outcomes (NCI data)
–– 2-yr OS: 55%, 5-yr OS = 44%
–– 3-y/5-y OS: 61%/56% Treatment
–– 3-y OS: 56% –– Trimodality: neoadj cis/etop +45Gy
–– Check CT at last week of RT
Studies –– If resectable, → surgery →EP consolidation or
–– INT study (Loehrer, 1997). Evaluating PAC docetaxel per 0220
chemo. Pts get 2–4c q3w cis (50 mg/m2), –– If unresectable at 45Gy, go to 60–66Gy + adj
doxorubicin (50 mg/m2), and cyclophospha- chemo. Thus, need to save immobilization
mide (500 mg/m2). For those without PD, devices until surgeon reviews CTs and pt has
mediastinal RT to 54 Gy was delivered. surgery. No evidence for boost in patients w
n = 23, with 5 CRs and 11 PRs to chemo. RT R+ resection (per 0160)
converted 80% of pts w SD to a CR or PR. –– Contraindications to surgery: involves bra-
5-year OS 53%. chial plexus, subclavian artery, VB, esopha-
–– SWOG 9416/INT 0160 (Rusch, 2007): supe- gus, mediastinal LNs, or DMs
rior sulcus phase II of T3/4 N0/1 induction
Lung Cancer 215
Studies
Xie, 2012: SEER. Improved outcomes w RT. If
treated w RT alone, MST 12 m vs 5 m w/ RT
Diagnostic
–– Broch. Mediastinoscopy if LNs on CT
Neuroendocrine
–– Typical carcinoid – (<2 mitoses/10 hpf)
–– Atypical carcinoid – (2–10 mitoses/10 hpf
or + necrosis)
–– Large cell neuroendocrine carcinoma (>=11
mitoses/10 hpf, large polygonal cells,
necrosis)
–– Small cell lung cancer (15% of lung cancers)
Lung Cancer 219
inferior). The better comparison is 70 and before RT. Mostly verbal memory, frontal
45 Gy BID. MDACC arm was less acceptable. lobe dysfunction, fine motor coordination. In
PCI is given 3–6 w after completion of all 4 c 11 pts who had post-RT testing, no SS differ-
chemo. ence in neuro status pre vs post RT.
–– CONVERT trial (Faivre-Finn, 2017): –– NRG hippocampal avoidance trial: improved
International. Concurrent CRT w 45 Gy BID Hopkins verbal learning.
vs 66 QD for LS-SCLC and ECOG 0–1. –– Takahashi, 2017: PCI in ES-SCLC with neg
ASCO 2016: 2-y OS 56% vs 51% and MST MRI prior to randomization. PCI did not
30 m vs 25 m (NSS). No difference in toxicity. improve OS.
Currently supporting either regimen as SOC.
–– RTOG 0239: phase II → RT to 61.2 Gy, par- Extensive Stage (ES-SCLC, i.e., Stage IV)
tially BID. Improved OS compared to Turrisi. –– 9-Month OS
–– CALGB 8837: phase I trial, dose escalated to –– Definition: can’t fit in one RT port.
over 70 Gy with good results. –– Yugoslavia, Jeremic 1999: PR or CR after
–– RTOG 0538: pending. Turrisi vs RTOG 0329 chemo→ chest RT vs more chemo. RT
(0239) vs 70/35 (CALGB 8837). This trial improved 5-yr OS (3.7 → 9.1%).
includes ENI. The spinal cord tolerance is –– Bonner, Mayo, 1995. ES-SCLC gets chemo.
36 Gy in the BID arm. If PR or CR, +/−TRT. 3/19 pts alive
>5 years.
Induction Chemo –– CREST (Chest Radiotherapy Extensive-Stage
–– SWOG 7924 (Kies, 1987): All got induction Small Cell Lung Cancer Trial), Slotman 2014:
chemo for LS-SCLC and then randomize to ES-SCLC get chemo. If PR or CR → +/− tho-
post-op tx. If CR, then rando to TRT or further racic RT (30/10). PCI is for all. 90% of pts had
chemo. Those with PR or had SD to induction residual thoracic dz after initial chemo, only
were rando to TRT to either pre-induction 5% w CR. For TRT, PTV = post-chemo vol-
(“wide field”) or post-induction (“reduced ume + 1.5 cm margin, also all LN stations
field”) target volume. No difference in OS, involved pre-chemo, including hilum + medi-
patterns of failure astinum, even w CR. Lung (defined as lungs
minus PTV) V20 <35%. Pts only get CNS
PCI imaging if there are clinical sx. 1-year OS
–– 5.4% OS3 absolute improvement. 25 Gy/10 fx. similar 28–33%, NSS. However, RT improved
–– Contraindications: poor mental status, stable, 2-yr OS (3 → 13%). Intrathoracic PFS
or progressing disease. improved 20% vs 47%. Toxicity similar be
–– Auperin 1999: meta-analysis of PCI. PCI arms, G3+ esophagitis <2%.
reduced 3-yr brain mets (59 → 33%) and –– Giuliani, PMH, 2011: ES-SCLC pts get con-
increased 3-yr OS (15.3 → 20.7%). Better if solidative TRT, 30+ Gy. LR was 26% in 1 y
given early. and 39% in 2 y.
–– Le Pechoux 2003: randomized LS cCR –– RTOG 0937: ES-SCLC w up to four extracra-
patients to 25/10 vs 36/18 PCI. Same brain nial mets get PCI 25/10 +/− consolidative
met rate, worse OS in high dose. TRT and met-directed RT. Must have at least
–– EORTC 08993 (Slotman 2007): ES with any radiographic PR in one site and no PD. Can
response to 4–6c chemo→ +/− PCI. PCI have max four extracranial metastatic lesions.
improved 1-yr OS (13 → 27%). No routine Rec max dose is 45 Gy/15 fx at 3 Gy.
MRIs done. Extracranial progression Alternative is 30–40 Gy/10 fx. Closed due to
unchanged (89% vs 93%, NSS). poor accrual and futility boundary for
–– Komaki, 1995: MDACC neurocog study. 97% OS. High G4+ toxicity in TRT arm.
of pts had neuro decline after chemo but
Lung Cancer 221
On Treatment
–– Daily KV imaging
222 N. G. Zaorsky et al.
Stage Tx paradigm
SIADH and hypoNa Fluid restriction
Give hypertonic saline, correct 1–2 nmol/L/h
Demeclocycline
Antineoplastic therapy
Vasopressin receptor inhibitors
Cushing syndrome Consider ketoconazole and then metyrapone
Consider before initiation of antineoplastic tx
T1–2 N0 Mediastinal sampling. If neg → lobectomy and node dissection
pN0: post-op chemo
pN+: post-op CRT
Still recommend PCI
LS-SCLC: “Concurrent platin doublet + RT” (Turrisi and Pignon meta-analysis)
“fits in one port” Four cycles of EP chemo: etoposide, 120 mg/m2, d1–3 + cisplatin, 60 mg/m2, d1 q3w
Concurrent RT at 1st cycle if possible. Some pts w massive LAD may need 1–2 cycle
induction chemo before TRT. In this case, RT is to post-induction volume, no apparent
increase in marginal failure (SWOG 7924, Kies, 1987; Liengswanwong, JCO, 1994)
RT: 45Gy/1.5Gy/fx >6 h apart or 60–70 Gy 1.8–2Gy (Turrisi or CALGB 39808)
“Early RT” (Murray and Fried meta): start date to end date of RT should be 3 weeks w
BID
Wait for 3–6 w after all chemo, and do CT chest and MRI brain. If CR or PR, then PCI,
25 Gy/10 fx
If progression use topotecan
LS-SCLC w SVC Try chemo first and then proceed per above.
syndrome
ES-SCLC: “doesn’t” Platin doublet, four cycle EP chemo (80/80)
Then CT chest, to bottom of adrenals and MRI brain
If CR or PR: PCI for all. Concurrent consolidative TRT to 30 Gy in ten fractions to
GTV + 1.5 cm and ENI if involved up front (EORTC CREST/Slotman)
RT to other sites not recommended (RTOG 0937)
If progression: topotecan
M1, non-brain met SVC/lobar obstruction/bone mets: Chemo + RT
Cord compression: palliative RT first
M1, brain met Asx: may do chemo 1st, then WBRT
Sx: WBRT and then chemo
Lung Cancer 223
RT details
Margin Histology Post-op paradigm (“just say 50 or 60”)
R0 Thymoma or I: Surveillance w CT chest q6–12 months for 2 y and
carcinoma then annually for 5y for carcinoma and 10 y for
thymoma
Thymoma II–IV: +/− post-op RT Stage II R0–1 B2–3:
45–50 Gy
Carcinoma II–IV: + post-op RT (cat 2b) Stage II R0–1 B3: 50–54Gy
Note R0 for Masaoka Stage
III difficult to achieve
R1 Thymoma Post-op RT 50–54Gy
Carcinoma Post-op RT +/− chemo 50–54Gy
Then surveillance w CT chest q6–12 months for 2 y
and then annually for 5 y for carcinoma and 10 y for
thymoma
R2 Thymoma Post-op RT +/− chemo 50–54Gy
Carcinoma Post-op RT + chemo Unresectable or R2,
60–70 Gy
Unresectable Chemo first. Then CT with contrast and PET-CT. Then
reconsider for resection. If unresectable, then RT
+/− chemo
226 N. G. Zaorsky et al.
AJCC8 AJCC8 T1 T2 T3 T4
• T1 – ipsilateral pleural disease only N0 IA IB
• T2 – into diaphragm muscle or pulmonary N1 II IIIA IIIB
parenchyma
N2 IIIB
• T3 – endothoracic fascia, mediastinal fat,
chest wall, outer pericardium (potentially M1 IV
resectable)
• T4 – multifocal, into peritoneum, contralateral pleura, mediastinal organ, spine, internal pericar-
dium (unresectable)
–– N1 – ipsi hilar/mediastinal
–– N2 – contralateral or SVC
AJCC7
• T1 AJCC7 T1a T1b T2 T3 T4
• T1a – ipsi parietal pleura, no visceral N0 IA IB II III
• T1b – ipsi parietal pleura, +visceral N1 IV
• T2 – into diaphragm muscle or pulmonary III
N2
parenchyma
• T3 – endothoracic fascia, mediastinal fat, N3
IV
chest wall, outer pericardium M1
• T4 – multifocal, into peritoneum, contralateral
pleura, mediastinal organ, spine, internal pericardium
• N1 – ipsi hilar
• N2 – ipsi mediastinal/IM, or subcarinal, or peridiaphragmatic
• N3 – contra mediastinal/IM, supraclav
Work-Up Surgery
–– H&P – asbestos exposure, if transthoracic –– Extrapleural pneumonectomy (EPP)
extension, then SC/Ax LNs at increased risk. removes parietal/visceral pleura, lung, medi-
–– CT CAP. astinal nodes, pericardium, and ipsi dia-
–– MRI chest/abd to rule diaphragm invasion. phragm. ~5–15% mortality. 2-year OS is 35%.
–– Pleural bx: VATS preferred over open. Needle Contraindicated if extension through
bx can seed track. diaphragm.
–– If surgery planned (clinical Stages I–III):
Lung Cancer 227
–– Pleurectomy/decortication (P/D): more like –– *Avoid aggressive surgery if poor PS, N+,
debulking, resection of pleura w/o lung or sarcomatoid b/c poor prognosis
removal. Possible removal/recon of dia- • For N+, palliative chemo is the SOC.
phragm or pericardium. Only 2% operative –– Chemo: anti-folate/platinum doublet (e.g., cis/
mortality but only curative for T1, so it is pem).
reserved for early stage. Adjuvant RT limited –– Thymic carcinoma needs adjuvant therapy.
by remaining underlying intact lung.
Radiation
Adj RT –– Post-op after EPP: R0: 50–54Gy; R+
–– Adj RT typically part of package after EPP, 54–60Gy. 41.4 Gy is prescribed to contra
but some centers have published good out- vertebral.
comes of post-op IMRT and IMPT after P/D. –– Prophylactic to prevent tract recurrence:
–– Rusch 2001: phase II → EPP and hemitho- 21Gy/3 fx at 7 Gy.
racic 54 Gy. MS 34 m for Stages I–II, 10 m for
Stages III–IV. Technique
–– Flores 2006: induction gem/ –– Sim w supine arms akimbo since tx usually
cis → EPP → 54Gy. MS 33 m. AP PA and en face electron patches under
–– Rosenzweig (2012) Good review esp toxicity shielding.
of IMRT and ways to minimize this. –– Simulation, CTV:
–– Sup: top of T1
Chemotherapy –– Inf: bottom of L2
–– Vogelzang 2003: metastatic→cis +/−peme- –– Lat: flash skin
trexed. Pem is 500 mg/m2 and cis is 75 mg/m2 –– Medial: 1.5–2 cm beyond contra VB
q3w. This is superior to cis alone in patients border
unfit for surg for MST (12 vs 9 m), PFS (6 vs –– Stomach (for L-sided lesion) and liver (for
4 m), and radiographic response (41% vs R-sided lesion) blocked through entire hemi-
17%). thoracic RT course.
–– If on L, ant heart block at 19.8 Gy to prevent
RT pericarditis.
–– O’Rourke: n = 61. Rate of tract seeding simi- –– Cord blocked after 41.4 Gy.
lar bw RT arm (13%) and obs (10%). Questions –– Electron field patch is used centrally to sup-
need for prophylactic tract RT. plement pleural space under block, to treat the
diaphragmatic recesses around the liver. Dose
NCCN usually 1.53 Gy/d. This dose is used because
Early Stage/Resectable photon field scatters 15% of dose to blocked
–– Highly controversial. Radical pleurectomy portion. Electrons prescribed to 90% IDL.
increasingly option over EPP. RT after radical
pleurectomy only on trial. Constraints
–– If RT after P/D: lung V20Gy <20% and
Advanced Stage/Unresectable MLD <9.5 Gy
–– Induction chemo (cis and pem) →surgery → –– Hemithoracic RT 4–8 wks post-op to 54 Gy
+/−RT. –– Contralateral lung V20 <7%, V5 <50%,
–– Surgery (definitive/palliative): mean <8.5Gy
–– EPP –– Heart V40Gy <50%
–– P/D
228 N. G. Zaorsky et al.
tic body radiotherapy. Int J Radiat Oncol Biol Phys. favorable patients with Radiation Therapy Oncology
2010;76:796–801. Group stage III non-small-cell lung carcinoma: report
29.
Stephans KL, Djemil T, Reddy CA, et al. of Radiation Therapy Oncology Group 83-11. J Clin
Comprehensive analysis of pulmonary function Test Oncol. 1990;8:1543–55.
(PFT) changes after stereotactic body radiotherapy 41. Dillman RO, Seagren SL, Propert KJ, et al. A random-
(SBRT) for stage I lung cancer in medically inoper- ized trial of induction chemotherapy plus high-dose
able patients. J Thorac Oncol. 2009;4:838–44. radiation versus radiation alone in stage III non-small-
30. Videtic GM, Stephans K, Reddy C, et al. Intensity- cell lung cancer. N Engl J Med. 1990;323:940–5.
modulated radiotherapy-based stereotactic body 42. Gore EM, Bae K, Wong SJ, et al. Phase III compari-
radiotherapy for medically inoperable early-stage son of prophylactic cranial irradiation versus observa-
lung cancer: excellent local control. Int J Radiat tion in patients with locally advanced non-small-cell
Oncol Biol Phys. 2010;77:344–9. lung cancer: primary analysis of radiation therapy
31. Nagata Y, Takayama K, Matsuo Y, et al. Clinical out- oncology group study RTOG 0214. J Clin Oncol.
comes of a phase I/II study of 48 Gy of stereotactic 2011;29:272–8.
body radiotherapy in 4 fractions for primary lung can- 43. Keller SM, Adak S, Wagner H, et al. A randomized
cer using a stereotactic body frame. Int J Radiat Oncol trial of postoperative adjuvant therapy in patients with
Biol Phys. 2005;63:1427–31. completely resected stage II or IIIA non-small-cell
32. Hara R, Itami J, Kondo T, et al. Clinical outcomes of lung cancer. Eastern Cooperative Oncology Group. N
single-fraction stereotactic radiation therapy of lung Engl J Med. 2000;343:1217–22.
tumors. Cancer. 2006;106:1347–52. 44. Perez CA, Stanley K, Rubin P, et al. Patterns of tumor
33. Strauss GM, Herndon JE 2nd, Maddaus MA, et al. recurrence after definitive irradiation for inoperable
Adjuvant paclitaxel plus carboplatin compared with non-oat cell carcinoma of the lung. Int J Radiat Oncol
observation in stage IB non-small-cell lung cancer: Biol Phys. 1980;6:987–94.
CALGB 9633 with the Cancer and Leukemia Group 45. Rosell R, Gomez-Codina J, Camps C, et al.
B, Radiation Therapy Oncology Group, and North Preresectional chemotherapy in stage IIIA non-small-
Central Cancer Treatment Group Study Groups. J cell lung cancer: a 7-year assessment of a randomized
Clin Oncol. 2008;26:5043–51. controlled trial. Lung Cancer. 1999;26:7–14.
34. Spoelstra FO, Senan S, Le Pechoux C, et al. Variations 46. Roth JA, Atkinson EN, Fossella F, et al. Long-term
in target volume definition for postoperative radio- follow-up of patients enrolled in a randomized trial
therapy in stage III non-small-cell lung cancer: analy- comparing perioperative chemotherapy and surgery
sis of an international contouring study. Int J Radiat with surgery alone in resectable stage IIIA non-small-
Oncol Biol Phys. 2010;76:1106–13. cell lung cancer. Lung Cancer. 1998;21:1–6.
35.
Postoperative radiotherapy for non-small cell 47. Rusch VW, Giroux DJ, Kraut MJ, et al. Induction
lung cancer. Cochrane Database Syst Rev. 2005; chemoradiation and surgical resection for non-small
CD002142. cell lung carcinomas of the superior sulcus: initial
36. PL03.05. An intergroup randomized phase III com- results of Southwest Oncology Group Trial 9416
parison of standard-dose (60 Gy) vs high-dose (74 (Intergroup Trial 0160). J Thorac Cardiovasc Surg.
Gy) chemoradiotherapy (CRT) +/− cetuximab (cetux) 2001;121:472–83.
for stage III non-small cell lung cancer (NSCLC): 48. Sause W, Kolesar P, Taylor SI, et al. Final results
results on cetux from RTOG 0617. Clin Adv Hematol of phase III trial in regionally advanced unresect-
Oncol. 2014;12(2–4). able non-small cell lung cancer: Radiation Therapy
37. Albain KS, Swann RS, Rusch VW, et al. Radiotherapy Oncology Group, Eastern Cooperative Oncology
plus chemotherapy with or without surgical resection Group, and Southwest Oncology Group. Chest.
for stage III non-small-cell lung cancer: a phase III ran- 2000;117:358–64.
domised controlled trial. Lancet. 2009;374:379–86. 49. Thomas M, Rube C, Hoffknecht P, et al. Effect of
38. Ball D, Bishop J, Smith J, et al. A randomised phase preoperative chemoradiation in addition to pre-
III study of accelerated or standard fraction radio- operative chemotherapy: a randomised trial in
therapy with or without concurrent carboplatin in stage III non-small-cell lung cancer. Lancet Oncol.
inoperable non-small cell lung cancer: final report 2008;9:636–48.
of an Australian multi-centre trial. Radiother Oncol. 50. van Meerbeeck JP, Kramer GW, Van Schil PE, et al.
1999;52:129–36. Randomized controlled trial of resection versus
39. Belani CP, Choy H, Bonomi P, et al. Combined
radiotherapy after induction chemotherapy in stage
chemoradiotherapy regimens of paclitaxel and car- IIIA-N2 non-small-cell lung cancer. J Natl Cancer
boplatin for locally advanced non-small-cell lung Inst. 2007;99:442–50.
cancer: a randomized phase II locally advanced multi- 51. Weisenburger TH. Effects of postoperative mediasti-
modality protocol. J Clin Oncol. 2005;23:5883–91. nal radiation on completely resected stage II and stage
40. Cox JD, Azarnia N, Byhardt RW, et al. A randomized III epidermoid cancer of the lung. LCSG 773. Chest.
phase I/II trial of hyperfractionated radiation therapy 1994;106:297S–301S.
with total doses of 60.0 Gy to 79.2 Gy: possible sur- 52. Werner-Wasik M, Paulus R, Curran WJ Jr, et al.
vival benefit with greater than or equal to 69.6 Gy in Acute esophagitis and late lung toxicity in concurrent
Lung Cancer 231
chemoradiotherapy trials in patients with locally tion therapy for limited-stage unresectable thymoma:
advanced non-small-cell lung cancer: analysis of the an intergroup trial. J Clin Oncol. 1997;15:3093–9.
radiation therapy oncology group (RTOG) database. 64. Xie L, Fan M, Sheets NC, et al. The use of radia-
Clin Lung Cancer. 2011;12:245–51. tion therapy appears to improve outcome in patients
53. Stinchcombe TE, Hodgson L, Herndon JE, et al.
with malignant primary tracheal tumors: a SEER-
Treatment outcomes of different prognostic groups based analysis. Int J Radiat Oncol Biol Phys.
of patients on Cancer and Leukemia Group B trial 2012;84:464–70.
39801: induction chemotherapy followed by chemo- 65. Urdaneta AI, Yu JB, Wilson LD. Population based
radiotherapy compared with chemoradiotherapy cancer registry analysis of primary tracheal carci-
alone for unresectable stage III non-small cell lung noma. Am J Clin Oncol. 2011;34:32–7.
cancer. J Thorac Oncol. 2009;4:1117–25. 66. Auperin A, Arriagada R, Pignon JP, et al. Prophylactic
54. Wang L, Wu S, Ou G, et al. Randomized phase II cranial irradiation for patients with small-cell lung
study of concurrent cisplatin/etoposide or paclitaxel/ cancer in complete remission. Prophylactic Cranial
carboplatin and thoracic radiotherapy in patients with Irradiation Overview Collaborative Group. N Engl J
stage III non-small cell lung cancer. Lung Cancer. Med. 1999;341:476–84.
2012;77:89–96. 67. Choi NC, Herndon JE 2nd, Rosenman J, et al. Phase
55. Santana-Davila R, Devisetty K, Szabo A, et al.
I study to determine the maximum-tolerated dose of
Cisplatin and etoposide versus carboplatin and pacli- radiation in standard daily and hyperfractionated-
taxel with concurrent radiotherapy for stage III non- accelerated twice-daily radiation schedules with
small-cell lung cancer: an analysis of Veterans Health concurrent chemotherapy for limited-stage small-cell
Administration data. J Clin Oncol. 2015;33:567–74. lung cancer. J Clin Oncol. 1998;16:3528–36.
56.
Sundstrom S, Bremnes R, Aasebo U, et al. 68. Jeremic B, Shibamoto Y, Nikolic N, et al. Role of radi-
Hypofractionated palliative radiotherapy (17 Gy per ation therapy in the combined-modality treatment of
two fractions) in advanced non-small-cell lung car- patients with extensive disease small-cell lung cancer:
cinoma is comparable to standard fractionation for a randomized study. J Clin Oncol. 1999;17:2092–9.
symptom control and survival: a national phase III 69. Le Pechoux C, Dunant A, Senan S, et al. Standard-
trial. J Clin Oncol. 2004;22:801–10. dose versus higher-dose prophylactic cranial irradia-
57. Auperin A, Le Pechoux C, Rolland E, et al. Meta- tion (PCI) in patients with limited-stage small-cell
analysis of concomitant versus sequential radioche- lung cancer in complete remission after chemotherapy
motherapy in locally advanced non-small-cell lung and thoracic radiotherapy (PCI 99-01, EORTC 22003-
cancer. J Clin Oncol. 2010;28:2181–90. 08004, RTOG 0212, and IFCT 99-01): a randomised
58. Eberhardt WE, Pottgen C, Gauler TC, et al. Phase clinical trial. Lancet Oncol. 2009;10:467–74.
III study of surgery versus definitive concurrent 70. Pignon JP, Arriagada R, Ihde DC, et al. A meta-
chemoradiotherapy boost in patients with resectable analysis of thoracic radiotherapy for small-cell lung
stage IIIA(N2) and selected IIIB non-small-cell lung cancer. N Engl J Med. 1992;327:1618–24.
cancer after induction chemotherapy and concur- 71. Slotman B, Faivre-Finn C, Kramer G, et al.
rent chemoradiotherapy (ESPATUE). J Clin Oncol. Prophylactic cranial irradiation in extensive small-cell
2015;33:4194–201. lung cancer. N Engl J Med. 2007;357:664–72.
59. Suntharalingam M, Paulus R, Edelman MJ, et al.
72. Slotman BJ, van Tinteren H, Praag JO, et al. Use of
Radiation therapy oncology group protocol 02-29: a thoracic radiotherapy for extensive stage small-cell
phase II trial of neoadjuvant therapy with concurrent lung cancer: a phase 3 randomised controlled trial.
chemotherapy and full-dose radiation therapy fol- Lancet. 2015;385:36–42.
lowed by surgical resection and consolidative therapy 73. Takada M, Fukuoka M, Kawahara M, et al. Phase III
for locally advanced non-small cell carcinoma of the study of concurrent versus sequential thoracic radio-
lung. Int J Radiat Oncol Biol Phys. 2012;84:456–63. therapy in combination with cisplatin and etoposide
60.
Gandara DR, Chansky K, Albain KS, et al. for limited-stage small-cell lung cancer: results of the
Consolidation docetaxel after concurrent chemora- Japan Clinical Oncology Group Study 9104. J Clin
diotherapy in stage IIIB non-small-cell lung cancer: Oncol. 2002;20:3054–60.
phase II Southwest Oncology Group Study S9504. J 74. Turrisi AT 3rd, Kim K, Blum R, et al. Twice-daily
Clin Oncol. 2003;21:2004–10. compared with once-daily thoracic radiotherapy
61. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab ver- in limited small-cell lung cancer treated concur-
sus docetaxel in advanced nonsquamous non-small- rently with cisplatin and etoposide. N Engl J Med.
cell lung cancer. N Engl J Med. 2015;373:1627–39. 1999;340:265–71.
62. Yuan S, Sun X, Li M, et al. A randomized study of 75. Schild SE, Bonner JA, Shanahan TG, et al. Long-term
involved-field irradiation versus elective nodal irra- results of a phase III trial comparing once-daily radio-
diation in combination with concurrent chemotherapy therapy with twice-daily radiotherapy in limited-stage
for inoperable stage III nonsmall cell lung cancer. Am small-cell lung cancer. Int J Radiat Oncol Biol Phys.
J Clin Oncol. 2007;30:239–44. 2004;59:943–51.
63. Loehrer PJ Sr, Chen M, Kim K, et al. Cisplatin, doxo- 76. Kies MS, Mira JG, Crowley JJ, et al. Multimodal
rubicin, and cyclophosphamide plus thoracic radia- therapy for limited small-cell lung cancer: a ran-
232 N. G. Zaorsky et al.
domized study of induction combination chemother- trialists. Federation Nationale des Centres de Lutte
apy with or without thoracic radiation in complete Contre le Cancer. Int J Radiat Oncol Biol Phys.
responders; and with wide-field versus reduced-field 1995;32:651–9.
radiation in partial responders: a Southwest Oncology 87. Wright CD. Management of thymomas. Crit Rev
Group Study. J Clin Oncol. 1987;5:592–600. Oncol Hematol. 2008;65:109–20.
77. Liengswangwong V, Bonner JA, Shaw EG, et al.
88. Wright CD, Choi NC, Wain JC, et al. Induction
Limited-stage small-cell lung cancer: patterns of chemoradiotherapy followed by resection for locally
intrathoracic recurrence and the implications for tho- advanced Masaoka stage III and IVA thymic tumors.
racic radiotherapy. J Clin Oncol. 1994;12:496–502. Ann Thorac Surg. 2008;85:385–9.
78. Komaki R, Khalid N, Langer CJ, et al. Penetration 89. Hamaji M, Ali SO, Burt BM. A meta-analysis of
of recommended procedures for lung cancer staging induction therapy for advanced thymic epithelial
and management in the United States over 10 years: tumors. Ann Thorac Surg. 2015;99:1848–56.
a quality research in radiation oncology survey. Int J 90. Flores RM, Krug LM, Rosenzweig KE, et al.
Radiat Oncol Biol Phys. 2013;85:1082–9. Induction chemotherapy, extrapleural pneumonec-
79. Komaki R, Meyers CA, Shin DM, et al. Evaluation of tomy, and postoperative high-dose radiotherapy for
cognitive function in patients with limited small cell locally advanced malignant pleural mesothelioma: a
lung cancer prior to and shortly following prophylac- phase II trial. J Thorac Oncol. 2006;1:289–95.
tic cranial irradiation. Int J Radiat Oncol Biol Phys. 91. Rusch VW, Rosenzweig K, Venkatraman E, et al.
1995;33:179–82. A phase II trial of surgical resection and adjuvant
80. Giuliani ME, Atallah S, Sun A, et al. Clinical out- high-dose hemithoracic radiation for malignant
comes of extensive stage small cell lung carcinoma pleural mesothelioma. J Thorac Cardiovasc Surg.
patients treated with consolidative thoracic radiother- 2001;122:788–95.
apy. Clin Lung Cancer. 2011;12:375–9. 92. Vogelzang NJ, Rusthoven JJ, Symanowski J, et al.
81. Brahmer JR, Ettinger DS. The Role of Topotecan in Phase III study of pemetrexed in combination with
the Treatment of Small Cell Lung Cancer. Oncologist. cisplatin versus cisplatin alone in patients with
1998;3:11–4. malignant pleural mesothelioma. J Clin Oncol.
82. Takahashi T, Yamanaka T, Seto T, et al. Prophylactic 2003;21:2636–44.
cranial irradiation versus observation in patients with 93. Stahel RA, Riesterer O, Xyrafas A, et al. Neoadjuvant
extensive-disease small-cell lung cancer: a multi- chemotherapy and extrapleural pneumonectomy of
centre, randomised, open-label, phase 3 trial. Lancet malignant pleural mesothelioma with or without
Oncol. 2017;18:663–71. hemithoracic radiotherapy (SAKK 17/04): a ran-
83. Forquer JA, Rong N, Fakiris AJ, et al. Postoperative domised, international, multicentre phase 2 trial.
radiotherapy after surgical resection of thymoma: dif- Lancet Oncol. 2015;16:1651–8.
fering roles in localized and regional disease. Int J 94. O'Rourke N, Garcia JC, Paul J, et al. A randomised
Radiat Oncol Biol Phys. 2010;76:440–5. controlled trial of intervention site radiotherapy in
84. Haniuda M, Miyazawa M, Yoshida K, et al. Is post- malignant pleural mesothelioma. Radiother Oncol.
operative radiotherapy for thymoma effective? Ann 2007;84:18–22.
Surg. 1996;224:219–24. 95. Wilson LD, Detterbeck FC, Yahalom J. Clinical prac-
85. Mangi AA, Wright CD, Allan JS, et al. Adjuvant radi- tice. Superior vena cava syndrome with malignant
ation therapy for stage II thymoma. Ann Thorac Surg. causes. N Engl J Med. 2007;356:1862–9.
2002;74:1033–7. 96. Loeffler JS, Leopold KA, Recht A, et al. Emergency
86. Mornex F, Resbeut M, Richaud P, et al. Radiotherapy prebiopsy radiation for mediastinal masses: impact on
and chemotherapy for invasive thymomas: a multicen- subsequent pathologic diagnosis and outcome. J Clin
tric retrospective review of 90 cases. The FNCLCC Oncol. 1986;4:716–21.
Breast Cancer
6
Nicholas G. Zaorsky, Daniel M. Trifiletti,
and Jennifer Rosenberg
Abstract
This chapter discusses the general manage-
ment of patients with breast cancers, with spe-
cial focus on principles that guide radiotherapy
management. Several key components of tri-
modality care and accelerated partial breast
irradiation (APBI) are discussed.
T1 – ≤2 cm T1 T2 T3 T4
T1mi – ≤0.1 cm N0 IA IIA IIB
N1mi = IB IIIB
T1a – >0.1 and ≤0.5 cm N1 IIA IIB IIIA
T1b – >0.5 and ≤1 cm N2 IIIA
T1c – >1.0 and ≤2 cm N3 IIIC
M1 IV
T2 – >2.0 and ≤5 cm
T3 – >5 cm
T4 - AJCC 8th Edition Changes/Staging
T4a –
chest wall (not pec, but serratus, Clarifications
intercostal, ribs) –– LCIS no longer included in Tis, it is benign.
T4b – ulceration, peau d’orange, nodules, and/or –– Multifocal: 2+ tumors in the same quadrant.
edema that is not T4d –– Multicentric: 2+ tumors in diff quadrants,
T4c – 4a + 4b separated by >5 cm.
T4d – inflammatory breast cancer (IBC): –– For multifocal disease, use size of max focus
Rapid onset of breast erythema, edema, (do not sum).
and/or peau d’orange involving >1/3 of –– Anatomic stage: Only based on TNM.
breast with or without palpable mass. –– Prognostic stage: Includes TNM, ER/PR/
<6-month duration. Her2 status, +/− Oncotype testing
Pathological confirmation: core bx con- –– pT1–2 N0 that is ER/PR+ and HER2- and
firms invasive ca, skin punch bx (minimum Oncotype <11 is stage IA.
of 2). DLI pathognomonic but not required. –– Intramammary LN count as axillary nodes for
staging.
cN0 No nodes pN0 No nodes
cN1 Mobile level I-II nodes pN1mi Micromets (0.2-2mm)
pN1a 1-3 nodes (>2mm)
pN1b Micro IM nodes
pN1c pN1a +pN1b
cN2a Fixed/matted axillary nodes pN2a 4-9 nodes (>2 mm)
cN2b Clinical IM nodes only pN2b Clinical IM nodes with
pathologically neg axilla
cN3a Infraclav nodes (ax level III) pN3a 10+ axillary or any infraclav
cN3b IM + axillary nodes pN3b IM + axillary nodes
cN3c Ipsilateral Supraclav pN3c Ipsilateral Supraclav
236 N. G. Zaorsky et al.
Anatomy
lat dorsi
4
1 2 3 4
Breast Cancer 237
MLO CC
R L R L
Pe
c
ma
jor
upper outer
nipple
lower inner
Nicholas G Zaorsky, MD
240 N. G. Zaorsky et al.
Surgery Types
Surgery type Breast removed Muscle removed Nodes removed Predictors of LC Predictors of OS
RM Yes Pec major and minor ALND I, II, III 1. Axl LNs
MRM Yes Pec major fascia ALND I, II 1. # Axl LNs 2. T stage
2. T stage
TM Yes Pec major fascia No
WLE Partial No No
Excisional bx Partial No No
BCS Partial No Typically w/ SLN (I) 1. R+
2. young age
chemo, then RT vs (2) RT, then chemo. Chemo after BCS. For any size tumor with pCR, LRR
was CMF, prednisone, leucovorin. RT was rates were 0%. Highest LRR rates if ypN+.
45 + 16 Gy. Long-term FU: no benefit for –– After MRM, predictors of recurrence were
LRR, time to failure, DMs, OS. (1) tumor size prior to chemo, (2) LN status
prior to chemo, and (3) ypN status.
Recht trial Chemo→RT RT → chemo –– Predictors of recurrence in BCS were (1)
10 y rate of any event 46% 51%, p = 0.88
age, (2) cLN status, and (3) ypN status.
(IBTR, CBTR, breast
cancer, second –– The very low LRR rates are the rationale
malignancy, or death) for NSABP B-51.
10 y distant metastases 35% 36%, p = 0.70 –– NSABP B-28: LN+ pts randomized to AC vs
10 y mortality 28% 33%, p = 0.41 ACT. Taxane improved 5-yr DFS 72 → 76%
(RR 0.83). No OS benefit. MA-20 showed
–– EBCTCG chemo/HT: meta-analysis, 6 m similar DSS benefit to adding RNI to early
anthracycline-based chemo reduced breast ca stage N+ patients (HR 0.76, DFS 82 vs 77%).
death by 38% if <50 yo and 20% if 50–69 yo. –– NCCTG N9831: HER2 pos women rando to
Tamoxifen x5 yrs reduced breast ca death by AC, then T vs AC then T then trastuzumab vs
31%. AC, then T+ trastuzumab, then transtuzumab
–– NSABP B-20: surgery and pN0, ER+. alone.
Randomized to Tamox vs Tamox +MF chemo –– NSABP B-31: HER2+ pts: ACT vs
vs Tamox + CMF. Chemo improved 12 yr ACT+H. Herceptin improved 3-yr DFS
DFS, but not OS (p = 0.068). These pts were (75 → 87%) and OS (92 → 94%).
used in the validation of Oncotype recurrence –– N9831 and B-31 joint analysis (Perez, 2011):
score. AC→T +/− trastuzumab. 10y DFS 75 vs 84%,
–– CALGB 9741: showed that dose dense ACT OS 62 vs 74%.
improved 4 yr DFS 75 → 82% as well as OS. –– ATAC Trial (Howell, 2005; Cuzick, 2010):
–– NSABP B-14: ER+ pts. Tamox 10 mg BID x5 ER+/− postmen pts: anastrozole 1 mg QD,
yrs vs placebo. Tamox improved 15-yr DFS/ tamoxifen 20 mg QD, or both. AI improved
OS. DFS over tamox (89 vs 87%); both were 87%,
–– NSABP B-18: cT1–3 N01. Preop vs postop but only for ER+ pts. No dif in OS.
AC x4c. n = 1523. At 9 yrs, no difference in
DFS or OS. More BCT in preop group (~25%
converted), but more LR in those patients Systemic Therapy, Clinical Use
(10.7 vs 7.6%). More LR if mastectomy con-
verted to BCT (9.6 vs 15.7%). Main benefit of I ndications for Adjuvant Chemo
NA CT was downstaging tumor for BCT. At –– ER+, HER2-:
time of surgery, pts w/ NA CT were ypN = 41% 1. + LN (>2 mm) or
of time vs 57% if no NA CT. 2. Tumor >0.5 cm → Oncotype, ≥31
–– NSABP B-27: Neoadj ACx4→surg vs –– All other subtypes:
ACx4 →docetaxel→surg vs ACx4→surg→ 1. + LN (>2 mm) or
docetaxel. More toxicity with AC then 2. Tumor >1 cm
docetaxel (G4 = 23% vs 10%). However,
response increased (40% vs 64% cCR; 14% vs gents Used and Schedule
A
26% pCR). Similar rates of BCS. –– ddAC x 4 → T x 4, q 2 weeks (dose dense).
–– B-18 + B-27 combined analysis (Mamounas, *Adriamycin 60 mg/m2 and cyclophospha-
2012): n = 3088. NA chemo with AC or mide 600 mg/m2 are both on day 1, cycled q
AC-T. Patients who had MRM did not get 14 days for 4 cycles (2 months total).
adjuvant RT. Patients w/ BCS had WBI. Paclitaxel 175 mg/m2 weekly x 4 weeks.
10-year LR was 12% after MRM and 10% Preferred for LN+.
Breast Cancer 245
–– TC –– Indications:
Docetaxel 75 mg/m2 IV day 1. LN(−) and ER(+) who will be tx with
Cyclophosphamide 600 mg/m2 IV day 1. tamoxifen
Cycled q21d x 4 cycles with filgrastim support SWOG 8814 also demonstrated predictive
for each cycle. magnitude in ER+ LN+ patients
Some use AC, but not “preferred” per NCCN. –– 0–17 low risk. 31+ high risk
–– ER+: Tam (20 mg QD) or anastrozole (1 mg
QD) x 5–10 years. elative Indications for Neoadjuvant
R
–– HER2+: Herceptin (start with first dose of Chemotherapy (NACT)
paclitaxel if w/ CT) x 1 year. 4 mg/kg once per –– To get to BCS (T2, T3)
week. Cardiac monitoring at 3, 6, and 9 months. –– Locally advanced tumors (N2, T3N1, T4, N3)
–– + herceptin if + LN, > 2 cm (Herceptin: –– Inflammatory
4 mg/kg loading then 2 mg/kg weekly during
chemo, then 6 mg/kg q3wks x 1 y after) o Ensure Post-NACT SLNBx FNR < 10%
T
–– FNA w/ clip placement so the primary is not
lost
AI Follow-Up –– Dual radiocolloid and blue dye tracer
–– Check DEXA at baseline –– IHC to define any positivity
–– Toxicities: Osteoporosis, arthralgia, SOB, –– 3+ SLN
cough, fever/chills
Hot Flashes/Antidepressants
–– Venlafaxine, paroxetine, fluoxetine, and ser-
Oncotype Dx traline all decrease hot flashes vs placebo.
–– PCR study of 21 genes (5 reference/16 cancer) Sertraline has been shown to decrease hot
–– Prediction/prognostication: flashes in women on hormonal therapy.
Likelihood of disease recurrence and survival –– Fluoxetine/paroxetine may decrease forma-
(prognostic) tion of tamoxifen metabolites.
Response to chemo (predictive)
246 N. G. Zaorsky et al.
Omission of RT
Consider if age > 65–70, ER/PR+, T1 N0 tumors,
on hormonal therapy (similar to CALGB 9343,
PRIME II inclusion).
256 N. G. Zaorsky et al.
PORT (tangents). 97% got systemic therapy NCCN for T3+, N2+
(HT or chemo) at discretion of physician. –– Neoadj chemo → MRM → PMRT.
5-yr regional control same (99 + %), OS –– OR MRM, then chemo and RT.
same (92%). Radiation field design varied –– If doing NACT, place markers to localize
widely: 89% got WBRT, 15% got RNI tumor bed.
(though it wasn’t allowed), high tangents –– If NACT, and given AC x 4 but no response,
used in 50%. try taxol x 4 (per B-27).
–– Which patients do not quality for Z0011? If –– Contraindications to BCT after NACT:
cannot find the node, if gross ECE, if clini- Multicentric, IBC (T4d), diffuse microcalcs,
cally LN+, R+, >3 LNs involved, matted LNs, residual skin involvement.
neoadjuvant systemic tx.
260 N. G. Zaorsky et al.
Overview Overview
–– Large, fast growing. –– Eczematoid changes, crusting, redness,
–– Arises from periductal stromal cells. discharge
–– Accounts for <1% of breast neoplasms.
–– 30% are malignant. Workup
–– Stomal overgrowth, tumor size >7 cm. –– Full thickness skin biopsy
–– Infiltrative borders, necrosis, and high mitotic –– PE + MMG; if both (−) → MRI for patients
count are the main high-risk features for who are candidates for BCT
DMs.
–– Classification: benign, borderline, and malig-
nant (only malignant get RT). Treatment
–– Paget alone (Tis); Paget + DCIS (Tis); Paget +
Differential invasive disease (staged per invasive)
–– Fibroadenoma, IDC –– BCS (with removal of full nipple areolar com-
plex) + RT vs TM
Treatment –– EORTC 10873, Bijker, 2001: 5-year LR 5%
–– Optimal management is surgery. after complete excision of nipple-areolar com-
–– Re-excise for R+. No axillary staging. plex and underlying breast + WBI 50 Gy
–– Consider PORT if unresectable (cat 2b) or if
very large and removed w/ MRM (e.g., >
10 cm) or if >5 cm and removed w/ BCS.
262 N. G. Zaorsky et al.
References 16. Perez EA, Romond EH, Suman VJ, et al. Four-year
follow-up of trastuzumab plus adjuvant chemotherapy
for operable human epidermal growth factor receptor
1. Ho AL, Bovill ES, Macadam SA, et al. 2-positive breast cancer: joint analysis of data from
Postmastectomy radiation therapy after immediate NCCTG N9831 and NSABP B-31. J Clin Oncol.
two-stage tissue expander/implant breast reconstruc- 2011;29:3366–73.
tion: a University of British Columbia perspective. 17. Cuzick J, Sestak I, Baum M, et al. Effect of anastro-
Plast Reconstr Surg. 2014;134:1e–10e. zole and tamoxifen as adjuvant treatment for early-
2. Borgen PI, Senie RT, McKinnon WM, et al. stage breast cancer: 10-year analysis of the ATAC
Carcinoma of the male breast: analysis of prognosis trial. Lancet Oncol. 2010;11:1135–41.
compared with matched female patients. Ann Surg 18. Martinez-Monge R, Gomez-Iturriaga A, Cambeiro M,
Oncol. 1997;4:385–8. et al. Phase I-II trial of perioperative high-dose-rate
3. Botteri E, Bagnardi V, Rotmensz N, et al. Analysis of brachytherapy in oral cavity and oropharyngeal can-
local and regional recurrences in breast cancer after cer. Brachytherapy. 2009;8:26–33.
conservative surgery. Ann Oncol. 2010;21:723–8. 19. Cook B DT. Wikibooks radiation oncology. https://
4. Amin MB, Edge SB, Byrd DR, et al. American Joint en.wikibooks.org/wiki/Radiation_Oncology. Accessed
Committee on Cancer (AJCC) cancer staging manual. April 2018.
8th ed. New York, NY: Springer; 2017. 20. Edge S, Byrd D, Compton C, et al. AJCC cancer stag-
5. Chagpar AB, Killelea BK, Tsangaris TN, et al. A ran- ing manual. New York, NY: Springer; 2010.
domized, controlled trial of cavity shave margins in 21. Hansen ERM. Handbook of evidence-based radiation
breast cancer. N Engl J Med. 2015;373:503–10. oncology. 2nd ed: Springer; 2010.
6. Finn RS, Martin M, Rugo HS, et al. Palbociclib and 22. Hristov BLS, Christodouleas JP. Radiation oncology -
letrozole in advanced breast cancer. N Engl J Med. a question based review. 2nd ed: LWW; 2014.
2016;375:1925–36. 23. Bijker N, Meijnen P, Peterse JL, et al. Breast-
7. Hortobagyi GN, Stemmer SM, Burris HA, et al. conserving treatment with or without radiotherapy in
Ribociclib as first–line therapy for HR–positive, ductal carcinoma-in-situ: ten-year results of European
advanced breast cancer. N Engl J Med. 2016;375: Organisation for Research and Treatment of Cancer
1738–48. randomized phase III trial 10853--a study by the
8. Cristofanilli M, Turner NC, Bondarenko I, et al. EORTC Breast Cancer Cooperative Group and EORTC
Fulvestrant plus palbociclib versus fulvestrant plus pla- Radiotherapy Group. J Clin Oncol. 2006;24:3381–7.
cebo for treatment of hormone-receptor-positive, HER2- 24. Fisher B, Dignam J, Wolmark N, et al. Tamoxifen
negative metastatic breast cancer that progressed on in treatment of intraductal breast cancer: National
previous endocrine therapy (PALOMA-3): final analy- Surgical Adjuvant Breast and Bowel Project B-24 ran-
sis of the multicentre, double-blind, phase 3 randomised domised controlled trial. Lancet. 1999;353:1993–2000.
controlled trial. Lancet Oncol. 2016;17:425–39. 25. Holmberg L, Garmo H, Granstrand B, et al. Absolute
9. Pagani O, Regan MM, Walley BA, et al. Adjuvant risk reductions for local recurrence after postoperative
exemestane with ovarian suppression in premeno- radiotherapy after sector resection for ductal carcinoma
pausal breast cancer. N Engl J Med. 2014;371:107–18. in situ of the breast. J Clin Oncol. 2008;26:1247–52.
10. Brufsky A, Harker WG, Beck JT, et al. Zoledronic 26. Houghton J, George WD, Cuzick J, et al. Radiotherapy
acid inhibits adjuvant letrozole-induced bone loss in and tamoxifen in women with completely excised
postmenopausal women with early breast cancer. J ductal carcinoma in situ of the breast in the UK,
Clin Oncol. 2007;25:829–36. Australia, and New Zealand: randomised controlled
11. Bundred NJ, Campbell ID, Davidson N, et al. Effective trial. Lancet. 2003;362:95–102.
inhibition of aromatase inhibitor-associated bone 27. Wapnir IL, Dignam JJ, Fisher B, et al. Long-term
loss by zoledronic acid in postmenopausal women outcomes of invasive ipsilateral breast tumor recur-
with early breast cancer receiving adjuvant letrozole: rences after lumpectomy in NSABP B-17 and B-24
ZO-FAST study results. Cancer. 2008;112:1001–10. randomized clinical trials for DCIS. J Natl Cancer
12. Van Poznak C, Hannon RA, Mackey JR, et al. Prevention Inst. 2011;103:478–88.
of aromatase inhibitor-induced bone loss using risedro- 28. Dunne C, Burke JP, Morrow M, et al. Effect of margin
nate: the SABRE trial. J Clin Oncol. 2010;28:967–75. status on local recurrence after breast conservation
13. Margolese RG, Cecchini RS, Julian TB, et al. Anastrozole and radiation therapy for ductal carcinoma in situ. J
versus tamoxifen in postmenopausal women with ductal Clin Oncol. 2009;27:1615–20.
carcinoma in situ undergoing lumpectomy plus radio- 29. Solin LJ, Gray R, Baehner FL, et al. A multigene
therapy (NSABP B-35): a randomised, double-blind, expression assay to predict local recurrence risk for
phase 3 clinical trial. Lancet. 2016;387:849–56. ductal carcinoma in situ of the breast. J Natl Cancer
14. Early Breast Cancer Trialists Collaborative Group
Inst. 2013;105:701–10.
(EBCTCG). Aromatase inhibitors versus tamoxifen 30. Hughes LL, Wang M, Page DL, et al. Local exci-
in early breast cancer: patient-level meta-analysis of sion alone without irradiation for ductal carcinoma in
the randomised trials. Lancet. 2015;386:1341–52. situ of the breast: a trial of the Eastern Cooperative
15. Goss PE, Ingle JN, Pritchard KI, et al. Extending aro- Oncology Group. J Clin Oncol. 2009;27:5319–24.
matase–inhibitor adjuvant therapy to 10 years. N Engl 31. Allred DC, Clark GM, Molina R, et al. Overexpression
J Med. 2016;375:209–19. of HER-2/neu and its relationship with other prog-
Breast Cancer 263
nostic factors change during the progression of in tectomy, total mastectomy, and total mastectomy fol-
situ to invasive breast cancer. Hum Pathol. 1992; lowed by irradiation. N Engl J Med. 2002;347:567–75.
23:974–9. 46. Haviland JS, Owen JR, Dewar JA, et al. The UK
32. McCormick B, Winter K, Hudis C, et al. RTOG 9804: standardisation of breast radiotherapy (START) tri-
a prospective randomized trial for good-risk ductal als of radiotherapy hypofractionation for treatment
carcinoma in situ comparing radiotherapy with obser- of early breast cancer: 10-year follow-up results
vation. J Clin Oncol. 2015;33:709–15. of two randomised controlled trials. Lancet Oncol.
33. Wong JS, Kaelin CM, Troyan SL, et al. Prospective 2013;14:1086–94.
study of wide excision alone for ductal carcinoma in 47. Hughes KS, Schnaper LA, Berry D, et al. Lumpectomy
situ of the breast. J Clin Oncol. 2006;24:1031–6. plus tamoxifen with or without irradiation in women
34. Fisher ER, Sass R, Fisher B, et al. Pathologic findings 70 years of age or older with early breast cancer. N
from the National Surgical Adjuvant Breast Project Engl J Med. 2004;351:971–7.
(protocol 6). I. Intraductal carcinoma (DCIS). Cancer. 48. Krag DN, Anderson SJ, Julian TB, et al. Technical
1986;57:197–208. outcomes of sentinel-lymph-node resection and con-
35. Fisher ER, Leeming R, Anderson S, et al. Conservative ventional axillary-lymph-node dissection in patients
management of intraductal carcinoma (DCIS) of the with clinically node-negative breast cancer: results
breast. Collaborating NSABP investigators. J Surg from the NSABP B-32 randomised phase III trial.
Oncol. 1991;47:139–47. Lancet Oncol. 2007;8:881–8.
36. Fisher B, Costantino JP, Wickerham DL, et al.
49. Romestaing P, Lehingue Y, Carrie C, et al. Role of
Tamoxifen for prevention of breast cancer: report of the a 10-Gy boost in the conservative treatment of early
National Surgical Adjuvant Breast and Bowel Project breast cancer: results of a randomized clinical trial in
P-1 study. J Natl Cancer Inst. 1998;90:1371–88. Lyon, France. J Clin Oncol. 1997;15:963–8.
37. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen 50. van Dongen JA, Voogd AC, Fentiman IS, et al. Long-
for the prevention of breast cancer: current status of the term results of a randomized trial comparing breast-
National Surgical Adjuvant Breast and Bowel Project conserving therapy with mastectomy: European
P-1 study. J Natl Cancer Inst. 2005;97:1652–62. Organization for research and treatment of cancer
38. Bartelink H, Horiot JC, Poortmans PM, et al. Impact 10801 trial. J Natl Cancer Inst. 2000;92:1143–50.
of a higher radiation dose on local control and sur- 51. Vernon CC, Hand JW, Field SB, et al. Radiotherapy
vival in breast-conserving therapy of early breast can- with or without hyperthermia in the treatment of super-
cer: 10-year results of the randomized boost versus ficial localized breast cancer: results from five ran-
no boost EORTC 22881-10882 trial. J Clin Oncol. domized controlled trials. International Collaborative
2007;25:3259–65. Hyperthermia Group. Int J Radiat Oncol Biol Phys.
39. Bellon JR, Come SE, Gelman RS, et al. Sequencing 1996;35:731–44.
of chemotherapy and radiation therapy in early-stage 52. Veronesi U, Cascinelli N, Mariani L, et al. Twenty-year
breast cancer: updated results of a prospective ran- follow-up of a randomized study comparing breast-
domized trial. J Clin Oncol. 2005;23:1934–40. conserving surgery with radical mastectomy for early
40. Bentzen SM, Agrawal RK, Aird EG, et al. The UK breast cancer. N Engl J Med. 2002;347:1227–32.
standardisation of breast radiotherapy (START) trial 53. Whelan TJ, Pignol JP, Levine MN, et al. Long-term
A of radiotherapy hypofractionation for treatment of results of hypofractionated radiation therapy for
early breast cancer: a randomised trial. Lancet Oncol. breast cancer. N Engl J Med. 2010;362:513–20.
2008;9:331–41. 54. Giuliano AE, Hunt KK, Ballman KV, et al. Axillary
41. Buchholz TA, Lehman CD, Harris JR, et al. Statement dissection vs no axillary dissection in women with
of the science concerning locoregional treatments invasive breast cancer and sentinel node metastasis: a
after preoperative chemotherapy for breast cancer: a randomized clinical trial. JAMA. 2011;305:569–75.
National Cancer Institute conference. J Clin Oncol. 55. Galimberti V, Cole BF, Zurrida S, et al. Axillary
2008;26:791–7. dissection versus no axillary dissection in patients
42. Cascinelli N, Greco M, Bufalino R, et al. Prognosis with sentinel-node micrometastases (IBCSG
of breast cancer with axillary node metastases after 23-01): a phase 3 randomised controlled trial.
surgical treatment only. Eur J Cancer Clin Oncol. Lancet Oncol. 2013;14:297–305.
1987;23:795–9. 56. Donker M, van Tienhoven G, Straver ME, et al.
43. Fisher B, Anderson S, Bryant J, et al. Twenty-year Radiotherapy or surgery of the axilla after a positive
follow-up of a randomized trial comparing total mas- sentinel node in breast cancer (EORTC 10981-22023
tectomy, lumpectomy, and lumpectomy plus irradia- AMAROS): a randomised, multicentre, open-label, phase
tion for the treatment of invasive breast cancer. N Engl 3 non-inferiority trial. Lancet Oncol. 2014;15:1303–10.
J Med. 2002;347:1233–41. 57. Bijker N, Rutgers EJ, Duchateau L, et al. Breast-
44. Fisher B, Bryant J, Dignam JJ, et al. Tamoxifen,
conserving therapy for Paget disease of the nipple:
radiation therapy, or both for prevention of ipsilateral a prospective European Organization for Research
breast tumor recurrence after lumpectomy in women and Treatment of cancer study of 61 patients. Cancer.
with invasive breast cancers of one centimeter or less. 2001;91:472–7.
J Clin Oncol. 2002;20:4141–9. 58. van der Ploeg IM, Nieweg OE, van Rijk MC, et al.
45. Fisher B, Jeong JH, Anderson S, et al. Twenty-five-year Axillary recurrence after a tumour-negative sentinel
follow-up of a randomized trial comparing radical mas- node biopsy in breast cancer patients: a systematic
264 N. G. Zaorsky et al.
review and meta-analysis of the literature. Eur J Surg chemotherapy for node-positive breast cancer: results
Oncol. 2008;34:1277–84. from NSABP B-28. J Clin Oncol. 2005;23:3686–96.
59. Clarke M, Collins R, Darby S, et al. Effects of radio- 71. McGale P, Taylor C, Correa C, et al. Effect of radiother-
therapy and of differences in the extent of surgery for apy after mastectomy and axillary surgery on 10-year
early breast cancer on local recurrence and 15-year recurrence and 20-year breast cancer mortality: meta-
survival: an overview of the randomised trials. Lancet. analysis of individual patient data for 8135 women in
2005;366:2087–106. 22 randomised trials. Lancet. 2014;383:2127–35.
60. Strnad V, Ott OJ. Hildebrandt G, et al: 5-year results 72. Olson JA Jr, McCall LM, Beitsch P, et al. Impact of
of accelerated partial breast irradiation using sole immediate versus delayed axillary node dissection on
interstitial multicatheter brachytherapy versus whole- surgical outcomes in breast cancer patients with posi-
breast irradiation with boost after breast-conserving tive sentinel nodes: results from American College of
surgery for low-risk invasive and in-situ carcinoma Surgeons Oncology Group trials Z0010 and Z0011. J
of the female breast: a randomised, phase 3, non- Clin Oncol. 2008;26:3530–5.
inferiority trial. Lancet. 2016;387:229–38. 73. Overgaard M, Hansen PS, Overgaard J, et al.
61. Livi L, Meattini I, Marrazzo L, et al. Accelerated Postoperative radiotherapy in high-risk premeno-
partial breast irradiation using intensity-modulated pausal women with breast cancer who receive adjuvant
radiotherapy versus whole breast irradiation: 5-year chemotherapy. Danish Breast Cancer Cooperative
survival analysis of a phase 3 randomised controlled Group 82b trial. N Engl J Med. 1997;337:949–55.
trial. Eur J Cancer. 2015;51:451–63. 74. Overgaard M, Jensen MB, Overgaard J, et al.
62. Formenti SC, Hsu H, Fenton-Kerimian M, et al. Prone Postoperative radiotherapy in high-risk postmeno-
accelerated partial breast irradiation after breast- pausal breast-cancer patients given adjuvant tamoxi-
conserving surgery: five-year results of 100 patients. fen: Danish Breast Cancer Cooperative Group DBCG
Int J Radiat Oncol Biol Phys. 2012;84:606–11. 82c randomised trial. Lancet. 1999;353:1641–8.
63. Correa C, Harris EE, Leonardi MC, et al. Accelerated 75.
Romond EH, Perez EA, Bryant J, et al.
partial breast irradiation: executive summary for Trastuzumab plus adjuvant chemotherapy for oper-
the update of an ASTRO evidence-based consen- able HER2-positive breast cancer. N Engl J Med.
sus statement. Pract Radiat Oncol. 2017;7(2):73–9. 2005;353:1673–84.
https://doi.org/10.1016/j.prro.2016.09.007. 76. Truong PT, Olivotto IA, Kader HA, et al. Selecting
64. Katz A, Buchholz TA, Thames H, et al. Recursive breast cancer patients with T1-T2 tumors and one to
partitioning analysis of locoregional recurrence pat- three positive axillary nodes at high postmastectomy
terns following mastectomy: implications for adjuvant locoregional recurrence risk for adjuvant radiother-
irradiation. Int J Radiat Oncol Biol Phys. 2001;50: apy. Int J Radiat Oncol Biol Phys. 2005;61:1337–47.
397–403. 77. Wolmark N, Wang J, Mamounas E, et al. Preoperative
65. Taghian A, Jeong JH, Mamounas E, et al. Patterns of chemotherapy in patients with operable breast cancer:
locoregional failure in patients with operable breast nine-year results from National Surgical Adjuvant
cancer treated by mastectomy and adjuvant chemo- Breast and Bowel Project B-18. J Natl Cancer Inst
therapy with or without tamoxifen and without radio- Monogr. 2001:96–102.
therapy: results from five National Surgical Adjuvant 78. Recht A, Come SE, Henderson IC, et al. The sequenc-
Breast and Bowel Project randomized clinical trials. J ing of chemotherapy and radiation therapy after con-
Clin Oncol. 2004;22:4247–54. servative surgery for early-stage breast cancer. N Engl
66.
Citron ML, Berry DA, Cirrincione C, et al. J Med. 1996;334:1356–61.
Randomized trial of dose-dense versus conventionally 79. Cristofanilli M, Gonzalez-Angulo AM, Buzdar AU,
scheduled and sequential versus concurrent combina- et al. Paclitaxel improves the prognosis in estrogen
tion chemotherapy as postoperative adjuvant treatment receptor negative inflammatory breast cancer: the M.
of node-positive primary breast cancer: first report of D. Anderson cancer center experience. Clin Breast
Intergroup Trial C9741/Cancer and Leukemia Group Cancer. 2004;4:415–9.
B trial 9741. J Clin Oncol. 2003;21:1431–9. 80. Liao Z, Strom EA, Buzdar AU, et al. Locoregional
67. Fisher B, Dignam J, Bryant J, et al. Five versus more irradiation for inflammatory breast cancer: effective-
than five years of tamoxifen for lymph node-negative ness of dose escalation in decreasing recurrence. Int J
breast cancer: updated findings from the National Radiat Oncol Biol Phys. 2000;47:1191–200.
Surgical Adjuvant Breast and Bowel Project B-14 81. Bristol IJ, Woodward WA, Strom EA, et al.
randomized trial. J Natl Cancer Inst. 2001;93:684–90. Locoregional treatment outcomes after multimodal-
68. Fisher B, Dignam J, Wolmark N, et al. Tamoxifen ity management of inflammatory breast cancer. Int J
and chemotherapy for lymph node-negative, estrogen Radiat Oncol Biol Phys. 2008;72:474–84.
receptor-positive breast cancer. J Natl Cancer Inst. 82. Haagensen CD, Stout AP. Carcinoma of the breast:
1997;89:1673–82. II. Criteria of operability. Ann Surg. 1943;118:859–70.
69. Howell A, Cuzick J, Baum M, et al. Results of the 83. Pezner RD, Schultheiss TE, Paz IB. Malignant phyl-
ATAC (Arimidex, Tamoxifen, alone or in combina- lodes tumor of the breast: local control rates with
tion) trial after completion of 5 years' adjuvant treat- surgery alone. Int J Radiat Oncol Biol Phys. 2008;
ment for breast cancer. Lancet. 2005;365:60–2. 71:710–3.
70. Mamounas EP, Bryant J, Lembersky B, et al. Paclitaxel 84. National Comprehensive Cancer Network Clinical
after doxorubicin plus cyclophosphamide as adjuvant Practice Guidelines. Breast cancer v 2.2016.
Gastrointestinal Cancers
7
Daniel M. Trifiletti, Leila Tchelebi,
Nicholas G. Zaorsky, and Einsley Marie Janowski
Abstract
This chapter discusses the general manage-
ment of patients with gastrointestinal cancers,
with special focus on principles that guide
radiotherapy management. Several key com-
ponents of trimodality care and stereotactic
body radiation therapy (SBRT) are discussed.
D. M. Trifiletti (*)
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
L. Tchelebi · N. G. Zaorsky
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
E. M. Janowski
University of Virginia, Charlottesville, VA, USA
e-mail: ej8t@hscmail.mcc.virginia.edu
Esophageal Cancer [1–23] Siewert (assess for all adenoCA of GEJ; from Siewert,
1998)
• T1 Epicenter Treat
like
–– T1a – into lamina propria or muscular 1 Lower esophagus within Above Esoph
mucosae 1–5 cm above GEJ GEJ
–– T1b – into submucosa 2 Cardia within 1 cm above and At GEJ Esoph
• T2 – muscularis propria 2 cm below GEJ
• T3 – adventitia 3 Subcardinal with center 2–5 Below Gastric
below GEJ, infiltrates GEJ GEJ
• T4 – adj structures from below
–– T4a – pleura, pericardium, azygos, dia- (Tx 1–2 like esophageal; 3 like gastric)
phragm, peritoneum
–– T4b – aorta, vertebrae, trachea AJCC esophagus cm from
• N1 – 1–2 nodes section incisors Landmark
• N2 – 3–6 nodes 15 Cricoid
Cervical 15–20
• N3 – 7+ nodes
20 Thoracic inlet,
sternal notch
Additions to AJCC 8th, from 7th Upper T 20–25
25 Azygous, carina
–– Changed definition of location of tumor from Mid T 25–30
top to its epicenter. 30 Inf pulmonary vein
–– Any tumor extending <2 cm into stomach is Lower T 30–40
40–42 GEJ
esophageal; if >2 cm, then it is gastric.
–– Added ypTNM prognostic staging, which
does not distinguish between adenoCA vs AJCC 8 T1a T1b T2 T3 T4a T4b
N0 IA-SCC IB-SCC IB-SCC IIA-SCC IIIB IVA
squam. G1 G1-3 G1 G1up/mid
IB-SCC IB-aden IIA-SCC or G1-3
low
General Anatomy/Staging G2/3 G1-2 G2/3
IA-aden IC-aden IIB-SCC
G1 G1/2 G2/3
–– Tumors arising at the GEJ or in the cardia of IB-aden up/mid
the stomach within 5 cm of GEJ and extend G2
IC-aden IIA-aden IIB-aden-
into the GEJ are staged as esophageal. G3 G3 G1-3
–– All others with an epicenter in the stomach N1 IIB IIIA IIIB IIIB
>5 cm from the GEJ or those <5 cm without N2 IIIA IIIB IIIB IVA
N3 IVA
extension into the GEJ are staged as gastric. M1 IVB
–– The staging system differs for N0 patients; it
depends on histology.
–– Once a pt. is LN+, staging and prognosis are AJCC 7 T1 T2 T3 T4a T4b
similar. N0 Depends on histo, G; loc IIIA IIIC
for SC
–– Location matters for SCC; prognosis is worse
N1 IIB IIB IIIA IIIC IIIC
with more proximal tumors.
N2 IIIA IIIA IIIB IIIC IIIC
N3 IIIC
M1 IV
Gastrointestinal Cancers 267
AJCC 8 ypT0 ypT1 ypT2 ypT3 ypT4a –– Thus, CTV margin <30 mm OK in 94% of
ypTNM cases, except distal GEJ adenoCA, which
ypN0 I I I II IIIB need 50 mm.
ypN1 IIIA IIIA IIIA IIIA IVA
ypN2 IIIB IIIB IIIB IIIB IVA Surgical Techniques
ypN3 IVA
–– Endoscopic mucosal resection
M1 IVB
–– No ulceration, no LVSI, <2 cm, G1–2,
T1aN0 (SCC), T1a/superficT1b (adenoCA)
Overview –– 98% OS (Ell 2007)
–– 17, 300 cases/yr., 15, 800 deaths/yr. –– Transhiatal esophagectomy: no thoracotomy,
–– Barrett esophagus annual conversion 0.2– two incisions, pull up Inability to perform a
2%/yr. full thoracic LN dissection, but better toler-
–– Risk factors: tobacco, ETOH, nitrosamines, ated, no mediastinitis risk
Plummer-Vinson syndrome, achalasia, –– Ivor-Lewis (right thoracotomy): good expo-
GERD. sure, risk mediastinitis
–– Note: no serosal lining of esophagus and rich –– Left thoracotomy: good for lower third
submucosal lymphatic network; thus, locore- resections
gional spread common. 90% of tx failures after –– Optimum # nodes = 23 (Peyre 2008); min 15
definitive CRT are within GTV (Welsh, 2012). (NCCN)
–– Surgery alone ~20% 3-year OS
T Tis T1a T1b T2 T3
Risk LN+ <3% 7 20 40 >40
Preop/Periop Chemo
–– RTOG 8911 (Kelsen 1998): T1–2Nx → sur-
Workup
gery +/− preop cis/5-FU. n = 440. Same OS,
–– EGD, EUS with FNA.
2.5% pCR.
–– On US, layers 1, 3, and 5 are bright, whereas 2
–– MAGIC trial (Cunningham 2006): T1–3 N0–1
and 4 are dark. Layer 3, submucosa (T1); layer
gastric/GE/lower esophagus adenosCAs→
4, muscularis propria (T2); layer 5, serosa
surgery +/− periop epirubicin, cisplatin, 5-FU.
(T4).
n = 503. Only 42% of patients assigned to
–– PET-CT or CT chest (40% have lung mets).
periop chemo arm are able to complete all
–– PFTs if planning surgery.
cycles. Only 49% of pts who got all preop
–– Barium swallow as needed.
chemo completed all three cycles of adjuvant
–– Nutrition eval.
chemo. Chemo ↑ 5-yr OS (23 → 36%).
–– Port/J-tube; consider lap (in GEJ adenoCA);
–– FLOT4 (abstract only): MAGIC regimen (3c
Her-2 in M1; CBC; CMP; LFT; EtOH, tobacco
ECF preop and post-op) vs FLOT (4c
abuse.
docetaxel, oxaliplatin, 5-FU preop and post-
–– Bronch if tumor is above carina (at ~25 cm)
op). FLOT improved MST (35 m vs 50 m) and
and M0 to rule out a tracheoesophageal
3-yr OS (48% vs 57%).
fistula.
–– Staging lap and thoracoscopy in LA dz. to
limit futile CMT.
–– Assess Siewert.
–– Pathological microscopic spread (Gao,
IJROBP):
–– AdenoCA: 10 ± 7 mm prox, 18 ± 16 mm
distal.
–– SCC: 10 ± 14 mm prox, 11 ± 8 distal.
268 D. M. Trifiletti et al.
NCCN Technique
1. Definitive surgery –– Generally sim supine, arms up, IV and PO
–– Tis-T1a: EMR vs esophagectomy. Risk of contrast, motion assessment/management for
LNMs <3% lower tumors.
–– T1bN0: esophagectomy (EMR for superfi- –– Cervical eso: supraclav, neck nodes
cial adenosCAs) –– Upper thoracic: include SCV and mediasti-
–– Adjuvant treatment: nal nodes
–– AdenosCAs: surveillance or CRT for pT3– –– Mid thoracic: paraesophageal nodes
T4, bad T2 (high-grade, LVI, PNI, or –– Low thoracic: consider gastric nodes,
age <50 yo), LN+, or R1 or R2 resection celiac axis
–– Squams: CRT only for + margins –– No benefit to dose escalation (INT 0123,
–– Nonsurgical candidate: Minsky).
–– 45–50 Gy + 2 drug chemo. –– PET SUV of 2.5 at baseline (not post chemo)
–– Higher doses may be considered in cervical. is good threshold.
2. Neoadjuvant CRT –– Assimilate EGD and EUS report.
–– T2–T4a or N+. –– IMRT:
–– ChemoRT → surgery (non-cervical) by week –– CTV = 4 cm sup/inf mucosal expansion
6. (only 2 cm into stomach if distal) and
–– RT: 41.4–50.4 Gy. 1–1.5 cm radial expansion for primary
Chemo: weekly carbo (2AUC)/taxol(50) x 5 tumor +1–1.5 cm expansion for gross nodal
wks; CP 75–100 on d1 and d29 + 5-FU 750–1000 disease. Incorporate CT, US, PET. CTV
CI d1–4&d29–32. should be shaved off the bones, aorta, and
3. Definitive CRT (cervical esoph; if decline
heart (limit 0.5 cm into the heart but can
surg; not candidate for surgery). Consider sal- shave off if motion management is used).
vage esophagostomy for persistent disease. –– Upper 1/3: + SCV.
RT: 50–50.4 Gy. –– Distal 1/3: + celiac LNs to the bifurcation.
Chemo: CP 75–100 d1 + 5-FU 750–1000 –– PTV = 0.5 expansion w daily kV imaging.
CI d1–4. Q28d for 2–4 cycles. Another option
is FOLFOX. (Note that the chemo is different Constraints
from the recommended preop carbo/taxol, but –– Lung: V20 <20%; V10 <40%; V5 <50%;
weekly carbo/taxol can be considered). mean ALARA, <5–8 Gy (Wang, 2013)
4. Esophagectomy (non-cervical) – if low risk –– Heart: V50 <33%; V30 <30%; V25 <50%;
(T1 N0, <2 cm, well-differentiated). mean <30 Gy
5. Defin CRT –– Liver: mean <25 Gy; V30 <20%; V20 <30%
–– If cervical or decline surg (50–50.4) –– Kidney: 2/3 <18 Gy; combined mean <18 Gy
–– Then PET/EGD. Can observe if cCR and
SCC
–– T4b: defin CRT (50–50.4)
–– Dysphagia palliation: RT preferred over
stent or CRT
270 D. M. Trifiletti et al.
Gastrointestinal Cancers 271
Gastric Cancer [1–5, 9, 24–34] –– GEJ, fundus: 35%, diffuse. Seiwert III
esophageal CA treated like gastric
• T1 –– Body: 25%
• T1a – into lamina propria or muscular –– Antrum: 40%, intestinal
mucosae
• T1b – into submucosa Workup:
• T2 – muscularis propria (this is the “fourth –– H&P, nodal eval, CBC, CMP, H pylori eval,
layer” on EUS). EGD, EUS, bx, CT CAP w/ IV + oral
• T3 – subserosa contrast
• T4 –– Nutritional assessment and counseling.
• T4a – serosa Consider J-tube (not G)
• T4b – adj structures –– HER2 and MSI/MMR testing if M+ adenocar-
• N1 – 1–2 nodes cinoma is documented/suspected
• N2 – 3–6 nodes –– Assess Siewert category
• N3 – 7+ nodes –– Smoking cessation
• N3a – 7–15 nodes –– Screen for FH
• N3b – 16+ nodes –– PET for T2+, suspicious LN
–– Laparoscopy if T1b + or N+: 20–35% have
M1: metastatic occult peritoneal mets
Mnemonic: IA (T# + N#; i.e., T1 N0) adds to
1; IB adds to 2 (i.e., T2 N0 or T1 N1); IIA adds to Surgical Anatomy
3; IIB adds to 4 –– What is unresectable? Infiltrating mesenteric
root, PA-LNs+, invades major vasculature
(not splenic vessels), DMs, peritoneal
AJCC8 T1 T2 T3 T4a T4b seeding.
N0 IA IB IIA IIB IIIA –– Need subtotal gastrectomy for distal lesion.
N1 IB IIA IIB IIIB –– Aim for >3–5 cm margins, ≥15 LNs.
IIIA
N2 IIA IIB IIIA IIIB –– D0: no or incomplete perigastric nodal
N3a IIB IIIA IIIB IIIB IIIC dissection.
N3b IIIB IIIB IIIC IIIC IIIC –– D1: perigastric nodes.
M1 IV –– D2: D1+ left gastric, common hepatic, splenic,
celiac (“Gotta Harvest Supplementary Cancer”).
–– D3: D2 + hepatoduodenal, retropancreatic,
Overview mesenteric root, PA, middle colic.
–– 22,700 cases/yr., 12,000 deaths/yr. –– Extent of dissection controversial. More com-
–– Risk factors: salt, nitrates, H pylori, perni- plications with D2+ but seemingly improved
cious anemia. CSS.
–– ≈ 20% HER2 positive; MSI/MMR and EBV –– Supplement fat-soluble vitamins (ADEK) and
are other relevant biomarkers B12 post-op.
–– CDH1 mutation: AD truncating mutation,
gene encodes cell adhesion molecule LN Anatomy
E-cadherin. Found in ≈ 40% of hereditary dif- –– Regional LNs of gastric cancer:
fuse gastric cancer (HDGC). For pts w syn- –– Greater curvature: greater curvature,
drome, avg. age of gastric cancer 37 yo, >70% greater omental, gastroduodenal, gastroep-
develop it by age 80. On path, appears diffuse, iploic, pyloric, pancreaticoduodenal
w signet rings. –– Pancreatic and splenic: pancreatolienal,
–– Anatomy peripancreatic, splenic
272 D. M. Trifiletti et al.
–– Lesser curvature: lesser curvature, lesser docetaxel, oxaliplatin, 5-FU preop and post-
omental, L gastric, cardioesophageal, com- op). FLOT improved MST (35 m vs 50 m) and
mon hepatic, celiac, hepatoduodenal 3-yr OS (48% vs 57%).
–– LNs/spread that is metastatic: retropancreatic, –– RTOG 9904 (Ajana 2006): phase II: preop
PA, portal, retroperitoneal, mesenteric, posi- CRT (induction chemo followed by 45 Gy
tive peritoneal cytology with 5-FU + taxol). pCR 26%.
–– Critics (ASCO 2016 abstract): phase II trial of
Surgery stage IB-IVM0 resectable gastric or GE junc-
–– Note, no RCT reveals OS benefit of D2 versus tion cancers rand to (1) periop ECX/EOX vs
D1 resection, but there is a CSS (Dutch trial). (2) ECX/EOX X 3 → surgery → CRT
–– Gouzi 1989: subtotal vs total gastrectomy for (45 Gy + XP). No difference in OS (41%) or
gastric antrum cancers: no difference in OS. PFS with higher heme tox for arm 1 and higher
–– MRC trial (Cuschieri 1996,1999): D1 vs D2. GI tox for arm 2. 87% of patients had at least
Same 5-yr OS, SS higher M/M with D2 Dutch a D1+ dissection with median 20 LNs.
trial (Bonenkamp 1999,Songun 2010): D1 vs –– TOPGEAR. Phase III perioperative ECF
D2. D2 had more complications/deaths. Same chemo versus perioperative chemo + preop
5-yr OS but the 15 yr. data showed D2 ↑CSS CRT – awaiting final results.
(37 → 48%) and ↓ locoregional recurrence.
–– For Dutch and MRC, criticism is 10% op mor- Post-op CRT and/or Chemo
tality, perhaps from inexperience of surgeons. –– INT 0116 (Macdonald 2001,2012): 556 pts,
–– Yu, 2006. RCT of splenectomy vs splenic R0 surgery +/− CRT. Stage IB-IVM0. 54%
preservation in proximal gastric cancer getting D0 resection. Chemo was 5-FU, leucovorin
gastrectomy. No OS benefit. NS higher M/M given once before RT started, then x 5d per w
w splenectomy. No impact on OS in pts w #1 and #5 of RT. RT was 45/25. CRT improved
mets to LNs at hilum of spleen or along splenic DFS and OS. 3-year OS 50% vs 41%; 3-year
artery. RFS 48% vs 31%.
–– Taiwanese trial (Wu 2006): D1 vs D3. D3 ↑ –– Kim 2005: retrospective. 990 pts with D2
5-yr OS (54 → 60%). No adj chemo or RT. +/-CRT. 5-yr OS improved with CRT
–– JCOG9501 trial (Sasako 2008): D2 vs (51 → 57%).
D2 + PAs. Longer surgery; no OS or RFS –– ARTIST: Adjuvant Chemoradiotherapy in
benefit. Stomach Tumors (Lee, 2012; Park, 2015):
2004–2008. n = 458. South Korea. Gastric ade-
Periop/Preop Chemo/CRT noCA s/p R0 gastrectomy + D2 LND. Rand:
–– Theoretical disadvantage is tumor regression (1) chemo alone (“XP”), 6c cape 2000 mg/mg
and perforation, targeting accuracy, and gas- on d1, 14 and cis 60 mg/mg2 on d1, repeat q3w.
tric ulceration. vs (2) 2c of cape and cis +, then 45 Gy + con-
–– MAGIC (Cunningham 2006): mostly gastric current cape 825 mg/m2 BID, and then 2c of
but some GEJ/eso → surgery +/− pre-/post-op cape and cis. RT decreased LRR (13% vs 7%),
chemo (ECF). 42% completed all 6c chemo. but no change in 5-year OS (~74%), DFS
Only 49% of pts completing preop chemo (~75%), DMs (~26%). No benefit in DFS for
completed all 3c adjuvant chemo. CR rate 0%. entire group (p = 0.08) but CRT beneficial if
Chemo improved downstaging, R0, 5 yr OS LN+ (p = 0.04), and in pts w intestinal-type GC
(23 → 36%). Bc of difficulty completing (3 yr. 94% vs 83%). Criticisms: no surg
chemo, benefit of post-op chemo component QA. Borderline significance.
unclear. –– CLASSIC (Noh 2014): 37 centers in SK,
–– FLOT4 (abstract only): MAGIC regimen (3c China, Taiwan. D2 resected II-IIIB gastric
ECF preop and post-op) vs FLOT (4c CA. n = 1035. Surgically resected then rand:
Gastrointestinal Cancers 273
(1) adjuvant oral cape 1000 mg/m2 BID on Post-surgical R1–R2 ± Preop Chemo
d1,14 x 8c + oxali 130 mg/m2 on d1 for 6 m; Treatment
vs (2) obs only. 3-year DFS 75% vs 60%. –– R1: CRT (chemo alone if received preop can
5-year OS 78% vs 69%. G3–4 tox in 56% vs be considered)
6%, favoring surg only. Chemo arm won for –– R2: CRT or palliative
OS, but >9x the severe toxicity.
–– CALGB 80101 (Fuchs 2017): Stage IB-IVM0
patients treated with surgery→ chemo→CRT Technique
(5-FU + 45 Gy) → chemo sandwich, testing –– Fast for 3 hrs before sim.
either ECF versus 5-FU/leucovorin. 3-yr data –– Supine, arms up, IV contrast, oral contrast, 4D.
show similar outcomes. –– Fuse w preop scans.
–– ARTIST II: ongoing. Looking at adjuvant
chemo versus CRT in LN+ patients; chemo is “Classic” Four-Field Technique (from
S1 vs S1 and oxaliplatin. Gunderson Patterns of Failure Paper)
–– 3D CRT w/ four fields (heavy weighting of
Metastatic APPA with RAO/LPO)
–– ToGA trial (Bang 2010): metastatic gastric, –– Superior = Level of L hemidiaphragm
cis/5-FU +/− trastuzumab. Improved median (unless anastomosis above diaphragm), often
OS (11.1 → 13.8 m). T9/10.
–– For proximal T3 lesions, cover medial 2/3 L
NCCN hemidiaphragm.
–– Inferior = L3/L4 for most pts. If upper 1/3
Pre-surgical Recommendations
lesion w/ only 1–2 nodes (+), consider raising
–– Tis-1aN0: endoscopic resection or gastrec-
border to L2 b/c lower risk of sub-pyloric
tomy alone
nodes.
–– T1b: surgery alone
–– Right lateral = 3 cm to right of vertebral
–– T2+ or N+
body. Look at preop/post-op imaging to
1. Perioperative chemotherapy + surg [5-FU
include porta hepatis, gastroduodenal nodes,
and cis x3 →surgery →5-FU and cis] (cat 1)
antrum, medial duodenal wall.
2.
Preoperative CRT (carbo/taxol;
–– Left lateral = Variable. 2/3 of L hemidia-
CP/5-FU) + surgery (cat 2B)
phragm +1 cm. Shield L kidney in lower por-
–– Unresectable: 5-FU-based CRT
tion of field if possible.
–– 6 MV beam energy.
Post-surgical R0 with No Preop Treatment
–– Block edge margin = 5 mm.
–– Tis or T1: surveillance.
–– T2 N0 – surveillance or MacDonald or chemo
–– Volume based:
(if D2 dissection); CRT can be considered for
–– CTVp: residual dz. + remaining stomach +
pT2 N0 w unfav features (LVSI, <D2 LN
anastomosis, 3–5 cm margin. No need to treat
dissection).
entire gastric volume.
–– T3+/N+: MacDonald [MacDonald: surgery +
–– Consider boost for close SM (<5 mm).
post-op chemoRT: 5-FU (425 mg/m2) + leucovo-
–– CTVn (contouring atlas available Wo et al.
rin (20 mg/m2) X 1 cycle → concomitant chemo/
2013).
RT (45 Gy w/ 2 cycles 5-FU(400 mg/m2)/
–– IGRT Mandatory!
LV(20 mg/m2) → 2 additional cycles 5-FU(425 mg/
m2)/LV (5 cycles of chemo, RT w/cycles 2 + 3)].
274 D. M. Trifiletti et al.
Predictors of Malignancy
–– >5 mitoses/50 HPF = “high”
–– High MIB1 index
Gastrointestinal Cancers 277
–– Conroy 2011: 342 stage IV pancreatic cancer Post-op. Indications: R+, LN+
rand: FOLFIRINOX vs gem. MS was 11.1 vs –– Abdominal nodal volumes (peripancreatic,
6.8 m favoring FOLFIRINOX. celiac, SMA, porta hepatis, para-aortic), anas-
–– VonHoff 2013: Stage IV pancreatic cancer tomoses, tumor bed
patients rando: gem vs gem+abraxane. MS 6.7 –– Post-op nodal beds +1 cm
vs 8.5 m favoring gem+abraxane. –– Classic borders: Sup T10/11. Inf L3/4
–– Type I involves CHD; Type II is bifurcation. mets. No PV, HA invasion. No organ invasion.
IIIA is R IH duct. IIIB is L IH duct. IV is bilat- No DMs.
eral IH ducts. IV is multicentric. –– Transplant: only an option of EHCC (hilar).
–– Distal – 30%.
–– Surgery is a Whipple. Studies
–– Intrahepatic/diffuse/multifocal – 10%.
Papillary – best prognosis. CRT After Surgery
–– Venous drainage – PV (posterior to CBD). –– Limited data.
–– Lymphatic drainage – submucosa to porta –– Mayo Clinic (Gold 2009). Retrospective.
hepatis, celiac axis, pancreaticoduodenal LN. Stage I (T1–2 N0) or II (T3 N0 or T1–3 N1)
gallbladder. Median dose 50.4/28. 5-FU-based
–– GB cancer natural history: tends to chemo. Of 73 pts included, 25 received adj
metastasize. CRT. Adj CRT assoc. w improved OS.
–– HCC cancer natural history: LR spread pre- –– Wang, 2011: Nomogram of OS benefit with
dominates (e.g., liver, stomach, duodenum, adjuvant chemo or CRT for resect gallbladder
pancreas, colon, omentum, abd wall). LN CA.
mets in 40–80%. First echelon: cystic and –– Adjuvant chemo recommended if T4 and/or
pericholedochal. Second echelon: pancreati- N+.
coduodenal, celiac, PA. –– Adjuvant CRT has small benefit in T2–3 N0
–– DM rate: adenoCA of GB > IHCC > hilar CC. and has large benefit in T4 N0 or any N+.
–– LR continues over time (unlike HCC, where –– SWOG 0809 (Ben-Josef 2015). Phase II feasi-
LR is in 1 year). bility study of 79 patients with gallbladder or
extrahepatic cholangios (pT2–4 or N+ or posi-
–– E/IHCC risk factors: gallstones, PSC, UC, tive resection margins) treated with adjuvant
family hx, parasites, meds (e.g., INH, chemo + CRT. Chemo is cape 1500 mg/m2/d
thorotrast). (in two doses: 750 mg/m2 q12h) for 14d; and
–– GB risk factors: cholelithiasis; chronic gem 1 g/m2 on d1 and d8; give for 4 cycles
inflammation (e.g., salmonella); porcelain (12 w). Reimage. Then concurrent cape
GB; polyps; congenital cysts; obesity. 1330 mg/m2 (in two doses: 665 mg/m2
q12h) + RT 45, 5x per week for 5–6w. 2-yr OS
–– Workup: HP; labs: CBC, LFT, CA 19–9, was 65%.
coags, CEA, Hep panel; imaging: US will ID
obstruction location; CT chest w contrast; CRT Followed by Transplant
multiphasic abdominal/pelvic CT/MRI with –– Mayo (Rea, 2005). Hilar cholangiocarcinoma
contrast; cholangiogram; ERCP/MRCP. get NA-CRT 45 Gy/1.5 Gy BID w concurrent
5-FU, then 12–16 Gy intracavity BT of 6 Gy
–– Markers: CA 19–9 and CA 50 can be elevated x2 or 4 Gy x 4 BID, w concurrent 5-FU, fol-
in cholangiocarcinoma. lowed by the liver txp compared to similar
–– CA19–9 also seen in other GI malignancies, patients treated with resection. 5-year OS 82%
e.g., HCC, esophageal, CRC. for transplant versus 21% after resection.
–– CT, MRI, fiducial marker, 4DCT, IV con- –– Soliman 2013 (Toronto): phase II of 8 Gy x1
trast, arms up, vac lock, Abd compression to whole liver for patients with HCC of liver
or other motion management. tumor-related pain. Exclusion criteria: sys-
–– Late arterial phase of CT should be used bc temic tx in 2 w or TACE in 1 month. Plts >25,
contrast enhances within tumor allowing Hgb <7, INR >3, bili >100 umol/L, AST or
superior delineation vs liver parenchyma. This ALT >10 x normal. Premed with granisetron,
is in contrast to liver mets or IHCC. decadron 2 mg 1 h prior to RT. GTV. CTV is
–– GTV + ITV + 5 mm = PTV. majority of the liver. CTV + 1 cm = PTV. 8 Gy
–– SBRT dose varies by institution. 10–20 Gy x x1. 95% of PTV >7 Gy; improved QOL, 48%
3, 5–12 Gy x 5 are most common. Get BED10 had ↓symptoms at 1 month.
to >100.
–– More details in liver mets section. Non-SBRT Constraint: Mean <25 Gy
–– QUANTEC Constraints
Toxicity
QUANTEC Treating HCC Treating mets
Mean liver dose 3 fx 6 fx 3 fx 6fx
(liver – GTV)
–– Classic RILD: anicteric hepatomegaly, asci-
<13 Gy <18 Gy <15 Gy <20 Gy
Critical liver vol >700 mL normal liver receives
tes, elevated liver enzymes. 1–2 months after
≤15 Gy in 3 fx of ≤21 Gy in 5 fx RT. Fatigue, RUQ pain, ascites, high LFTs
(esp ALP). Pathologic: VOD, central venous
congestion, spares large veins, entraps RBCs.
Supportive management. 10% mortality
Whole-Liver RT Palliation
–– Non-Classic RILD: Elevation of liver
enzymes, reactivation of hepB or C, liver
–– Russell 1993: 1.5 Gy BID dose escalation for
function decline
patients with liver metastases. Recommended
–– Biliary obstruction
dose of 30 Gy (33 Gy was unsafe). No longer
–– GI Bleed, obstruction, fistula
performed.
–– Patients w baseline dysfunction, anything not
–– Hanson recommends 21 Gy in 7 fx.
fitting classic
Gastrointestinal Cancers 289
290 D. M. Trifiletti et al.
–– TROG 01.04 (Ngan 2012): T3 N0–2 rectal –– Endoscopically removed polyp with cancer
adenoCA. (1) 5x5, wait 1 month, surg, then 6c of indeterminate pathology
of 5-FU vs (2) 50.4/1.8 + 5-FU, surg, then –– No LVI, PNI
5-FU x4c. LR were 4.4% vs 7.5% (NS). ~28%
had DMs in both arms (NS). 5y OS similar, –– T1 N0: transanal vs APR/LAR
72%. Similar G3+ acute tox: 6%; chronic: 8%. –– High-risk features: T2, +margin, LVSI, G3.
Increased pCR in long-course arm 15% vs –– Post-op dose is 45 Gy + 5.4–9 Gy boost.
1%. “Either no difference or favoring LC for –– T2 N0 R0: FOLFOX x 2 m, then capeRT,
distal tumors.” then FOLFOX.
–– NSABP R-04 (Allegra 2015): Stage II-III rec- –– Post-op CRT if R+, T3+ or N+, may also
tal cancer getting preop RT 45/25 + 5.4– consider after transanal w/ high-risk fea-
10.8 Gy boost. Chemo CVI 5-FU vs tures that does not have surgical resection
capecitabine +/− oxaliplatin: no difference in as either definitive or neoadjuvant
OS or LC. Adding oxaliplatin only worsened treatment).
tox. Cap is SOC. –– Adjuvant chemo is FOLFOX (preferred)
for 6 months, q 2 weeks.
Local Recurrence
–– MDACC (Das 2010). 39 Gy in 1.5 Gy BID if
re treat >1 y or 30 Gy if <1 yr. Concurrent –– T3+ or N+: neoadj CRT, then TME, then
chemo in 98%. 36% had surg after RT. 3-year adj chemo
OS 39% for all; better if received surg, 66% vs –– Preoperative capeRT (45 Gy + 5.4 Gy to
27%. 3-yr freedom from LP was 33%. G3–4 primary) → surgery → FOLFOX x 8
tox 35%. (4 months).
–– Italy (Valentini 2006). 30 Gy in 1.2 Gy BID, –– Chemo is CI 5-FU 225 mg/m2 over 24 h (or
then 10.8 Gy boost w concurrent 5-FU. Then capecitabine 825 mg BID) during RT.
surg in 6–8 w. All received adjuvant Ralitrexed. –– Surgery is 6 weeks after CRT.
CR and PR 44%. Surgery in 51%. 5-year OS –– Low-lying = <5 cm from anal verge, need
67% if R0, 22% if R+ or no surg. G3 tox 5%. APR.
–– IORT (12.5–15 Gy) for positive margin.
NCCN –– If obstructing distal lesion → temporary
–– For 5 × 5, NCCN recommends surgery diverting colostomy. Avoid stent.
1–2 weeks after radiation, but there is some –– Unresectable: ChemoRT to 59.4 Gy.
evidence that waiting 4–8 weeks will ↑ pCR
rate and ↓ postoperative complications. 5 × 5 Resectable Mets
is not recommended for T4 tumors. –– FOLFOX × 6 (3 months) → capeRT → surger-
–– Even if patient needs APR, need to do NA- ies (synchronous/staged)
CRT because of the high recurrence rate. –– Liver/lung rules: Primary tumor out or can be
taken out, no unresectable extrahepatic/extra-
Rectal pulmonary disease, Complete resection
–– Local excision alone requirements (rule of possible
3 s):
–– <1/3 bowel circumference Technique
–– <3 cm –– Prone, belly board, PO ± IV contrast, full
–– SM > 3 mm bladder, (wire scar and include if post-op),
–– T1 N0 fuse CT sim with MRI.
–– Mobile –– If there is gross residual in a paracolic gutter,
–– G1–2 then sim the patient in the contra lateral decu-
–– <8 cm from anal verge bitus (e.g., if dz. in R, then sim L lat decub).
Gastrointestinal Cancers 297
• N1a – inguinal, mesorectal, internal iliac AJCC 7th Edition Staging (N Stage Differs
• N1b – external iliac only from 8th)
–– N1c – external iliac and any N1a node –– N1 – perirectal nodes
–– M1 – DMs, e.g., dermal involvement of vulva –– N2 – No perirectal
–– But, unilat internal iliac, unilat inguinal, or both
–– N3 – perirectal+inguinal, bilateral internal
iliac, bilateral inguinal
–– M1 – DMs, e.g., dermal involvement of vulva
AJCC 7 TNM T1 T2 T3 T4
N0 I II IIIA
N1: Perirectal IIIA IIIB
N2: Unilat int iliac or unilat IIIB
inguinal or both
N3: perirectal+inguinal,
bilateral internal iliac,
bilateral inguinal
M1 IV
Local Excision Alone or RT Alone 2 cm lat to greater sciatic notch. Then true pel-
–– Boman 1984: T1, G1, neg margins, <40% cir- vis (to 45 Gy): same field, but sup border
cumference, no sphincter involvement (LC lowed to inf border of the SI joints. If N0, then
>90%). field reduced off inguinal nodes also. 5 yr OS
–– Deniaud-Alexandre 2003: RT alone for with mitoC better (71 → 78%). 5-year
T1 N0, 100% LC. colostomy-free survival better w MMC 72%
vs 65%. 5 yr. DFS (68% vs 58%). G3+ heme
RT vs CRT acute tox worse w MMC, 62% vs 41%.
–– Nigro 1974: 30/15 + 5-FU + mitoC. 5-FU is –– EXTRA-(Glynne-Jones 2008): phase II data
100 mg/m2 CI x2c, and MMC is 15 mg/m2 of mitomycin combined with capecitabine
bolus d1. RT was AP/PA to pelvis and ingui- instead of 5-FU. Good results.
nals. Then surgery for some. 71% pCR. Same –– UKCCCR ACT II (James 2013): Can we
control with or without APR (~80%). replace MMC w cis? 50.4 Gy with
Foundation trial for definitive CRT. 5-FU + mitoC vs cisplatin. Then +/− mainte-
–– UKCCCR ACT I (Lancet 1996, Northover nance chemo. Superiority trial. n = 472. 3-year
2010): anal SCC, excluding T1 N0. Randomize PFS was 75%. No diff in PFS, OS, colostomy-
45 Gy/20–25 fx (institutional prefer- FS, or CR at 26w. At 26w, CR was 91% in
ence) + boost w Ir-192 BT+/− MMC arm and 90% in cis arm. 5-FU/
5-FU + mitoC. Split course RT. If no response, MMC + 50.4 Gy RT in 28 fx w/o maintenance
then pt. go to salvage surgery. At 12y, CRT recommended as SOC bc this was not a non-
improved LRC 66% vs 41%, colostomy-free inferiority trial.
survival 89% vs 73%, but not OS (27–33%). –– RTOG 0529 (Kachnic 2013): phase II of dose
CRT w 5-FU/MMC became SOC. painted IMRT. Entire pelvis gets up to 54/1.8
–– RTOG 8704 (Flam 1996): 45–50.4 Gy with (30 fx) depending on stage. Inguinal LNs get
5-FU +/− mitoC. 5-FU was 1000 mg/m2 on 45 Gy if negative, 50.4 if <3 cm (1.68 Gy/fx),
d1–4 and d29–32. MitoC was 10 mg/m2 on d1 and 54 Gy if >3 cm. A T2 N0 tumor in this
and 29. MMC improved CR rate (85 → 92%) protocol is treated w 50.4 Gy/1.8 Gy to pri-
and ↓colostomy rate (22% vs 9%) and 4-year mary and 42 Gy/1.5 Gy fx to pelvis and
DFS (51% vs 73%) but same OS. However, inguinal LNs. Less grade 2+ heme, grade 3+
MMC had more G4–5 tox, 26% vs 8%. GI, and grade 3+ derm toxicity but primary
–– EORTC trial (Bartelink 1997): similar ACT endpoint (combined acute grade 2 toxicity GI
I. 45/25 + boost of 15–20 Gy if CR or PR after and GU).
6w vs CRT 45/25 + 5-FU CI and MMC. CRT –– Contours per RTOG 0529.
arm had better LC, 68% vs 50% and
colostomy-free survival 72% vs 40%. 5-yr OS Anal Cancer Treatment Paradigm
similar – 56%. –– Everyone gets definitive CRT.
–– Salama 2007: 53 pts s/p che- –– MMC and 5-FU/capecitabine.
moIMRT. Favorable results. –– Chemotherapy wks 1 and 5.
–– CALGB 9281 (Meropol 2008): neoadj chemo –– Capecitabine 825 mg/m2 PO BID M-F.
phase II trial for T3–4 and/or N2–3. At 4 yrs. –– MMC 10 mg/m2, D1 and 29 (or 12 mg/m2 on
showed 68% OS and 50% sphincter D1 only if capecitabine).
preservation. –– Do not use cisplatin in place of MMC, unless
–– RTOG 9811 (Ajani 2008, Gunderson 2012): AIDS. RTOG 98–11: Worse OS and DFS with
T2–4,NX, n = 644 patients. 5-FU + cis with cis, although confounded by addition of
induction vs 5-FU + mitoC without induction. induction cis/5-FU in cis arm. UK ACT II: No
RT was 3D CRT. sup L5/S1. Whole pelvis difference b/w MMC and cis; no benefit to
(30.6 Gy): Inf 2.5 cm margin around tumor outback chemo.
and anus. Lat to include lat inguinal LNs and
302 D. M. Trifiletti et al.
upper third
(12-16 cm)
middle third
(6-12 cm)
peritoneal reflection
anal columns of
anal location
Morgagni terminology
rectal mucosa
levatorani
drainage to pelvic LNs lower third
pectinate/dentate line anoderm
analvalve (<6 cm)
squamous mucosa; anal crypts, glands
drainage to inguinal LNs pecten
bifurcate to ext./int iliacs (~sacral promontory) presacral space at the level. around anal verge (or from GTV, if perieanal skin involved).
Lateral: extend to lateral pelvic sidewall musculature or Same for 50.4 Gy or 5x5 Gy Lat, include <3 mm beyond levator ani (unless levator ani
bone. Unless evidence of extension into ischiorectal fossa, CTV45 = at least 2 cm inferior to involved, then 1–2 cm margin up to bone)
no need to go more >mm beyond levator m gross disease or to the pelvic floor, CTV node boost = GTV node +5-7 mm. No accepted
Anterior: 1 cm into the posterior bladder to account for whichever is lower. Lateral to standard.
day-to-day variation pelvic sidewall muscles, include CTV(42–50): remainder of the CTVA + B + C
Posterior: posterior internal obturator vessels (lie posterior portion of internal T3–4/N+ (30 fractions)
between external and internal iliacs), + 7–8 mm around obturator vessels. Sup to PTV_A = 54/30 at 1.8
vessels bifurcation of common iliacs to int/ PTV_A_B_C = 45/30 at 1.5
ext. iliacs. PTV_CD ≤3 cm = 50.4/30 at 1.68
PTV = CTV + 3-5 mm, inside skin PTV_CD ≥3 cm = 54/30 at 1.8
CTVB External iliac region Sometimes treated: T1–2/N0 (28 fractions)
Inf extent of internal obturator vessels (bony landmark: Cover if tumor extending anteriorly PTV_A = 50.4/28 at 1.8
upper edge of sup pubic rami) (i.e., T4 lesion – GI/GU structures), PTV_A_B_C = 42/28 at 1.5
recommend 7 mm margin in soft tissue around the iliac or abutting anal canal
vessels but at least 1 cm ant, especially if cN+
Trim off uninvolved muscle and bone
CTVC Inguinal region. Cover for tumors invading anal canal. Sometimes treated:
Should be 2 cm below saphenous/femoral junction Cover if invading anal canal
(continued)
305
Volume Description Rectal cancer (per RTOG 0822) Anal cancer
306
Dose Constraints [112] 9. Cunningham D, Allum WH, Stenning SP, et al.
Perioperative chemotherapy versus surgery alone for
RTOG 0822 RTOG 0529 resectable gastroesophageal cancer. N Engl J Med.
(rectal) (anal) 2006;355:11–20.
10. Gebski V, Burmeister B, Smithers BM, et al. Survival
Small bowel Dmax <50 Gy
benefits from neoadjuvant chemoradiotherapy or
V45 <65 cc V45 <20 cc
chemotherapy in oesophageal carcinoma: a meta-
V40 <100 cc analysis. Lancet Oncol. 2007;8:226–34.
V35 <180 cc V35 <150 cc 11. Kelsen DP, Winter KA, Gunderson LL, et al. Long-
V30 <300 cc V30 <200 cc term results of RTOG trial 8911 (USA Intergroup
Large bowel V45 <20 cc 113): a random assignment trial comparison of
V35 <150 cc chemotherapy followed by surgery compared with
V30 <200 cc surgery alone for esophageal cancer. J Clin Oncol.
Femoral heads V50 <0.5 cc V44 <5% 2007;25:3719–25.
V40 <35% 12. Minsky BD, Pajak TF, Ginsberg RJ, et al. INT 0123
V30 <50% (Radiation Therapy Oncology Group 94-05) phase
Bladder V40 <40% V35 <50% III trial of combined-modality therapy for esopha-
V50 <0.5 cc V40 <35% geal cancer: high-dose versus standard-dose radia-
V50 <5% tion therapy. J Clin Oncol. 2002;20:1167–74.
Kidneys V18 <2/3 of 13. Peyre CG, Hagen JA, DeMeester SR, et al. Predicting
kidney systemic disease in patients with esophageal cancer
Iliac crest V50 <5% after esophagectomy: a multinational study on the
V40 <35% significance of the number of involved lymph nodes.
V30 <50% Ann Surg. 2008;248:979–85.
14. Stahl M, Walz MK, Stuschke M, et al. Phase III
Pelvic bone
comparison of preoperative chemotherapy compared
marrow
with chemoradiotherapy in patients with locally
Genitalia V20 <50%
advanced adenocarcinoma of the esophagogastric
V30 <35%
junction. J Clin Oncol. 2009;27:851–6.
V40 <5%
15. Tepper J, Krasna MJ, Niedzwiecki D, et al. Phase III
trial of trimodality therapy with cisplatin, fluoroura-
cil, radiotherapy, and surgery compared with surgery
alone for esophageal cancer: CALGB 9781. J Clin
References Oncol. 2008;26:1086–92.
16. van Hagen P, Hulshof MC, van Lanschot JJ, et al.
1. Martinez-Monge R, Gomez-Iturriaga A, Cambeiro Preoperative chemoradiotherapy for esophageal or
M, et al. Phase I-II trial of perioperative high-dose- junctional cancer. N Engl J Med. 2012;366:2074–84.
rate brachytherapy in oral cavity and oropharyngeal 17. Sjoquist KM, Burmeister BH, Smithers BM, et al.
cancer. Brachytherapy. 2009;8:26–33. Survival after neoadjuvant chemotherapy or chemo-
2. Cook B DT. Wikibooks radiation oncology. radiotherapy for resectable oesophageal carci-
3. Edge S, Byrd D, Compton C, et al. AJCC cancer noma: an updated meta-analysis. Lancet Oncol.
staging manual. New York: Springer; 2010. 2011;12:681–92.
4. Hansen ERM. Handbook of evidence-based radia- 18. Sai H, Mitsumori M, Araki N, et al. Long-term results
tion oncology. 2nd ed: Springer; 2010. of definitive radiotherapy for stage I esophageal can-
5. Hristov BLS, Christodouleas JP. Radiation oncol- cer. Int J Radiat Oncol Biol Phys. 2005;62:1339–44.
ogy – A question based review. 2nd ed: LWW; 2014. 19. Gaspar LE, Winter K, Kocha WI, et al. A phase I/II
6. Bosset JF, Gignoux M, Triboulet JP, et al. study of external beam radiation, brachytherapy, and
Chemoradiotherapy followed by surgery compared concurrent chemotherapy for patients with local-
with surgery alone in squamous-cell cancer of the ized carcinoma of the esophagus (Radiation Therapy
esophagus. N Engl J Med. 1997;337:161–7. Oncology Group Study 9207): final report. Cancer.
7. Burmeister BH, Smithers BM, Gebski V, et al. 2000;88:988–95.
Surgery alone versus chemoradiotherapy followed 20. Siewert JR, Stein HJ. Classification of adenocarci-
by surgery for resectable cancer of the oesophagus: a noma of the oesophagogastric junction. Br J Surg.
randomised controlled phase III trial. Lancet Oncol. 1998;85:1457–9.
2005;6:659–68. 21. Urba SG, Orringer MB, Turrisi A, et al. Randomized
8. Cooper JS, Guo MD, Herskovic A, et al. trial of preoperative chemoradiation versus surgery
Chemoradiotherapy of locally advanced esophageal alone in patients with locoregional esophageal carci-
cancer: long-term follow-up of a prospective ran- noma. J Clin Oncol. 2001;19:305–13.
domized trial (RTOG 85-01). Radiation Therapy 3/ 22. Walsh TN, Noonan N, Hollywood D, et al. A
yj;Oncology Group. JAMA. 1999;281:1623–7. comparison of multimodal therapy and surgery
308 D. M. Trifiletti et al.
for esophageal adenocarcinoma. N Engl J Med. 37. Joensuu H, Eriksson M, Sundby Hall K, et al. One vs
1996;335:462–7. three years of adjuvant imatinib for operable gastro-
23. Welsh J, Settle SH, Amini A, et al. Failure patterns in intestinal stromal tumor: a randomized trial. JAMA.
patients with esophageal cancer treated with defini- 2012;307:1265–72.
tive chemoradiation. Cancer. 2012;118:2632–40. 38. Chauffert B, Mornex F, Bonnetain F, et al. Phase
24. Ajani JA, Winter K, Okawara GS, et al. Phase II III trial comparing intensive induction chemoradio-
trial of preoperative chemoradiation in patients with therapy (60 Gy, infusional 5-FU and intermittent cis-
localized gastric adenocarcinoma (RTOG 9904): platin) followed by maintenance gemcitabine with
quality of combined modality therapy and patho- gemcitabine alone for locally advanced unresectable
logic response. J Clin Oncol. 2006;24:3953–8. pancreatic cancer. Definitive results of the 2000-01
25. Bang YJ, Van Cutsem E, Feyereislova A, et al. FFCD/SFRO study. Ann Oncol. 2008;19:1592–9.
Trastuzumab in combination with chemotherapy 39. Conroy T, Desseigne F, Ychou M, et al. FOLFIRI-
versus chemotherapy alone for treatment of HER2- NOX versus gemcitabine for metastatic pancreatic
positive advanced gastric or gastro-oesophageal cancer. N Engl J Med. 2011;364:1817–25.
junction cancer (ToGA): a phase 3, open-label, ran- 40. Corsini MM, Miller RC, Haddock MG, et al.
domised controlled trial. Lancet. 2010;376:687–97. Adjuvant radiotherapy and chemotherapy for pan-
26. Bonenkamp JJ, Hermans J, Sasako M, et al. creatic carcinoma: the Mayo Clinic experience
Extended lymph-node dissection for gastric cancer. (1975-2005). J Clin Oncol. 2008;26:3511–6.
N Engl J Med. 1999;340:908–14. 41. Hazard L, Tward JD, Szabo A, et al. Radiation
27. Cuschieri A, Weeden S, Fielding J, et al. Patient sur- therapy is associated with improved survival in
vival after D1 and D2 resections for gastric cancer: patients with pancreatic adenocarcinoma: results
long-term results of the MRC randomized surgical of a study from the Surveillance, Epidemiology,
trial. Surgical Co-operative Group. Br J Cancer. and End Results (SEER) registry data. Cancer.
1999;79:1522–30. 2007;110:2191–201.
28. Gouzi JL, Huguier M, Fagniez PL, et al. Total versus 42. Herman JM, Swartz MJ, Hsu CC, et al. Analysis
subtotal gastrectomy for adenocarcinoma of the gas- of fluorouracil-based adjuvant chemotherapy and
tric antrum. A French prospective controlled study. radiation after pancreaticoduodenectomy for duc-
Ann Surg. 1989;209:162–6. tal adenocarcinoma of the pancreas: results of a
29. Hartgrink HH, van de Velde CJ, Putter H, et al. Neo- large, prospectively collected database at the Johns
adjuvant chemotherapy for operable gastric cancer: Hopkins Hospital. J Clin Oncol. 2008;26:3503–10.
long term results of the Dutch randomised FAMTX 43. Kalser MH, Ellenberg SS. Pancreatic cancer.
trial. Eur J Surg Oncol. 2004;30:643–9. Adjuvant combined radiation and chemother-
30. Kim HH, Hyung WJ, Cho GS, et al. Morbidity and apy following curative resection. Arch Surg.
mortality of laparoscopic gastrectomy versus open 1985;120:899–903.
gastrectomy for gastric cancer: an interim report--a 44. Krishnan S, Rana V, Janjan NA, et al. Induction che-
phase III multicenter, prospective, randomized Trial motherapy selects patients with locally advanced,
(KLASS Trial). Ann Surg. 2010;251:417–20. unresectable pancreatic cancer for optimal benefit
31. Macdonald JS, Smalley SR, Benedetti J, et al. from consolidative chemoradiation therapy. Cancer.
Chemoradiotherapy after surgery compared with 2007;110:47–55.
surgery alone for adenocarcinoma of the stom- 45. Loehrer PJ Sr, Feng Y, Cardenes H, et al. Gemcitabine
ach or gastroesophageal junction. N Engl J Med. alone versus gemcitabine plus radiotherapy in
2001;345:725–30. patients with locally advanced pancreatic cancer: an
32. Sasako M, Sano T, Yamamoto S, et al. D2 lymphad- Eastern Cooperative Oncology Group trial. J Clin
enectomy alone or with para-aortic nodal dissection Oncol. 2011;29:4105–12.
for gastric cancer. N Engl J Med. 2008;359:453–62. 46. Neoptolemos JP, Dunn JA, Stocken DD, et al.
33. Yu W, Choi GS, Chung HY. Randomized clinical Adjuvant chemoradiotherapy and chemotherapy
trial of splenectomy versus splenic preservation in in resectable pancreatic cancer: a randomised con-
patients with proximal gastric cancer. Br J Surg. trolled trial. Lancet. 2001;358:1576–85.
2006;93:559–63. 47. Oettle H, Post S, Neuhaus P, et al. Adjuvant chemo-
34. Tepper JE, Gunderson LL. Radiation treatment therapy with gemcitabine vs observation in patients
parameters in the adjuvant postoperative therapy of undergoing curative-intent resection of pancre-
gastric cancer. Semin Radiat Oncol. 2002;12:187–95. atic cancer: a randomized controlled trial. JAMA.
35. Le Cesne A, Ray-Coquard I, Bui BN, et al. 2007;297:267–77.
Discontinuation of imatinib in patients with 48. Regine WF, Winter KA, Abrams RA, et al.
advanced gastrointestinal stromal tumours after 3 Fluorouracil vs gemcitabine chemotherapy before
years of treatment: an open-label multicentre ran- and after fluorouracil-based chemoradiation follow-
domised phase 3 trial. Lancet Oncol. 2010;11:942–9. ing resection of pancreatic adenocarcinoma: a ran-
36. Bucher P, Taylor S, Villiger P, et al. Are there any domized controlled trial. JAMA. 2008;299:1019–26.
prognostic factors for small intestinal stromal 49. Rich T, Harris J, Abrams R, et al. Phase II study of
tumors? Am J Surg. 2004;187:761–6. external irradiation and weekly paclitaxel for non-
Gastrointestinal Cancers 309
metastatic, unresectable pancreatic cancer: RTOG- 63. Ben-David MA, Griffith KA, Abu-Isa E, et al.
98-12. Am J Clin Oncol. 2004;27:51–6. External-beam radiotherapy for localized extrahe-
50. Smeenk HG, van Eijck CH, Hop WC, et al. Long- patic cholangiocarcinoma. Int J Radiat Oncol Biol
term survival and metastatic pattern of pancreatic Phys. 2006;66:772–9.
and periampullary cancer after adjuvant chemora- 64. Hamamoto Y, Niino K, Ishiyama H, et al. Impact
diation or observation: long-term results of EORTC of pretreatment cholinesterase level on survival of
trial 40891. Ann Surg. 2007;246:734–40. inoperable intrahepatic or hepatic-hilar carcinomas
51. Stocken DD, Buchler MW, Dervenis C, et al. treated with three-dimensional conformal radiother-
Meta- analysis of randomised adjuvant ther- apy. Radiat Med. 2004;22:316–23.
apy trials for pancreatic cancer. Br J Cancer. 65. Gold DG, Miller RC, Haddock MG, et al. Adjuvant
2005;92:1372–81. therapy for gallbladder carcinoma: the Mayo
52. Valle JW, Palmer D, Jackson R, et al. Optimal dura- Clinic Experience. Int J Radiat Oncol Biol Phys.
tion and timing of adjuvant chemotherapy after 2009;75:150–5.
definitive surgery for ductal adenocarcinoma of the 66. Rea DJ, Heimbach JK, Rosen CB, et al. Liver
pancreas: ongoing lessons from the ESPAC-3 study. transplantation with neoadjuvant chemoradiation is
J Clin Oncol. 2014;32:504–12. more effective than resection for hilar cholangio-
53. Von Hoff DD, Ervin T, Arena FP, et al. Increased sur- carcinoma. Ann Surg. 2005;242:451–8; discussion
vival in pancreatic cancer with nab-paclitaxel plus 458–61.
gemcitabine. N Engl J Med. 2013;369:1691–703. 67. Bujold A, Massey CA, Kim JJ, et al. Sequential
54. Iacobuzio-Donahue CA, Fu B, Yachida S, et al. DPC4 phase I and II trials of stereotactic body radiotherapy
gene status of the primary carcinoma correlates with for locally advanced hepatocellular carcinoma. J
patterns of failure in patients with pancreatic cancer. Clin Oncol. 2013;31:1631–9.
J Clin Oncol. 2009;27:1806–13. 68. Dawson LA, Hashem S, Bujold A. Stereotactic body
55. Katz MH, Pisters PW, Evans DB, et al. Borderline radiation therapy for hepatocellular carcinoma. Am
resectable pancreatic cancer: the importance of Soc Clin Oncol Educ Book. 2012:261–4.
this emerging stage of disease. J Am Coll Surg. 69. Lau WY, Lai EC. Treatment of unresectable hepa-
2008;206:833–46.; discussion 846–8. tocellular carcinoma with transarterial radio-
56. Treatment of locally unresectable carcinoma of the embolization: iodine-131-lipiodol. ANZ J Surg.
pancreas: comparison of combined-modality therapy 2008;78:331–2.
(chemotherapy plus radiotherapy) to chemotherapy 70. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in
alone. Gastrointestinal Tumor Study Group. J Natl advanced hepatocellular carcinoma. N Engl J Med.
Cancer Inst. 1988;80:751–5. 2008;359:378–90.
57. Taniguchi CM, Murphy JD, Eclov N, et al. 71. Meng MB, Cui YL, Lu Y, et al. Transcatheter arte-
Dosimetric analysis of organs at risk during expi- rial chemoembolization in combination with radio-
ratory gating in stereotactic body radiation therapy therapy for unresectable hepatocellular carcinoma:
for pancreatic cancer. Int J Radiat Oncol Biol Phys. a systematic review and meta-analysis. Radiother
2013;85:1090–5. Oncol. 2009;92:184–94.
58. Ben-Josef E, Guthrie KA, El-Khoueiry AB, et al. 72. Russell AH, Clyde C, Wasserman TH, et al.
SWOG S0809: A Phase II Intergroup Trial of Accelerated hyperfractionated hepatic irradiation in
Adjuvant Capecitabine and Gemcitabine Followed the management of patients with liver metastases:
by Radiotherapy and Concurrent Capecitabine in results of the RTOG dose escalating protocol. Int J
Extrahepatic Cholangiocarcinoma and Gallbladder Radiat Oncol Biol Phys. 1993;27:117–23.
Carcinoma. J Clin Oncol. 2015;33:2617–22. 73. Rusthoven KE, Kavanagh BD, Cardenes H, et al.
59. Jarnagin WR, Fong Y, DeMatteo RP, et al. Staging, Multi-institutional phase I/II trial of stereotactic
resectability, and outcome in 225 patients with hilar body radiation therapy for liver metastases. J Clin
cholangiocarcinoma. Ann Surg. 2001;234:507–17; Oncol. 2009;27:1572–8.
discussion 517–9. 74. Soliman H, Ringash J, Jiang H, et al. Phase II trial of
60. Matsuo K, Rocha FG, Ito K, et al. The Blumgart pre- palliative radiotherapy for hepatocellular carcinoma
operative staging system for hilar cholangiocarci- and liver metastases. J Clin Oncol. 2013;31:3980–6.
noma: analysis of resectability and outcomes in 380 75. Tse RV, Hawkins M, Lockwood G, et al. Phase I
patients. J Am Coll Surg. 2012;215:343–55. study of individualized stereotactic body radiother-
61. Tao R, Krishnan S, Bhosale PR, et al. Ablative apy for hepatocellular carcinoma and intrahepatic
Radiotherapy Doses Lead to a Substantial cholangiocarcinoma. J Clin Oncol. 2008;26:657–64.
Prolongation of Survival in Patients With Inoperable 76. Klein J, Dawson LA. Hepatocellular carcinoma radi-
Intrahepatic Cholangiocarcinoma: A Retrospective ation therapy: review of evidence and future opportu-
Dose Response Analysis. J Clin Oncol. nities. Int J Radiat Oncol Biol Phys. 2013;87:22–32.
2016;34:219–26. 77. Wahl DR, Stenmark MH, Tao Y, et al. Outcomes
62. Valle J, Wasan H, Palmer DH, et al. Cisplatin plus after stereotactic body radiotherapy or radiofre-
gemcitabine versus gemcitabine for biliary tract can- quency ablation for hepatocellular carcinoma. J Clin
cer. N Engl J Med. 2010;362:1273–81. Oncol. 2016;34:452–9.
310 D. M. Trifiletti et al.
78. Penna C, Nordlinger B. Colorectal metastasis (liver vant treatment for colon cancer. N Engl J Med.
and lung). Surg Clin North Am. 2002;82:1075–90. 2004;350:2343–51.
x-xi. 92. Yu CS, Kim TW, Kim JH, et al. Optimal time interval
79. Eriguchi T, Takeda A, Sanuki N, et al. Acceptable between capecitabine intake and radiotherapy in pre-
toxicity after stereotactic body radiation therapy for operative chemoradiation for locally advanced rectal
liver tumors adjacent to the central biliary system. cancer. Int J Radiat Oncol Biol Phys. 2007;67:1020–6.
Int J Radiat Oncol Biol Phys. 2013;85:1006–11. 93. Myerson RJ, Genovesi D, Lockett MA, et al. Five
80. Collette L, Bosset JF, den Dulk M, et al. Patients with fractions of preoperative radiotherapy for selected
curative resection of cT3-4 rectal cancer after pre- cases of rectal carcinoma: long-term tumor control
operative radiotherapy or radiochemotherapy: does and tolerance to treatment. Int J Radiat Oncol Biol
anybody benefit from adjuvant fluorouracil-based Phys. 1999;43:537–43.
chemotherapy? A trial of the European Organisation 94. O'Connell MJ, Martenson JA, Wieand HS, et al.
for Research and Treatment of Cancer Radiation Improving adjuvant therapy for rectal cancer by
Oncology Group. J Clin Oncol. 2007;25:4379–86. combining protracted-infusion fluorouracil with
81. Fisher B, Wolmark N, Rockette H, et al. Postoperative radiation therapy after curative surgery. N Engl J
adjuvant chemotherapy or radiation therapy for rec- Med. 1994;331:502–7.
tal cancer: results from NSABP protocol R-01. J 95. Gerard JP, Azria D, Gourgou-Bourgade S, et al.
Natl Cancer Inst. 1988;80:21–9. Clinical outcome of the ACCORD 12/0405
82. Frykholm GJ, Glimelius B, Pahlman L. Preoperative PRODIGE 2 randomized trial in rectal cancer. J Clin
or postoperative irradiation in adenocarcinoma of Oncol. 2012;30:4558–65.
the rectum: final treatment results of a randomized 96. Bujko K, Nowacki MP, Nasierowska-Guttmejer A,
trial and an evaluation of late secondary effects. Dis et al. Long-term results of a randomized trial com-
Colon Rectum. 1993;36:564–72. paring preoperative short-course radiotherapy with
83. Gerard JP, Conroy T, Bonnetain F, et al. Preoperative preoperative conventionally fractionated chemoradi-
radiotherapy with or without concurrent fluoroura- ation for rectal cancer. Br J Surg. 2006;93:1215–23.
cil and leucovorin in T3-4 rectal cancers: results of 97. Das P, Delclos ME, Skibber JM, et al.
FFCD 9203. J Clin Oncol. 2006;24:4620–5. Hyperfractionated accelerated radiotherapy for rec-
84. Krook JE, Moertel CG, Gunderson LL, et al. tal cancer in patients with prior pelvic irradiation. Int
Effective surgical adjuvant therapy for high-risk rec- J Radiat Oncol Biol Phys. 2010;77:60–5.
tal carcinoma. N Engl J Med. 1991;324:709–15. 98. Cukier M, Smith AJ, Milot L, et al. Neoadjuvant
85. Martenson JA Jr, Willett CG, Sargent DJ, et al. chemoradiotherapy and multivisceral resection for
Phase III study of adjuvant chemotherapy and radia- primary locally advanced adherent colon cancer:
tion therapy compared with chemotherapy alone a single institution experience. Eur J Surg Oncol.
in the surgical adjuvant treatment of colon cancer: 2012;38:677–82.
results of intergroup protocol 0130. J Clin Oncol. 99. Ajani JA, Winter KA, Gunderson LL, et al.
2004;22:3277–83. Fluorouracil, mitomycin, and radiotherapy vs fluo-
86. Pahlman L, Glimelius B. Pre- or postoperative rouracil, cisplatin, and radiotherapy for carcinoma of
radiotherapy in rectal and rectosigmoid carcinoma. the anal canal: a randomized controlled trial. JAMA.
Report from a randomized multicenter trial. Ann 2008;299:1914–21.
Surg. 1990;211:187–95. 100. Bartelink H, Roelofsen F, Eschwege F, et al.
87. Peeters KC, Marijnen CA, Nagtegaal ID, et al. Concomitant radiotherapy and chemotherapy is
The TME trial after a median follow-up of 6 years: superior to radiotherapy alone in the treatment of
increased local control but no survival benefit in irra- locally advanced anal cancer: results of a phase III
diated patients with resectable rectal carcinoma. Ann randomized trial of the European Organization for
Surg. 2007;246:693–701. Research and Treatment of Cancer Radiotherapy and
88. Roh MS, Colangelo LH, O'Connell MJ, et al. Gastrointestinal Cooperative Groups. J Clin Oncol.
Preoperative multimodality therapy improves 1997;15:2040–9.
disease-free survival in patients with carci- 101. Boman BM, Moertel CG, O'Connell MJ, et al.
noma of the rectum: NSABP R-03. J Clin Oncol. Carcinoma of the anal canal. A clinical and patho-
2009;27:5124–30. logic study of 188 cases. Cancer. 1984;54:114–25.
89. Russell AH, Harris J, Rosenberg PJ, et al. Anal 102. Deniaud-Alexandre E, Touboul E, Tiret E, et al.
sphincter conservation for patients with adenocarci- Epidermoid carcinomas of the anal canal treated
noma of the distal rectum: long-term results of radi- with definitive radiation therapy in a series of 305
ation therapy oncology group protocol 89-02. Int J patients. Cancer Radiother. 2003;7:237–53.
Radiat Oncol Biol Phys. 2000;46:313–22. 103. Flam M, John M, Pajak TF, et al. Role of mitomycin
90. Sauer R, Becker H, Hohenberger W, et al. Preopera- in combination with fluorouracil and radiotherapy,
tive versus postoperative chemoradiotherapy for rec- and of salvage chemoradiation in the definitive non-
tal cancer. N Engl J Med. 2004;351:1731–40. surgical treatment of epidermoid carcinoma of the
91. Andre T, Boni C, Mounedji-Boudiaf L, et al. anal canal: results of a phase III randomized inter-
Oxaliplatin, fluorouracil, and leucovorin as adju- group study. J Clin Oncol. 1996;14:2527–39.
Gastrointestinal Cancers 311
104. Glynne-Jones R, Meadows H, Wan S, et al. EXTRA – moid anal cancer: 13-year follow-up of the first ran-
a multicenter phase II study of chemoradiation using domised UKCCCR Anal Cancer Trial (ACT I). Br J
a 5 day per week oral regimen of capecitabine and Cancer. 2010;102:1123–8.
intravenous mitomycin C in anal cancer. Int J Radiat 109. Salama JK, Mell LK, Schomas DA, et al. Concurrent
Oncol Biol Phys. 2008;72:119–26. chemotherapy and intensity-modulated radiation
105. James RD, Glynne-Jones R, Meadows HM, et al. therapy for anal canal cancer patients: a multicenter
Mitomycin or cisplatin chemoradiation with or experience. J Clin Oncol. 2007;25:4581–6.
without maintenance chemotherapy for treatment 110. Pineda CE, Berry JM, Jay N, et al. High-resolution
of squamous-cell carcinoma of the anus (ACT II): a anoscopy targeted surgical destruction of anal high-
randomised, phase 3, open-label, 2 × 2 factorial trial. grade squamous intraepithelial lesions: a ten-year
Lancet Oncol. 2013;14:516–24. experience. Dis Colon Rectum. 51:829–35; vdiscus-
106. Kachnic LA, Winter K, Myerson RJ, et al. RTOG sion 835–7, 2008.
0529: a phase 2 evaluation of dose-painted intensity 111. Ng M, Leong T, Chander S, et al. Australasian
modulated radiation therapy in combination with Gastrointestinal Trials Group (AGITG) contour-
5-fluorouracil and mitomycin-C for the reduction of ing atlas and planning guidelines for intensity-
acute morbidity in carcinoma of the anal canal. Int J modulated radiotherapy in anal cancer. Int J Radiat
Radiat Oncol Biol Phys. 2013;86:27–33. Oncol Biol Phys. 2012;83:1455–62.
107. Nigro ND, Vaitkevicius VK, Considine B Jr. 112. Marks LB, Yorke ED, Jackson A, et al. Use of
Combined therapy for cancer of the anal canal: a pre- normal tissue complication probability mod-
liminary report. Dis Colon Rectum. 1974;17:354–6. els in the clinic. Int J Radiat Oncol Biol Phys.
108. Northover J, Glynne-Jones R, Sebag-Montefiore D, 2010;76:S10–9.
et al. Chemoradiation for the treatment of epider-
Genitourinary Cancers
8
Nicholas G. Zaorsky, Daniel M. Trifiletti,
and Katherine Tzou
Abstract
This chapter discusses the general manage-
ment of patients with genitourinary tract can-
cers, with special focus on principles that
guide radiotherapy management. Several key
components of trimodality care and brachy-
therapy are discussed.
N. G. Zaorsky (*)
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
D. M. Trifiletti · K. Tzou
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu;
Tzou.Katherine@mayo.edu
Ataxia telangiectasia
Preexisting rectal fistula
Risk of bleeding (e.g. from anticoagulants) ** ** ** **
Moderate-severe urinary symptoms (e.g. high IPSS score, typically defined as > 20) Consider Consider Consider
CFRT CFRT CFRT
History of prior pelvic radiotherapy * *
Large prostate (e.g. >60 cm3) N/A
Large median lobes N/A
Inflammatory bowel disease * *
Pubic arch interference (e.g. prior pelvic fracture, irregular pelvic anatomy, or a penile
prosthesis)
Patient peak flow rate <10 cc/s and post void residual volume prior to brachytherapy > 100 N/A N/A N/A N/A
cc
RT treatment options for men with NCCN risk group-stratified prostate cancer, in relation to other treatment options.
NCCN risk group
Post-RP RT19
Very Low Adjuvant
T1c, Low Intermediate indications: pT3;
and GS <6, High Very High positive SMs, GS
T1-T2a, T2b-T2c,
and PSA <10 ng/mL, T3a, T3b-T4, 8-10, seminal
and GS < or GS 7 (3+4)/GG 2,
Options and subtypes and <3 positive cores or GS 8/GG 4, or primary GP vesicle involvement
6/GG 1, or GS 7 (4+3)/GG 3,
with <50% or GS 9-10/GG 5, 5/GG 5, Salvage
and PSA <10 or PSA ≥10 ng/ml ≤
cancer/core, or PSA > 20 ng/ml, or >4 cores with indications:
ng/ml, 20 ng/ml
and PSA density <0.15 GS 8-10/ GG 4 or 5 suspected LR (e.g.
ng/mL/g rising PSA,
imaging, biopsy-
proven)
Definitions
Prostate brachytherapy terms. Note, these are generally specific to prostate, do not apply to
other organs
D0.1 cc or Dmax The average dose to the hottest point of a volume. “D0.1 cc” is sometimes used because this
approximates the maximum dose to the smallest volume that can be calculated on a computer
D2cc The average dose to 2 cc of a volume
D10 The average dose to 10% of a volume, in Gy. The urethra D10 should be <150% of the
prescribed dose. This constraint limits the dose to the urethra
D30 The average dose to 30% of a volume, in Gy. The urethra D30 should be <130% of the
prescribed dose. This constraint limits the dose to the urethra
D90 In prostate cancer brachytherapy, this is the minimum dose in the hottest 90% of a volume, in
Gy. The prostate D90% should be >100%. This constraint ensures the prostate volume receives
adequate dose
Dwell time The time that the 192Ir source spends in a predetermined dwell position during HDR-BT. A
longer dwell time in a position translates to a greater dose deposited in the volume around the
position
Dwell position The position where a 192Ir source is located during HDR-BT. A combination of dwell
positions in different needles allows the delivery of a predetermined dose to the CTV
Hot spot A colloquialism used to describe volume outside the PTV which receives dose larger than
100% of the specified PTV dose
RV100 In prostate cancer brachytherapy, this is the volume of the rectum receiving 100% of the dose
and should be <1 cc
V100 In prostate cancer brachytherapy, this is the percentage of a structure receiving 100% of the
dose. For example, the V100 for the prostate should be >90%, meaning that 100% of the
prostate CTV should receive more than 90% of the prescribed dose. V100 is a surrogate for
post-implant D90
V150 In prostate cancer brachytherapy, this is the percentage of a structure receiving 150% of the
dose. The V150 for the prostate CTV should be <50–60%, meaning that <50–60% of the CTV
should receive >150% of the prescribed dose. V150 is surrogate for chronic urethral toxicity
UV5 In prostate cancer brachytherapy, this is the average dose to 5% of the urethral volume
receiving the highest dose. The UV5 should receive <150% of the dose
UV30 In prostate cancer BT, this is the average dose to 30% of the urethral volume receiving the
highest dose. The UV30 should be <125% of the dose
UV150 In prostate cancer BT, this is the volume of the urethra receiving 150% of the prescribe
dose. UV150 of the urethra should be 0%, meaning that 0% of the volume should receive
150% of the prescribed dose
Genitourinary Cancers 319
Average
Mono dose Boost dose t1/2 energy
Radionuclide (Gy) (Gy) (days) (keV) Prostate (CTV) Urethra Rectum
EBRT is D90 V100 V150
45–50.4
125I 145 110 59.4 28.4 >100% of >90– <50– UV150 ~0 (in volume) V100 D2cc < prescribed dose and
103Pd 110 100 17.0 20.7 dose 95% 60% UV5 <150% <1 cc on D0.1 cc <200 Gy
131Cs 120 9.7 30.4 UV30 <125% day 0; and
<1.3 cc on
day 30
192Ir 13.5 × 2 15 73.8 380 >90– D0.1 ≤120 Gy EQD2 D2cc ≤75 Gy EQD2
9.5 × 4 4 × 8.5 95% of D10 ≤120 Gy EQD2
dose D30 ≤105 Gy EQD2
V150 = 0 cc
No normal tissue constraints from ABS due to wide range of fractionation
options [7]
N. G. Zaorsky et al.
Genitourinary Cancers 321
EBRT SBRT
–– Fiducials for guidance.
Conventional and Hypofractionation –– Contour prostate urethra (insert Foley if no
–– Bowel prep. MRI).
–– Supine. –– GTV: prostate w/o SVs.
–– Comfortably full bladder. –– PTV: GTV + 3 mm.
–– Immobilized w/custom thermoplastic casts. –– D95 >100% prescription.
–– CT and MRI (3 mm) images obtained. No –– Maximum point dose <120%.
MRI for pacer or metal foreign bodies (orbital –– <15% of the PTV or <10 cc should be treated
XR). MRI before Calypso placed. to >115% of prescription dose.
–– For intermediate risk, proximal 1 cm of SVs –– Ratio of prescription isodose volume to PTV
should be covered by CTV (per RTOG 0815). volume of <1.2.
–– Definition of PTV: –– Volume outside PTV receiving >100% of dose
–– Prostate +8 mm (except posteriorly) should be <15% of PTV volume.
–– Prostate +5 mm posteriorly –– Hot spots in CTV if possible.
–– Proximal/distal SVs same as prostate –– Urethra: maximum point dose 110%,
–– LNs → no margin V39.3 Gy <20%.
–– CFRT: 75.6–79.2 Gy @ 1.8/fx, 80 Gy @
2 Gy / fx.
–– HFRT:
• FCCC: 70.2 Gy @ 2.7/fx
• MDACC: 72 Gy @ 2.4/fx.
• 0415: 70 Gy @ 2.5/fx
• Italy: 62 Gy @ 3.1/fx
–– NCIC 52.5 Gy @ 2.63/fx
–– No nodes/ADT for low risk
–– On treatment:
• Daily KV imaging if fiducials.
• Daily CBCT if no fiducials. Align to PTV.
322 N. G. Zaorsky et al.
• Abiraterone + • Abiraterone +
prednisone • Docetaxel • Abiraterone + prednisone
• Observation,
• Docetaxel • Radium-223 prednisone • Cabazitaxel • Palliative care
continued ADT • Enzalutamide
• GnRH agonist • Sipuleucel T • Enzalutamide • Radium-223
• Enzalutamide • Radium-223
• GnRH
antagonist • ADT +/-
• GnRH analog+
chemotherapy
Therapy
AA • 1st gen AA • Abiraterone + • Ketoconazole + • Ketoconazole +
• Abiraterone +
Indicated
• orchiectomy • Ketoconazole + prednisone steroid steroid
• 1st gen AA • Other prednisone
steroid • Ketoconazole +
• 1st gen ASI • Enzalutamide
• Observation radionuclide
steroid • ketoconazole +
Disease progression
• 1st gen ASI therapy • Docetaxel
• Mitoxantrone steroid
• Other continuation / • Radionuclide
radionuclide • Docetaxel retreatment therapy
therapy • Mitoxantrone
SRE prevention
• Systemic
chemotherapy* • Sipuleucel-T • Sipuleucel-T
• Immunotherapy*
Contraindicated
Standard of care Recommendation Option Expert opinion *= unless in clinical trial
• Benefits are < or > harms • Benefits are < or > harms • Benefits are = harms
KEY • Level of evidence: A or B • Level of evidence: C • Level of evidence: A, B, or C
• May be for select patients
NICHOLAS G. ZAORSKY. M.D.
N. G. Zaorsky et al.
Genitourinary Cancers 327
–– RTOG 8531, Lawton, 2005: Subset analysis 90d. 35% received aRT. The CSS rate at 5y
of N+ patients of 8531. 173 patients had pN+ was 96% vs 82%, favoring CMT. 2 groups
dz. in this group 98 received RT + immediate benefits (1) Patients with <3 + LNs and GS7–
goserelin, and 75 patients got RT alone with 10 with pT3b/pT4 dz. or R+; (2) Patients with
goserelin at relapse. 9 yr. OS 38 → 62%. 95 3–4 + LNs, regardless of any other character-
PFS 33% vs 4%. Thus, immediate ADT won. istics. Other patients (e.g., >5) did not benefit
–– Abdollah, 2014: Milan + Mayo data. 1107 from aRT, suggesting extra-pelvic dz.
pN1 patients analyzed to determine benefit of –– Zagars, 2001. 255, retrospective. ADT vs
aRT. All had RP + EPLND, then aRT within ADT + RT. improves 10y OS, 46% vs 67%.
332 N. G. Zaorsky et al.
Gynecomastia Prophylaxis 4/50, and 17/50. Breast pain: 29/51, 3/50, and
15/50. Thus, both tam and RT reduce risk of
Studies breast pain and gynecomastia for men taking
–– Perdona, 2005: tam vs RT for prevention of AA. RT good as tam alternative.
breast pain and gynecomastia during bicalu-
tamide monotherapy. n = 151. Rando to 3 Regimens
arms: (1) bicalutamide 150 mg QD; (2) bicalu- –– 4 Gy × 3 or 10 Gy × 1 with en face electrons
tamide 150 mg QD + tam 10 mg QD; (3) prescribed to 85% IDL. 10 cm diameter circu-
bicalutamide 150 mg QD + RT, one 12 Gy lar block centered over the nipple. No bolus
fraction when starting bic. In these arms, since skin not target.
development of gynecomastia was 35/51,
Genitourinary Cancers 333
–– Men: prostate, SVs, prox vas deferens, prox Reconstructive Options Following
urethra, and margin of fat and peritoneum. Cystectomy
–– PLND performed. pelvic LNs distal to com- –– Ileal conduit – conduit for urine to drain from
mon iliac bifurcation removed. Presacral LNs kidneys, exteriorized via stoma through
and common iliac LNs up to bifurcation abdominal wall (continuous drainage of urine).
removed. –– Continent cutaneous diversion – Kock,
–– 42% upstaging and 6% downstaging follow- Indiana, Miami pouch, low-pressure pouch
ing surgery. from bowel segments connected to the skin
require intermittent clean catheterization (no
stoma needed).
Common
AFP 6 5–7d <1K 1–10K >10K ng/mL NSGCT. HCC, cirrhosis, hepatitis
Never
GCT.
Need to have… all any any
Stage with N0 IS IS IS
Stage with N+ IIIB IIIC
Needed for risk M0 M0 M1
group
Risk group Good Intermed Poor
(need all (need all (need
above) above) ANY
above)
Genitourinary Cancers 343
Tumor size >4 cm and (3) rete testis invasion –– Long-term effects of treatment:
identified as risk factors. –– CV disease HR = 5.3 with CRT, HR = RT
–– Chung, 2010: validation study of the 2 RFs: alone, HR = 1 for surgery alone.
rete testes involvement did not predict relapse. –– Metabolic syndrome: 17% for CRT vs 10%
for carbo or surg.
NCCN –– Duodenal ulcer: 5% risk if orchiectomy.
–– All patients: radical transinguinal orchiec- –– Bleo-induced pneumonitis in 10–20%, of
tomy with high ligation of the spermatic cord those 10% die.
(never biopsy), then repeat tumor markers. –– Non-fatherhood with RT: 18%.
LNs not routinely dissected unless it is NSGCT. –– Nephro-/neurotoxicity with cisplatin.
• General options: surveillance (NCCN pre- –– Risk of contralateral cancer: 0.1% per year.
ferred); adjuvant RT (20 Gy to RP/PA –– Wait 1 year to have kid.
LNs), adjuvant single agent chemo. –– 50% have azoospermia at diagnosis
–– Risk factors of recurrence: size >4 cm. –– Only 30% can have kids.
–– Surveillance: At 15 yrs., 20% may relapse,
<1% CSM. Adjuvant Seminoma
–– Follow-up studies:
• AFP, b-hCG, LDH – q3–4 mo years 1–2, –– I: Obs* (13–16% LRF) or 20 Gy PA or carbo-
q6–12 mo years 3–4, then annually. platin AUC 6. CSS is 99% for all! Previously,
• CT abdo/pelvis – q6 mo years 1–2, rete testis invasion and tumor size >4 cm were
q6–12 mo year 3, annually yr. 4–5. risk factors for relapse; since 2010, these are
• CXR as clinically indicated. no longer used.
–– High-risk features for recurrence: LVI+, size –– For observation. HP q 3–6 m, CT at 3,6,12 m.
>4 cm, hCG >200 IU/L.
–– Stage I seminoma RT:
Counsel –– PA field is T11/12 (note, lower than Fossa/
–– Fertility and sperm banking (per above) MRC trial) to L5/S1, laterally through trans-
–– Surveillance (per above) verse process (~2 cm margin on nodes).
–– Second malignancy: 36% vs 23% in general –– For a right-sided tumor, includes paracaval,
population. RR = ~2 for RT, chemo, or precaval, and interaortocaval LNs.
combination –– For a left-sided tumor, include the above plus
at least the latero-aortic and preaortic LNs.
–– CTV = (IVC + 1.2 cm) + (aorta +1.9 cm).
–– PTV = CTV + 0.5 cm. A 0.7 cm uniform mar-
gin around PTV to block edge accounts for
penumbra.
–– AP-PA, never IMRT
• for a left-sided tumor, include the above
plus at least the lateroaortic and preaortic
LNs and include left renal hilum.
Genitourinary Cancers 345
–– IIA: 20 Gy* (dogleg+boost GTV to 30) or cis/ –– IIC-III: chemo (cis/etop +/− bleo) x 3 cycles.
etop x2c –– Adjuvant NSGCT: RT palliative only.
• Prior CTV for PA LNs for a stage I. –– I: obs (30% LRF) vs RP-LND (30% path
–– Note modified dogleg I used for N+ seminoma. +) vs cis/etop/bleo x 3c (usually chemo not
–– IIB: Cis/etop x2c is preferred OR 20 Gy recommended).
(dogleg+boost GTV to 36) if LN is <3 cm. –– II:
–– Contour the ipsi common, external, and proxi- • pN1: Surveillance preferred bc 60–90%
mal internal iliac veins and arteries down infe- of patients are cures with RPLND alone.
riorly to the upper border of the acetabulum. If there is a low LN burned and the
–– Dogleg: PA field but down to mid-obturator serum markers normalize after surgery,
foramen (near top of acetabulum). 20/10, then observe; if not, then consider 2c EP
boost IIA nodes to 30 Gy, IIB nodes to 36 Gy. or BEP.
–– CTV = PA CTV + 1.2 cm vessel expansion, • pN2: 2c EP or BEP.
exclude bone and bowel. • pN3: 3c EP or BEP.
–– CTV boost = GTV + 0.8 cm, exclude bone and –– IIIB or IIIC: BEP x 4 c.
bowel.
–– PTV = CTV + 0.7 cm to block edge Positive tumor markers after surgery → chemo
–– D50 <8 Gy for each kidney.
346 N. G. Zaorsky et al.
Treatment Paradigm
–– (1) Cis-based chemo. Chemo is usually BEP
x3 or EP x 4
–– (2) Surg for residual disease. +/− 40 Gy for
residual dz.
• objective response rate is 90%, with 2/3 of
patients having CR.
352 N. G. Zaorsky et al.
Background Background
–– 20% cN+ versus penile 50% cN+ –– Renal pelvis 3–4x more common than ureter.
–– But 90% of cN+ are pN+ –– UUT have worse stage-for-stage prognosis
than bladder.
Treatment –– 20% UUT tumors → synchronous/metachro-
–– Prostatic urethra = radical cystoprostatec- nous bladder cancer (only 1–4% vice versa).
tomy + PLND
–– Bulbomembranous = radical cystoprostatec- Treatment
tomy + en bloc penectomy + PLND Radical nephroureterectomy: radical nephrec-
–– Penile urethra = treat like penile tomy including ureter + part of bladder.
Genitourinary Cancers 355
seminal vesicles be included in the clinical target vol- 3): an open randomised phase III trial. Lancet.
ume? Int J Radiat Oncol Biol Phys. 2002;54:686–97. 2009;373:301–8.
32. Asbell SO, Krall JM, Pilepich MV, et al. Elective 45. Morris WJ, Tyldesley S, Rodda S, et al. Androgen
pelvic irradiation in stage A2, B carcinoma of the suppression combined with elective nodal and dose
prostate: analysis of RTOG 77-06. Int J Radiat Oncol escalated radiation therapy (the ASCENDE-RT
Biol Phys. 1988;15:1307–16. trial): an analysis of survival endpoints for a
33. Bolla M, Collette L, Blank L, et al. Long-term results randomized trial comparing a low-dose-rate
with immediate androgen suppression and external brachytherapy boost to a dose-escalated exter-
irradiation in patients with locally advanced prostate nal beam boost for high- and intermediate-risk
cancer (an EORTC study): a phase III randomised prostate Cancer. Int J Radiat Oncol Biol Phys.
trial. Lancet. 2002;360:103–6. 2017;98:275–85.
34. Bolla M, de Reijke TM, Van Tienhoven G, et al. 46. Hanks GE, Pajak TF, Porter A, et al. Phase III trial of
Duration of androgen suppression in the treatment of long-term adjuvant androgen deprivation after neo-
prostate cancer. N Engl J Med. 2009;360:2516–27. adjuvant hormonal cytoreduction and radiotherapy
35. Crook J, Patil N, Wallace K, et al. A phase III ran- in locally advanced carcinoma of the prostate: the
domized trial of the timing of meloxicam with radiation therapy oncology group protocol 92-02. J
iodine-125 prostate brachytherapy. Int J Radiat Clin Oncol. 2003;21:3972–8.
Oncol Biol Phys. 2010;77:496–501. 47. Pisansky TM, Hunt D, Gomella LG, et al. Duration
36. Denham JW, Steigler A, Lamb DS, et al. Short-term of androgen suppression before radiotherapy for
androgen deprivation and radiotherapy for locally localized prostate cancer: radiation therapy oncol-
advanced prostate cancer: results from the trans- ogy group randomized clinical trial 9910. J Clin
Tasman radiation oncology group 96.01 randomised Oncol. 2015;33:332–9.
controlled trial. Lancet Oncol. 2005;6:841–50. 48. Zapatero A, Guerrero A, Maldonado X, et al. High-
37. Horwitz EM, Bae K, Hanks GE, et al. Ten-year fol- dose radiotherapy with short-term or long-term
low-up of radiation therapy oncology group protocol androgen deprivation in localised prostate cancer
92-02: a phase III trial of the duration of elective (DART01/05 GICOR): a randomised, controlled,
androgen deprivation in locally advanced prostate phase 3 trial. Lancet Oncol. 2015;16:320–7.
cancer. J Clin Oncol. 2008;26:2497–504. 49. Spiotto MT, Hancock SL, King CR. Radiotherapy
38. Jones CU, Hunt D, McGowan DG, et al. Radiotherapy after prostatectomy: improved biochemical relapse-
and short-term androgen deprivation for localized free survival with whole pelvic compared with
prostate cancer. N Engl J Med. 2011;365:107–18. prostate bed only for high-risk patients. Int J Radiat
39. Lawton CA, DeSilvio M, Roach M 3rd, et al. An Oncol Biol Phys. 2007;69:54–61.
update of the phase III trial comparing whole pel- 50. Hanks GE, Buzydlowski J, Sause WT, et al. Ten-
vic to prostate only radiotherapy and neoadjuvant to year outcomes for pathologic node-positive patients
adjuvant total androgen suppression: updated analy- treated in RTOG 75-06. Int J Radiat Oncol Biol
sis of RTOG 94-13, with emphasis on unexpected Phys. 1998;40:765–8.
hormone/radiation interactions. Int J Radiat Oncol 51. Bolla M, van Poppel H, Collette L, et al.
Biol Phys. 2007;69:646–55. Postoperative radiotherapy after radical prostatec-
40. Pilepich MV, Winter K, Lawton CA, et al. Androgen tomy: a randomised controlled trial (EORTC trial
suppression adjuvant to definitive radiotherapy 22911). Lancet. 2005;366:572–8.
in prostate carcinoma--long-term results of phase 52. King CR, Presti JC Jr, Gill H, et al. Radiotherapy
III RTOG 85-31. Int J Radiat Oncol Biol Phys. after radical prostatectomy: does transient androgen
2005;61:1285–90. suppression improve outcomes? Int J Radiat Oncol
41. Roach M 3rd, Bae K, Speight J, et al. Short-term Biol Phys. 2004;59:341–7.
neoadjuvant androgen deprivation therapy and 53. Messing EM, Manola J, Yao J, et al. Immediate
external-beam radiotherapy for locally advanced versus deferred androgen deprivation treatment in
prostate cancer: long-term results of RTOG 8610. J patients with node-positive prostate cancer after
Clin Oncol. 2008;26:585–91. radical prostatectomy and pelvic lymphadenectomy.
42. Steigler A, Denham JW, Lamb DS, et al. Risk strati- Lancet Oncol. 2006;7:472–9.
fication after biochemical failure following cura- 54. Thompson IM, Tangen CM, Paradelo J, et al.
tive treatment of locally advanced prostate Cancer: Adjuvant radiotherapy for pathological T3N0M0
data from the TROG 96.01 trial. Prostate Cancer. prostate cancer significantly reduces risk of metasta-
2012;814724:2012. ses and improves survival: long-term follow-up of a
43. Stephenson AJ, Kattan MW, Eastham JA, et al. randomized clinical trial. J Urol. 2009;181:956–62.
Prostate cancer-specific mortality after radical pros- 55. Wiegel T, Bottke D, Steiner U, et al. Phase III post-
tatectomy for patients treated in the prostate-specific operative adjuvant radiotherapy after radical prosta-
antigen era. J Clin Oncol. 2009;27:4300–5. tectomy compared with radical prostatectomy alone
44. Widmark A, Klepp O, Solberg A, et al. Endocrine in pT3 prostate cancer with postoperative undetect-
treatment, with or without radiotherapy, in locally able prostate-specific antigen: ARO 96-02/AUO AP
advanced prostate cancer (SPCG-7/SFUO- 09/95. J Clin Oncol. 2009;27:2924–30.
Genitourinary Cancers 357
56. Zagars GK, Pollack A, von Eschenbach AC. Addition bladder preservation by combined radiation ther-
of radiation therapy to androgen ablation improves apy and chemotherapy: initial results of radia-
outcome for subclinically node-positive prostate tion therapy oncology group 89-03. J Clin Oncol.
cancer. Urology. 2001;58:233–9. 1998;16:3576–83.
57. Swanson GP, Hussey MA, Tangen CM, et al. 69. Tester W, Porter A, Asbell S, et al. Combined
Predominant treatment failure in postprostatectomy modality program with possible organ preservation
patients is local: analysis of patterns of treatment fail- for invasive bladder carcinoma: results of RTOG
ure in SWOG 8794. J Clin Oncol. 2007;25:2225–9. protocol 85-12. Int J Radiat Oncol Biol Phys.
58. Sella T, Schwartz LH, Swindle PW, et al. Suspected 1993;25:783–90.
local recurrence after radical prostatectomy: endorec- 70. James ND, Hussain SA, Hall E, et al. Radiotherapy
tal coil MR imaging. Radiology. 2004;231:379–85. with or without chemotherapy in muscle-invasive
59. Carrie C, Hasbini A, de Laroche G, et al. Salvage bladder cancer. N Engl J Med. 2012;366:1477–88.
radiotherapy with or without short-term hormone 71. Plimack ER, Hoffman-Censits JH, Viterbo R, et al.
therapy for rising prostate-specific antigen concen- Accelerated methotrexate, vinblastine, doxorubi-
tration after radical prostatectomy (GETUG-AFU cin, and cisplatin is safe, effective, and efficient
16): a randomised, multicentre, open-label phase 3 neoadjuvant treatment for muscle-invasive bladder
trial. Lancet Oncol. 2016;17:747–56. cancer: results of a multicenter phase II study with
60. Abdollah F, Karnes RJ, Suardi N, et al. Impact molecular correlates of response and toxicity. J Clin
of adjuvant radiotherapy on survival of patients Oncol. 2014;32:1895–901.
with node-positive prostate cancer. J Clin Oncol. 72. von der Maase H, Sengelov L, Roberts JT, et al.
2014;32:3939–47. Long-term survival results of a randomized trial
61. Stephenson AJ, Shariat SF, Zelefsky MJ, et al. comparing gemcitabine plus cisplatin, with meth-
Salvage radiotherapy for recurrent prostate can- otrexate, vinblastine, doxorubicin, plus cispla-
cer after radical prostatectomy. JAMA. 2004;291: tin in patients with bladder cancer. J Clin Oncol.
1325–32. 2005;23:4602–8.
62. Michalski JM, Lawton C, El Naqa I, et al. 73. Zehnder P, Studer UE, Skinner EC, et al. Super
Development of RTOG consensus guidelines for the extended versus extended pelvic lymph node dis-
definition of the clinical target volume for postopera- section in patients undergoing radical cystectomy
tive conformal radiation therapy for prostate cancer. for bladder cancer: a comparative study. J Urol.
Int J Radiat Oncol Biol Phys. 2010;76:361–8. 2011;186:1261–8.
63. Advanced Bladder Cancer Meta-analysis 74. Mak RH, Hunt D, Shipley WU, et al. Long-term
Collaboration. Neoadjuvant chemotherapy in inva- outcomes in patients with muscle-invasive blad-
sive bladder cancer: a systematic review and meta- der cancer after selective bladder-preserving
analysis. Lancet. 2003;361:1927–34. combined-modality therapy: a pooled analysis of
64. Cole CJ, Pollack A, Zagars GK, et al. Local control of radiation therapy oncology group protocols 8802,
muscle-invasive bladder cancer: preoperative radio- 8903, 9506, 9706, 9906, and 0233. J Clin Oncol.
therapy and cystectomy versus cystectomy alone. Int 2014;32:3801–9.
J Radiat Oncol Biol Phys. 1995;32:331–40. 75. Neoadjuvant chemotherapy in invasive bladder can-
65. Coppin CM, Gospodarowicz MK, James K, et al. cer: update of a systematic review and meta-analysis
Improved local control of invasive bladder cancer by of individual patient data advanced bladder can-
concurrent cisplatin and preoperative or definitive cer (ABC) meta-analysis collaboration. Eur Urol.
radiation. The National Cancer Institute of Canada 2005;48:202–5; discussion 205-6.
clinical trials group. J Clin Oncol. 1996;14:2901–7. 76. Amin MB, Edge SB, Byrd DR, et al. American joint
66. Kaufman DS, Winter KA, Shipley WU, et al. The committee on Cancer (AJCC) Cancer staging man-
initial results in muscle-invading bladder cancer of ual. 8th ed. New York: Springer; 2017.
RTOG 95-06: phase I/II trial of transurethral sur- 77. Escudier B, Bellmunt J, Negrier S, et al. Phase
gery plus radiation therapy with concurrent cispla- III trial of bevacizumab plus interferon alfa-2a
tin and 5-fluorouracil followed by selective bladder in patients with metastatic renal cell carcinoma
preservation or cystectomy depending on the initial (AVOREN): final analysis of overall survival. J Clin
response. Oncologist. 2000;5:471–6. Oncol. 2010;28:2144–50.
67. Kaufman DS, Winter KA, Shipley WU, et al. Phase 78. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in
I-II RTOG study (99-06) of patients with muscle- advanced clear-cell renal-cell carcinoma. N Engl J
invasive bladder cancer undergoing transurethral Med. 2007;356:125–34.
surgery, paclitaxel, cisplatin, and twice-daily radio- 79. Kjaer M, Iversen P, Hvidt V, et al. A randomized trial
therapy followed by selective bladder preservation of postoperative radiotherapy versus observation in
or radical cystectomy and adjuvant chemotherapy. stage II and III renal adenocarcinoma. A study by the
Urology. 2009;73:833–7. Copenhagen renal Cancer study group. Scand J Urol
68. Shipley WU, Winter KA, Kaufman DS, et al. Phase Nephrol. 1987;21:285–9.
III trial of neoadjuvant chemotherapy in patients 80. Motzer RJ, Basch E. Targeted drugs for metastatic
with invasive bladder cancer treated with selective renal cell carcinoma. Lancet. 2007;370:2071–3.
358 N. G. Zaorsky et al.
81. Stein M, Kuten A, Halpern J, et al. The value of post- testicular cancer managed with active surveillance. J
operative irradiation in renal cell cancer. Radiother Clin Oncol. 2015;33:51–7.
Oncol. 1992;24:41–4. 97. Mortensen MS, Lauritsen J, Gundgaard MG, et al.
82. Patard JJ, Leray E, Rioux-Leclercq N, et al. A nationwide cohort study of stage I seminoma
Prognostic value of histologic subtypes in renal cell patients followed on a surveillance program. Eur
carcinoma: a multicenter experience. J Clin Oncol. Urol. 2014;66:1172–8.
2005;23:2763–71. 98. Paly JJ, Efstathiou JA, Hedgire SS, et al. Mapping
83. Kang DE, White RL Jr, Zuger JH, et al. Clinical patterns of nodal metastases in seminoma:
use of fluorodeoxyglucose F 18 positron emission rethinking radiotherapy fields. Radiother Oncol.
tomography for detection of renal cell carcinoma. J 2013;106:64–8.
Urol. 2004;171:1806–9. 99. Shamash J, Powles T, Wilson P, et al. Prognostic
84. Armstrong AJ, George DJ, Halabi S. Serum lactate factors in cytokine-refractory renal cell carcinoma
dehydrogenase predicts for overall survival benefit in treated with irinotecan, Cisplatin, and mitomycin
patients with metastatic renal cell carcinoma treated chemotherapy. J Clin Oncol. 2005;23:1323–5.
with inhibition of mammalian target of rapamycin. J 100. Travis LB, Fossa SD, Schonfeld SJ, et al. Second
Clin Oncol. 2012;30:3402–7. cancers among 40,576 testicular cancer patients:
85. Dalbagni G, Zhang ZF, Lacombe L, et al. Male focus on long-term survivors. J Natl Cancer Inst.
urethral carcinoma: analysis of treatment outcome. 2005;97:1354–65.
Urology. 1999;53:1126–32. 101. Willemse PM, Burggraaf J, Hamdy NA, et al.
86. Eng TY, Naguib M, Galang T, et al. Retrospective Prevalence of the metabolic syndrome and car-
study of the treatment of urethral cancer. Am J Clin diovascular disease risk in chemotherapy-treated
Oncol. 2003;26:558–62. testicular germ cell tumour survivors. Br J Cancer.
87. Heysek RV, Parsons JT, Drylie DM, et al. Carcinoma 2013;109:60–7.
of the male urethra. J Urol. 1985;134:753–5. 102. International Germ Cell Consensus Classification. A
88. Krieg R, Hoffman R. Current management of prognostic factor-based staging system for metastatic
unusual genitourinary cancers. Part 2: urethral can- germ cell cancers. International germ cell Cancer
cer. Oncology (Williston Park). 1520;13:1511–7. collaborative group. J Clin Oncol. 1997;15:594–603.
discussion 1523-4, 1999 103. Bieri S, Rouzaud M, Miralbell R. Seminoma of the
89. Fossa SD, Horwich A, Russell JM, et al. Optimal testis: is scrotal shielding necessary when radiother-
planning target volume for stage I testicular semi- apy is limited to the Para-aortic nodes? Radiother
noma: a Medical Research Council randomized trial. Oncol. 1999;50:349–53.
Medical Research Council testicular tumor working 104. Bruns F, Bremer M, Meyer A, et al. Adjuvant radio-
group. J Clin Oncol. 1999;17:1146. therapy in stage I seminoma: is there a role for fur-
90. Jones WG, Fossa SD, Mead GM, et al. Randomized ther reduction of treatment volume? Acta Oncol.
trial of 30 versus 20 Gy in the adjuvant treatment 2005;44:142–8.
of stage I testicular seminoma: a report on Medical 105. Warde P, Specht L, Horwich A, et al. Prognostic
Research Council trial TE18, European organ- factors for relapse in stage I seminoma managed
isation for the research and treatment of Cancer by surveillance: a pooled analysis. J Clin Oncol.
trial 30942 (ISRCTN18525328). J Clin Oncol. 2002;20:4448–52.
2005;23:1200–8. 106. Daugaard G, Petersen PM, Rorth M. Surveillance in
91. Oliver RT, Mason MD, Mead GM, et al. Radiotherapy stage I testicular cancer. APMIS. 2003;111:76–83.;
versus single-dose carboplatin in adjuvant treatment discussion 83-5.
of stage I seminoma: a randomised trial. Lancet. 107. Aparicio J, Maroto P, del Muro XG, et al. Risk-
2005;366:293–300. adapted treatment in clinical stage I testicular semi-
92. Brydoy M, Fossa SD, Klepp O, et al. Paternity fol- noma: the third Spanish germ cell Cancer group
lowing treatment for testicular cancer. J Natl Cancer study. J Clin Oncol. 2011;29:4677–81.
Inst. 2005;97:1580–8. 108. Bokemeyer C, Nichols CR, Droz JP, et al.
93. Classen J, Schmidberger H, Meisner C, et al. Extragonadal germ cell tumors of the mediastinum
Radiotherapy for stages IIA/B testicular seminoma: and retroperitoneum: results from an international
final report of a prospective multicenter clinical trial. analysis. J Clin Oncol. 2002;20:1864–73.
J Clin Oncol. 2003;21:1101–6. 109. Crook JM, Jezioranski J, Grimard L, et al. Penile
94. Fossa SD, Cvancarova M, Chen L, et al. Adverse brachytherapy: results for 49 patients. Int J Radiat
prognostic factors for testicular cancer-specific sur- Oncol Biol Phys. 2005;62:460–7.
vival: a population-based study of 27,948 patients. J 110. Grabstald H, Kelley CD. Radiation therapy of
Clin Oncol. 2011;29:963–70. penile cancer: six to ten-year follow-up. Urology.
95. Hamilton CR, Horwich A, Bliss JM, et al. 1980;15:575–6.
Gastrointestinal morbidity of adjuvant radiotherapy 111. Krieg RM, Luk KH. Carcinoma of penis. Review of
in stage I malignant teratoma of the testis. Radiother cases treated by surgery and radiation therapy 1960-
Oncol. 1987;10:85–90. 1977. Urology. 1981;18:149–54.
96. Kollmannsberger C, Tandstad T, Bedard PL, et al. 112. Mazeron JJ, Langlois D, Lobo PA, et al. Interstitial
Patterns of relapse in patients with clinical stage I radiation therapy for carcinoma of the penis using
Genitourinary Cancers 359
iridium 192 wires: the Henri Mondor experi- 115. Miralles-Guri C, Bruni L, Cubilla AL, et al. Human
ence (1970-1979). Int J Radiat Oncol Biol Phys. papillomavirus prevalence and type distribution in
1984;10:1891–5. penile carcinoma. J Clin Pathol. 2009;62:870–8.
113. McLean M, Akl AM, Warde P, et al. The results of 116. Zouhair A, Coucke PA, Jeanneret W, et al. Radiation
primary radiation therapy in the management of therapy alone or combined surgery and radiation
squamous cell carcinoma of the penis. Int J Radiat therapy in squamous-cell carcinoma of the penis?
Oncol Biol Phys. 1993;25:623–8. Eur J Cancer. 2001;37:198–203.
114. Sarin R, Norman AR, Steel GG, et al. Treatment 117. Crook J, Grimard L, Tsihlias J, et al. Interstitial
results and prognostic factors in 101 men treated for brachytherapy for penile cancer: an alternative to
squamous carcinoma of the penis. Int J Radiat Oncol amputation. J Urol. 2002;167:506–11.
Biol Phys. 1997;38:713–22.
Gynecologic Cancers
9
Daniel M. Trifiletti, Nicholas G. Zaorsky,
and Surbhi Grover
Abstract
This chapter discusses the general management
of patients with gynecologic system cancers,
with special focus on principles that guide
radiotherapy management. Several key com-
ponents of trimodality care and brachytherapy
are discussed.
D. M. Trifiletti (*)
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
N. G. Zaorsky
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
S. Grover
University of Pennsylvania, Philadelphia, PA, USA
e-mail: Surbhi.Grover@uphs.upenn.edu
FIGO staging is pathological FIGO staging is clinical, only allows CXR, IVP, FIGO staging is clinical FIGO staging is clinical,
barium enema, EUA, cysto, procto If involving or history of cervical/vulvar ca in 5y, pathological, radiographical
stage as primary/recurrence of those sites
D. M. Trifiletti et al.
Gynecologic Cancers 363
GYN LN Anatomy
Interior iliac LNs Obturator LNs External iliac LNs Inguinal LNs
Cranial Bifurcation of common iliac artery 3–5 mm cranial to Bifurcation of External iliac art leaves
obturator canal common iliac bony pelvis to become
artery femoral artery
Caudal Level of obturator canal; or where Obturator canal, Roof of Lower edge of ischial
no space between obturator where obturator acetabulum and tuberosities
internus and midline organs artery exits pelvis sup pubic rami
Post N/A Interior iliac LNs Interior iliac LNs Medial aspect of anterior
pectineal muscle
Ant Obturator internus Ant extent of 7 mm margin ant to Ant edge of sartorius
upper pelvis: 7 mm margin around obturator internus ext. iliac vessels muscle
internal iliac vessels
Lateral Medial edge of muscle or bone Obturator internus Iliopsoas muscle Medial edge iliopsoas
Medial Lower pelvis: mesorectum and Bladder Bladder or 7 mm Lateral aspect of adductor
presacral space margin around long or medial aspect of
upper pelvis: 7 mm around vessel vessel pectineus
364 D. M. Trifiletti et al.
int iliac LNs obturator LNs ext iliac LNs inguinal LNs
cranial bifurcation of 3-5 mm cranial bifurcation of ext iliac art
common iliac to obturator common iliac leaves bony
artery canal artery pelvis to
become
femoral artery
caudal level of obturator roof of lower edge of
obturator canal, where acetabulum ischial
canal; or where obt artery exits and sup pubic tuberosities
no space bw pelvis rami
obturator
internus and
midline organs
post N/A int iliac LNs int iliac LNs medial aspect
Int iliac
of anterior
pectineal
muscle
ant obturator ant extent of 7 mm margin ant edge of
internus obturator ant to ext iliac sartorius
upper pelvis: 7 internus vessels muscle
mm margin
around int iliac
vessels
uterus
uterus
rectum
Ureter
I and
pelvic
sidewall I
IIA IIB IIIB
bladder
IVA II IVA
IIIA
III
Nicholas G Zaorsky, MD
366 D. M. Trifiletti et al.
Stage Pelvic LN+ PA LN+ OS5 ↓LF (21% vs 14%), ↓mets, ↑PFS
IA1 <1 0 98 (65 → 78%). OS borderlines better 80% vs
IA2 6 3 95 71%. Thus, if these factors are present,
IB1 15 10 85 should recommend post-op WPRT.
IB2
–– INT 0107/SWOG 8797/RTOG 9112/GOG
IIA 30 15 73
IIB 30 20 66
109 (Peters, 2000): 243 pts. IA2, B-IIA
III 45 30 40 RadHys, had to have either (+margin, +LN,
IVA 60 40 22 +parametrium [i.e., pT2b]). Then randomize
IVB 9 → RT +/− cis/5FU. EBRT was whole pelvis
49.3/1.7, with 45 Gy to PA LNs if common
–– LN risk (%) by stage: “rule of 15 s” = stage x 15 iliacs +. No brachy. Chemo was cis 70 mg/m2
–– Risk for PA LN involvement = 50% risk of and 5FU 1000 mg/m3 q3w x4c total, 2c con-
pelvic LN involvement current, 2 cycles adj. CRT improved 4-yr. PFS
–– OS5 = rule of 15 s (63 → 80%) and OS (71 → 81%). Identified
the 3 Ps. Use of adjuvant chemo investiga-
tional; it is studied in RTOG 0724.
Surgery: Preserves ovarian function, treatment –– GOG 109 reanalysis (Monk, 2005): tumor
done in less time, does not cause second malig- size <2 cm and 1 LN involved have no OS
nancies (important in young women), takes only benefit of chemo.
up to half of the vagina (preserves sexual func- –– High-risk post-op (Peters RTOG 91–12):
tion), does not cause stenosis, but is more inva- IA2–IIA (with positive margins, positive
sive (risk of bleeding, infection), and some will nodes, and/or parametrial invasion), RT
require PORT (49.3Gy) vs cisplatin (70 mg/m2) + 5FU
(1000 mg/m2) q 3wks x 4 + RT.
Pathology Report Components: Stromal inva- –– RTOG 0724: early-stage, high-risk cervical
sion (and extent), LVSI, tumor size (esp. at 4 cm cut- ca, s/p rad hysterectomy. IA2, IB, and IIA,
off), parametrial invasion, margins, involved nodes who have +LNs, +parametria, or + PA LNs.
Stratify by BT, 3D vs IMRT, WPRT dose (45
Surgery vs RT vs 50.4). Rando to weekly cis (40 mg/
–– Italian, Landoni 1997: 343 pts. IB-IIA → RT m2) + RT vs concurrent weekly
vs RadHyst. RT was 47 Gy (range 40–53 Gy), cis + RT + adjuvant carbo (AUC 5) and pacli-
delivered to WP using AP/PA or 4 fields, then taxel (135 mg/m2). For 3D WPRT, sup is
LDR-BT to a total median dose of 76 Gy. For L4/5. Inferior is bottom of obturator foramen.
surgery, adjuvant RT 50.4 given for stage Lateral is 1–2 cm lateral of true pelvic diam-
>IIA, <3 mm of uninvolved cervical stroma, eter. If common iliac LNs+, then go up to L1/
cut through, LN+. Unchanged OS, DFS, but L2 sup. If PA LNs+, then go up to T11/T12.
morbidity worse in surgery arm (28% vs For lateral fields, ant border is 2 cm ant to the
12%). Limitation: 65% of surgery pts. received CB or 1 cm ant to contoured PA LNs. Post is
adjuvant RT, and tox highest in combined tx. 1–1.5 cm into the CB or >1 cm post to con-
Thus, for IB2 and IIA2, CRT is recommended toured PA LNs.
over surgery.
3D vs IMRT
Post-op RT and Chemo –– RTOG 1203 (TIME-C), Klopp, Yeung. 3D vs
–– GOG 92 (Sedlis 1999/Rotman 2006): 277 IMRT pelvis. Endometrial or cervical cancer
pts. IB2 → RadHys +/− WPRT. Had to have s/p hysterectomy and have indication for
2 of (>4 cm, LVSI, middle/deep stroma adjuvant RT. ASTRO 2016: reduced acute GI/
invasion – note these are simplified). PORT GU tox with IMRT. Sup border is L4–L5.
Gynecologic Cancers 369
–– Inferior border ≥3 cm from vaginal apex or • Lateral: 2 cm lateral to pelvic brim. Do
1 cm above bottom of obturator foramen not block femoral heads.
(whichever is lower) • Extended field:
–– ITV vagina/parametria: • (+) Pelvic lymph nodes = L1/L2.
–– Vaginal cuff = create vagina ITV, sum of vagi- • (+) PA nodes = T11/T12.
nal contours full + empty bladder, include –– Parametrial boost: boost sidewall 10 Gy for
vaginal cuff and at least 3 cm of vagina +parametria.
inferior. –– Sup = bottom of SI joint.
–– PTV45–50.4 = (CTV nodes + ITV vagina/ –– Inferior = same (bottom of obturator fora-
parametria) + 7 mm margin. Total PTV = 1.5– men); 5 cm midline block.
2.0 cm, including the ITV + SM. –– LN boost: LN GTV + 2 cm.
–– IMRT reduces acute and chronic GI toxicity –– Dose: 45 Gy to large fields w/ sequential CD
(per Mundt et al.) and decreases neutropenia, to 54 Gy PM boost, 59.4 Gy LN boost.
improving ability to give concurrent chemo –– First BT should be performed 4–6w after initi-
(per Brixey et al. IJROBP 2002). ating EBRT to allow for tumor shrinkage.
–– Dose: 50.4 Gy
–– Planning: IMRT.
–– On treatment: Daily CBCT and KV Chemo:
imaging. –– Cisplatin 40 mg/m2 weekly, given Mon/Tues,
–– If bladder not full enough, re-instruct patient/ before RT
drink more water.
–– If gas, insert red rubber catheter.
( 2) Definitive Brachytherapy Technique
–– Vaginal brachytherapy (VB) “cuff” boost
–– General anesthesia.
was not permitted on either GOG 92 or GOG
–– Patient in dorsal lithotomy.
109. However, ABS Consensus G uideline
–– EUA, confirm less than 4 cm for good implant
2012 specify it can be considered in patients at
geometry.
high risk of local relapse (e.g., R+).
–– Prep, drape.
–– Foley, inflate with 7 cc of 30% Renografin
Definitive RT Technique solution.
–– +/− gold seeds into cervix at 12 + 6 o’clock.
( 1) External Beam RT –– Sound the cervix. Grab with tenaculum to pro-
Simulation: vide countertraction and advance until slight
–– Full bladder (two cups an hour before) and resistance and note depth.
empty rectum (have a BM before sim). –– Dilate cervix using progressively larger dila-
–– Supine, arms on chest, Alpha Cradle. tors up to 16 French (6 mm). Note: 1 “French”
–– Place two gold seeds at the cervix. or “Fr” = 0.33 mm = 0.013″.
–– CT sim with IV contrast. –– Insert Smit sleeve: Smit length = sound
–– Anatomy based: traditional LN negative depth – 5 mm; suture in place.
WPRT field is 4-field box: –– Insert tandem: curvature = uterus shape;
• Anterior/post: 1 cm ant to pubic sym- length = sound depth – 5 mm.
physis, covering the entire sacrum/ –– Place ovoids over the tandem and slide ovoids
sacral hollow. into the vagina. Use largest ovoids that com-
• Superior: bifurcation of the common fortably fit. Why? Inverse square law: ↑ size, ↓
iliacs (L4/5). vaginal surface dose.
• Inferior: obturator foramen or 3 cm –– Pack radio-opaque gauze posteriorly first,
below most inferior extent of vaginal then anteriorly, using two separate packs.
involvement. –– Ensure bladder is fully drained, clamp Foley,
and inject 30–60 cc of dilute contrast.
372 D. M. Trifiletti et al.
–– Take AP and lateral films to evaluate T&O • Bladder pt: defined w/ 7 cc of contrast in
placement. balloon, perpendicular distance to surface
–– What to look for on orthogonals: of Foley catheter at the center in sup-
• AP: tandem bisects ovoids, tandem not inferior plane of Foley balloon and closest
rotated, phalange close to cervical marker, to applicator.
ovoids high in fornix with 0.5–1 cm spac- • Vaginal pt: at midpoint of lateral surface
ing, no packing above ovoids of ovoid on AP film.
• Lateral: tandem bisects ovoids, midway • Rectal point is most anterior point along
between the sacrum and bladder, at least line that bisects the ovoids.
3 cm away from sacral promontory, suffi- • IIB/parametrial involvement: needs para-
cient anterior, and post packing, Foley bal- metrial boost, 5.4–9 Gy.
loon pulled down • PA LNs: tx only if involved or in pt. who
–– HDR-BT T&O: cannot get chemo. Sup border is T12/L1
• 5.5 Gy x 5, for patients with <4 cm residual (higher than 90–01).
dz. This gives EQD2 >85 Gy
• 6 Gy in 5 fractions, for pts. with >4 cm –– Volume based:
residual dz. This has 35–45 EQD2 • GTVD – macroscopic tumor seen clinically
• 5 Gy x 6 or on MR at diagnosis.
• 7 Gy x 4 • GTVB1, GTVB2 – tumor seen clinically or
–– LDR-BT. Dose rate is 0.4–0.6 Gy/hr. (e.g., on MR at brachytherapy insertion 1, 2,
with Cs-137). Lower dose rates prolong tx etc.
time and have reduced efficacy. Higher dose • HR CTVB1: High-risk CTV includes whole
rates have more late toxicity. cervix and presumed extracervical exten-
–– LDR-BT target dose to point A is 80–90 Gy. sion (volume receiving total Rx dose).
After 45 Gy EBRT, 35–45 Gy prescribed with ~85Gy, D90 = 80–90 Gy.
LDR-BT. • IR CTVB1: Intermediate risk corresponds
to initial macroscopic disease ± margin;
–– ICRU 38 (1985) Guidelines HR CTV + 5–15 mm margin (5 mm AP,
• “Standard loading” of LDR-BT. Tandem 10 mm lateral/craniocaudal). ~60Gy.
and Fletcher-Suit-Delclos ovoid. Using • Primary: PTV identical to CTV.
137Cs. Per ABS, tandem is loaded 15-10- • LN: CTV to PTV expansion for nodal
10 mgRaeq from cephalad to caudad. Tip treatment should be 7 mm.
loaded with more activity to provide ade- • Cumulative EQD2 maximums.
quate coverage in lower uterine segment. • D2cc bladder ≤85 Gy.
Small ovoids then loaded with 10-15 • D2cc rectum ≤75Gy.
mgRaeq. Mini ovoid loaded with doses • D2cc sigmoid ≤75Gy.
5–7.5 mgRaeq because they lack internal –– Total Tx time <7–8 wks, i.e., 56 days. 0.5–
shielding. 1.0% decrement in LC for each day delay after
• Pt A: 2 cm sup and 2 cm lateral from exter- 8w, but no decrease in OS.
nal os (parametria) – 85 Gy EQD2.
• Pt B: 5 cm lateral from midline at the same
level at point A, but in the plane of the
patient/midline, and not from the tandem.
The dose to point B should be ¼ to 1/3 the
dose to point A. Represents obturator
nodes, gets 25% pt. A dose – 55 Gy.
Gynecologic Cancers 373
Parametrial boost
Sidewall +10 Gy for +parametria
Sup = bottom of SI joint;
Inf = same (bottom of obturator
foramen); 5 cm midline block
Conedown to 54 Gy
LN boost
Boost gross nodes to 60 Gy with IMRT
374 D. M. Trifiletti et al.
Chemo Toxicity
–– Cis: myelosuppression, n/v, renal toxicity,
neuropathy, hearing loss.
–– ANC <500 for >7 days or febrile neutropenia:
hold for a week and then repeat CBC.
–– Cr >2.0 mg/dl: hold for 1w. Prevent with
diuresis.
Gynecologic Cancers 375
• T1 Pathology
• T1a (IA) – <½ myometrium –– Simple hyperplasia→ cancer (<2%)
• T1b (IB) – ≥ ½ myometrium –– Complex hyperplasia→ cancer (40%)
• T2 (II) – cervical stroma –– Subtypes:
• T3 • Type I. 80%. Endometrioid histology.
• T3a (IIIA) – serosa/adnexa (SAy it ain’t so) Endometrial hyperplasia. G1–G2.
• T3b (IIIB) – vagina/parametrium (B agina, B Perimenopausal. ER driven. PI3K, PTEN,
arametria) MSI. Less aggressive.
• T4 (IVA) – bladder or rectum • Type II. 20%. G3 Endometrioid. Non-
• N1 (IIIC1) – regional (pelvic/up to common endometrioid histology, e.g., clear cell,
iliac) serous, and carcinosarcoma. Non-ER driven.
• N2 (IIIC2) – para-aortic nodes Postmenopausal. Poor prognosis. p53.
• M1 (IVB) – distant
Workup:
–– H&P: Postmenopausal bleeding, ask about
FIGO 1971 (Inoperable Endometrial
unopposed estrogen: nulliparity, obesity,
Cancer Staging)
tamoxifen, early menarche, late menopause,
I. Confined to corpus
T2DM. Ask about IBD, pelvic RT.
IA. Length of uterine cavity 8 cm or less
–– Labs: CBC, CMP, CA-125 (for trending in
IB. Length of uterine cavity >8 cm
III/IV).
II. Involves corpus and cervix, but no extension
–– US: Endometrial biopsy, imaging/scopes for
beyond the uterus
symptoms. Normal endometrial stripe <5 mm.
III. Extends outside uterus but not outside the
FNR is 10%. If negative and still symptom-
true pelvis
atic, do D/C.
IV. Outside the true pelvis or involves the blad-
–– CXR + CT AP, MRI pelvis if suspected
der or rectum
extrauterine involvement or not going to
IVA. Involves the bladder, rectum, sigmoid,
surgery.
or small bowel
IVB. Distant mets
376 D. M. Trifiletti et al.
–– HNPCC (5%): <50 yo or FH colon or endo- –– Type II MRH: Same as cervix above. Use
metrial → genetic testing. this for endometrial cancers with cervical
• HNPCC = colonic + extracolonic (endo- involvement.
metrial, other GI, ovarian).
• Median age = 45; 80% colon, 60% Surgery +/– LND +/– RT
endometrial.
• Genetic testing = MLH1, MSH2, MSH6, –– MRC ASTEC trial 2009: stage I dz.: first ran-
PMS2. domization: TAH/BSO +/− LND. second ran-
–– + pelvic nodes → 33% chance of + PA nodes. domization: intermed and high-risk pts. get
–– Creasman tables (uses AJCC 6 staging). obs vs adjuvant pelvic RT. No difference in
OS for LND, 5-year OS 80%.
–– NCIC EN.5 Study group.
Risk pelvic LN+ G1 G2 G3
Endometrium only 0 3 0
Inner 3 5 9 G1 G2 G3
Mid 0 9 4
I
Deep 11 19 34
II
Risk PA LN+ G1 G2 G3
(2/3 risk of pelvic LN)
Endometrium only 0 3 0 –– ASTEC/EN.5 combination, 2009: Reported
Inner 1 4 4
results of adjuvant RT. No difference in 5-year
Mid 5 0 0
Deep 6 14 23 OS (84%) or DSS (~90%). WPRT reduced
pelvic and vaginal recurrence, 6% vs 3%;
however, had higher toxicity. Adverse risk fac-
Surgical Procedure tors identified: age >60, LVSI, tumor>2 cm,
–– TAH-BSO + pelvic LND +/− PA sampling (if +cervical gland, ↑grade. These would later be
biopsy proven cervical ext.→ radical hysterec- used in PORTEC-1 and GOG 99.
tomy instead). G1 G2 G3
–– Make midline incision. I
–– Inspect serosal, peritoneal, and diaphragmatic II
surfaces.
–– Peritoneal washing (does not affect staging). Surgery +/− WPRT
–– Omental biopsy for serous, clear cell, and –– GOG-99, Keys 2004:
carcinosarcoma. –– Early-stage endometrial TAH/BSO + LND
–– Perform LND (in the U.S., not Europe) = com- +/−50.4 Gy WPRT. No VCBT. LND was selec-
mon iliacs, external iliacs, internal iliacs, tive for any suspicious LNs. Initially enrolled
obturators + PA sampling. intermediate risk (old stage I with any MI;
–– Sample PA for deeply invasive, high grade, occult II). Enrollment criteria revised while
serous, clear cell, and carcinosarcoma. ongoing to only high-intermediate risk: (1)
–– Perform TAH, bivalve the uterus, and BSO. age >70 + 1 RF; (2) age >50 + 2 RF; (3) any
age + 3 RFs. RFs are G2–3, LVSI, outer 1/3 MI.
Hysterectomy Types –– RT won. 4-yr. LRR 12 → 3% (SS), OS
–– Type I: extrafascial hysterectomy. Spares 86 → 92% (NSS). Mostly benefited high-risk
parametrium; ureters not mobilized; ligate patients. 72% recurrences in VC. Note differ-
UA at the uterus. Takes 1–2 mm of vaginal ence for “high-intermed” risk for GOG-99 vs
cuff. PORTEC-1.
–– Omental biopsy for uterine papillary serous
carcinoma (UPSC) clear cell carcinoma G1 G2 G3
(CCC). Use type I RH for most endometrial I
II
cancers.
Gynecologic Cancers 377
Second Malignancy
–– PORTEC 1 + 2 + Dutch TME pooled (2015):
n = 2554. No increased risk of malignancy
from RT: 26% in 15 years for both RT and
non-RT arms.
Gynecologic Cancers 379
Treatment Paradigm
–– All patients go for surgery if able: TAH/BSO, peritoneal inspection, fluid cytology, +/− LND (usu-
ally for grade 2/3), rad hysterectomy if cervical involvement.
G1 G2 G3
IA None None vs VCBT None vs VCBT
HR: ± VCBT HR: ± VCBT HR: ± WPRT
IB VCBT VCBT VCBT and/or WPRT
HR: ± WPRT HR: ± WPRT
II VCBT and/or WPRT VCBT and/or WPRT WPRT ± VCBT ± Chemo Chemo
IIIA WPRT ± VCBT + Chemo Chemo
*Chemo is carbo AUC5 , paclitaxel 175 x5c
IIIB Chemo and/or WPRT + VCBT
IIIC Chemo and/or WPRT ± VCBT
IV Debulk to R0 -1. Then Chemo ± WPRT ± VCBT
Nomogram to predict local (A) and distant (B) recurrence (Creutzberg, 2015)
Score
-15 -10 -5 0 5 10 15
Age (years)
35 40 45 50 55 60 65 70 75 80 85 90 95
Radiotherapy
EBRT VBT none
FIGO grade
G1 G2 G3
Myometrial invasion depth
< 50% > 50%
Vascular invasion
no yes
Sum of scores
-20 -10 0 10 20 30 40
Locoreginal relapse
within 5 years [%] 1 5 10 20 30 40 50 60 70 80 90
95
Score
-15 -10 -5 0 5 10 15
Age (years)
35 40 45 50 55 60 65 70 75 80 85 90 95
FIGO grade
G1 G2 G3
Vascular invasion
no yes
Sum of scores
-20 -10 0 10 20 30
Distance relapse
within 5 years [%] 1 5 10 20 30 40 50 60 70
380 D. M. Trifiletti et al.
Outcomes (OS5)
–– I – 90
–– II – 70
–– III – 50
Gynecologic Cancers 383
Adjuvant
LNM WPRT
Tumor Staging Symptoms Behavior incidence Surg, LND indications
Carcinosarcoma Treat and Vaginal Epithelial component 20–38 PLND, (Similar to
(MMMT) stage like bleeding dictates prognosis and PA-LND endometrial):
Malignant mixed regular treatment, not usually >60yo
mesodermal tumor/ high-grade sarcomatous recommended >50% MI
malignant mixed endometrial component Most have II
Mullerian tumor cancer (FIGO serous or G3 LN+
staging) endometrioid R+
histology and behave
as their pure uterine
cancer counterpart
Leiomyosarcoma IA/B: 5 cm Uterine 7–9 PLND, R+
(LMS) cutoff fibroid like PA-LND
Endometrial stromal IIA: adnexa symptoms Grade most important 10 usually not R+
sarcoma (ESS) IIB: pelvic (vs LMS or MMMT) recommended
tissues G1: hormone unless
IIIA: 1 sensitive, indolent extrauterine dz
abdominal courseG3: aggressive,
tissue site similar to LMS and
IIIB: >1 site MMMT Similar to
IVA: bladder undifferentiated
or rectum sarcoma
IV: DM
Adenosarcoma Like AJCC R+
6th edition
FIGO staging
384 D. M. Trifiletti et al.
T1a T1b T2 T3
≤2cm + >2cm 1/3 2/3
≤1mm or Urethra, Urethra,
DOI >1mm Vag; Vag; Anus;
DOI Anus other organ
II
N0 IA IB
(UVA)
1-2,
N1a
<5mm IIIA
N1b 1, ≥5mm IVA
N2a 3+, <5mm
IIIB
N2b 2+≥5mm
N2c ECE IIIC
N3 ulcer IVA
M1 IVB
386 D. M. Trifiletti et al.
Management Technique
IA: Simulation:
–– Radical vulvectomy vs WLE (if well • Full bladder and empty rectum
lateralized). • Supine, frog leg, Alpha Cradle
–– No need to address nodes. • Wire entire vulva, place additional fiducial
markers around gross disease
IB: • CT sim + IV contrast
–– Radical vulvectomy vs WLE (if well lateral-
ized) + SLNBx, if (+) SLN → b/l IFLND. Contours:
–– Only SLNBx if primary ≤4 cm, dual tracer • GTV = Primary/gross residual
(dye + radiotracer). If primary >4 cm need • CTV_Vulva = GTV + 1 cm and encompassing
upfront bilateral IFLND. at least entire vulva
–– In GROINSS-V, if SLN (−), only 3% LR in –– Anterior vulva includes at least 2 cm of the
untreated IFLN basin. urethra to bladder neck.
–– Complications, SLNBx vs IFLND: ↓lymph- –– Posterior vulva includes perineum between
edema (2% vs 25%), ↓wound breakdown/ posterior fourchette and anal verge.
cellulitis –– Vaginal extension: gross dz. + 3 cm.
Consider entire vagina with creation of
II (spread to lower 1/3 urethra, vagina, anus): vaginal ITV (w/ full and empty blad-
der) + presacral LN.
–– Radical vulvectomy + unilateral or bilateral –– Anal canal includes entire mesorectum and
inguinal LND +/− post-op RT. perirectal LN.
• PTV_Vulva = CTV_Vulva +1 cm
III-IVa – Radical vulvectomy+ bilateral ILND • CTV_LN = external iliacs, internal iliacs,
→Chemo RT obturators, inguinals (+ presacral LN for vagi-
Post-op CRT nal extension and + mesorectum/perirectal for
–– 50.4 Gy to CTV (vulva and elective LN) anal canal). If posterior vaginal, then presacral
–– 59.4 Gy to +SM or + LN nodes. In certain cases, lower common iliacs
–– 64.8 Gy to GRD (L5/S1)
–– Weekly cis (40 mg/m2) • Inguinal LN contours:
Unresectable = definitive CRT
–– 50.4 Gy to CTV (vulva and elective LN) ASTRO consensus for Radial margin around
inguinal LN coverage femoral vessels (mm)
–– 59.4 Gy to +SM or + LN Anteromedial ≥35
–– 64.8 Gy to GRD Anterior ≥23
–– Weekly cis (40 mg/m2) Anterolateral ≥25
–– GOG 205: cis + RT (57.6 Gy) → incisional Medial ≥22
biopsy to assess response, if (+) → radical Posterior 0
vulvectomy Lateral 0
–– cCR = 65%, pCR = 50%
Gynecologic Cancers 389
Treatment Technique
–– VAIN 1: observation –– Sim frog leg.
–– VAIN 2/3: WLE, laser, 5FU, Vcuff. –– Vaginal marker.
–– I: surgery or VCBT to 6 Gy x 5. –– Fields: pelvic, R/L inguinal.
• If >5 mm deep, >2 cm or G3: add LND or –– Sup border at L5–S1. Inferior is 2 cm below
WPRT. vag introitus. Lateral is 2 cm lateral to pelvic
brim.
–– II: –– If disease involves the rectovaginal septum
–– (Option 1) WPRT to 45 Gy + BT to 75 Gy (e.g., near pouch of Douglas) or bladder, then
(6 Gy x 3 HDR) + cisplatin-based chemother- include the perirectal LNs, do not BT because
apy (40 mg/m2 weekly) of high risk of fistula, and use IMRT boost.
–– Same simulation/fields as vulva (L5/S1) –– Vagina Tolerance
–– If involves lower 1/3 vagina, include inguinal • Upper vagina = 120 Gy
LN. • Middle vagina = 100 Gy
–– Reevaluate patient at 40 Gy for whether • Lower vagina = 80 Gy
<5 mm or ≥ 5 mm.
–– BT when possible: ABS/UPMC/MDACC Recommendations
• IC BT is for superficial lesions <0.5 cm Based on Location:
from surface. Apex: and <0.5 cm, use ICRT. If >0.5 cm, EBRT
• IS BT is for deep tumors >0.5 cm from or interstitial.
surface. Mid-vagina: interstitial therapy for anterior or
• Apex = template. Lateral and lateral. If posterior or massive, use EBRT boost.
distal = freehand. Distal: confined lesions gevt interstitial. Massive
tumors need EBRT boost.
(Option 2): Radical vulvovaginectomy + pel- Who should get vaginal cylinder alone?
vic LND + IFLND for distal 1/3 Superficial (<0.5 cm thickness).
Who should get interstitial brachy? Apical
tumor, well defined, mobile, and >0.5 cm thick.
Who gets chemo? Stage III+, high-risk dz.
392 D. M. Trifiletti et al.
tme, portec-1, and portec-2 trials. J Clin Oncol. carcinoma of the vulva: a gynecologic oncology
2015;33:1640–6. group study. Gynecol Oncol. 2012;124:529–33.
26. Blake P, Swart AM, Orton J, et al. Adjuvant external 38. Moore DH, Thomas GM, Montana GS, et al.
beam radiotherapy in the treatment of endometrial Preoperative chemoradiation for advanced vulvar
cancer (Mrc Astec and Ncic Ctg En.5 Randomised cancer: a phase II study of the gynecologic oncology
Trials): pooled trial results, systematic review, and group. Int J Radiat Oncol Biol Phys. 1998;42:79–85.
meta-analysis. Lancet. 2009;373:137–46. 39. Stehman FB, Bundy BN, Thomas G, et al. Groin dis-
27. Scholten AN, van Putten WL, Beerman H, et al.
section versus groin radiation in carcinoma of the
Postoperative radiotherapy for stage 1 endometrial vulva: a gynecologic oncology group study. Int J
carcinoma: long-term outcome of the randomized Radiat Oncol Biol Phys. 1992;24:389–96.
portec trial with central pathology review. Int J Radiat 40. Coleman RL, Ali S, Levenback CF, et al. Is bilateral
Oncol Biol Phys. 2005;63:834–8. lymphadenectomy for midline squamous carcinoma
28. Barillot I, Tavernier E, Peignaux K, et al. Impact of the vulva always necessary? an analysis from gyne-
of post operative intensity modulated radiotherapy cologic oncology group (Gog) 173. Gynecol Oncol.
on acute gastro-intestinal toxicity for patients with 2013;128:155–9.
endometrial cancer: results of the phase II rtcmien- 41. Viswanathan AN, Pinto AP, Schultz D, et al.
dometre french multicentre trial. Radiother Oncol. Relationship of margin status and radiation dose
2014;111:138–43. to recurrence in post-operative vulvar carcinoma.
29. Creutzberg CL, van Putten WL, Koper PC, et al.
Gynecol Oncol. 2013;130:545–9.
Survival after relapse in patients with endometrial 42. Stehman FB, Bundy BN, Ball H, et al. Sites of failure
cancer: results from a randomized trial. Gynecol and times to failure in carcinoma of the vulva treated
Oncol. 2003;89:201–9. conservatively: a gynecologic oncology group study.
30. Schwarz JK, Beriwal S, Esthappan J, et al. Consensus Am J Obstet Gynecol. 1996;174:1128–32; discussion
statement for brachytherapy for the treatment of med- 1132–1123.
ically inoperable endometrial cancer. Brachytherapy. 43. Gill BS, Bernard ME, Lin JF, et al. Impact of adjuvant
2015;14:587–99. chemotherapy with radiation for node-positive vulvar
31. Giuntoli RL 2nd, Metzinger DS, DiMarco CS, et al. cancer: a national cancer data base (Ncdb) analysis.
Retrospective review of 208 patients with leiomyo- Gynecol Oncol. 2015;137:365–72.
sarcoma of the uterus: prognostic indicators, surgical 44. Gaffney DK, King B, Viswanathan AN, et al.
Management, and adjuvant therapy. Gynecol Oncol. Consensus recommendations for radiation therapy
2003;89:460–9. contouring and treatment of vulvar carcinoma. Int J
32. Reed NS, Mangioni C, Malmstrom H, et al. Phase Radiat Oncol Biol Phys. 2016;95:1191–200.
III randomised study to evaluate the role of adjuvant 45. Montana GS, Thomas GM, Moore DH, et al.
pelvic radiotherapy in the treatment of uterine sar- Preoperative chemo-radiation for carcinoma of
comas stages I and II: an european organisation for the vulva with N2/N3 nodes: a gynecologic oncol-
research and treatment of cancer gynaecological can- ogy group study. Int J Radiat Oncol Biol Phys.
cer group STUDY (Protocol 55874). Eur J Cancer. 2000;48:1007–13.
2008;44:808–18. 46. de Crevoisier R, Sanfilippo N, Gerbaulet A, et al.
33. Wolfson AH, Brady MF, Rocereto T, et al. A
Exclusive radiotherapy for primary squamous
Gynecologic oncology group randomized phase cell carcinoma of the vagina. Radiother Oncol.
III trial of whole abdominal irradiation (Wai) Vs. 2007;85:362–70.
cisplatin-ifosfamide and mesna (Cim) as post-surgical 47. Frank SJ, Jhingran A, Levenback C, et al. Definitive
therapy in stage I-IV carcinosarcoma (Cs) of the radiation therapy for squamous cell carcinoma of
uterus. Gynecol Oncol. 2007;107:177–85. the vagina. Int J Radiat Oncol Biol Phys. 2005;62:
34. Wright JD, Seshan VE, Shah M, et al. The role of 138–47.
radiation in improving survival for early-stage car- 48. Lian J, Dundas G, Carlone M, et al. Twenty-year
cinosarcoma and leiomyosarcoma. Am J Obstet review of radiotherapy for vaginal cancer: an institu-
Gynecol. 2008;199:536 e531–8. tional experience. Gynecol Oncol. 2008;111:298–306.
3 5. Katz A, Eifel PJ, Jhingran A, et al. The role of 49. Hacker NF, Eifel PJ, van der Velden J. Cancer of
radiation therapy in preventing regional recur- the vagina. Int J Gynaecol Obstet. 2015;131(Suppl
rences of invasive squamous cell carcinoma of 2):S84–7.
the vulva. Int J Radiat Oncol Biol Phys. 2003;57: 50. McGuire WP, Hoskins WJ, Brady MF, et al.
409–18. Cyclophosphamide and cisplatin versus paclitaxel
36. Kunos C, Simpkins F, Gibbons H, et al. Radiation and cisplatin: a phase III randomized trial in patients
therapy compared with pelvic node resection for with suboptimal stage iii/iv ovarian cancer (from
node-positive vulvar cancer: a randomized controlled the gynecologic oncology group). Semin Oncol.
trial. Obstet Gynecol. 2009;114:537–46. 1996;23:40–7.
37. Moore DH, Ali S, Koh WJ, et al. A phase II trial of 51. Ozols RF, Bundy BN, Greer BE, et al. Phase III trial
radiation therapy and weekly cisplatin chemotherapy of carboplatin and paclitaxel compared with cisplatin
for the treatment of locally-advanced squamous cell and paclitaxel in patients with optimally resected
Gynecologic Cancers 395
stage iii ovarian cancer: a gynecologic oncology logic oncology group study. J Clin Oncol. 2007;25:
group study. J Clin Oncol. 2003;21:3194–200. 437–43.
52. Smith JP, Rutledge FN, Delclos L. Postoperative
54. Domchek SM, Friebel TM, Singer CF, et al.
treatment of early cancer of the ovary: a random trial Association of risk-reducing surgery in Brca1 or
between postoperative irradiation and chemotherapy. Brca2 mutation carriers with cancer risk and mortal-
Natl Cancer Inst Monogr. 1975;42:149–53. ity. JAMA. 2010;304:967–75.
53. Wenzel LB, Huang HQ, Armstrong DK, et al.
55. Finch AP, Lubinski J, Moller P, et al. Impact of
Health-related quality of life during and after oophorectomy on cancer incidence and mortality in
intraperitoneal versus intravenous chemotherapy women with a Brca1 or Brca2 mutation. J Clin Oncol.
for optimally debulked ovarian cancer: a gyneco- 2014;32:1547–53.
Hematologic Cancers
10
Nicholas G. Zaorsky, Daniel M. Trifiletti,
and Jennifer L. Peterson
Abstract
This chapter discusses the general manage-
ment of patients with hematologic cancers,
with special focus on principles that guide
radiotherapy management. Several key com-
ponents of trimodality care and radiotherapy
field design are discussed.
N. G. Zaorsky (*)
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
D. M. Trifiletti · J. L. Peterson
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu;
peterson.jennifer2@mayo.edu
R hilum
L hilum
Age
ESR and B
Mediastinal
#Nodal sites
Ann Arbor EORTC
R cervical, supraclav
R infraclav, subpec
R axilla
L cervical, supraclav
L infraclav, subpec
L axilla
Mediastinum
≥50
German
A: >50, B: >30
>3
NCCN
A: >50, B: >30
>2
N/A
≥50 or B
MTR > .35 MMR > .33MMR > .33
>3
#Nodal sites
E lesion any
Bulky >10 cm
–– Burkitt’s lymphoma
–– Lymphoblastic
–– Immunocompromised
–– BM+
–– Parameningeal
–– Testicular relapse
400 N. G. Zaorsky et al.
–– IFRT (do not use this anymore!) – 1995+; –– INRT: subtype of ISRT where pre-
20–30 Gy; site of clinically involved LN, chemo imaging available, no margin for
Kaplan/Rye group (per below); LN grouping imaging limitations; upfront PET-CT
not clearly defined; depends on bony anatomy; mandatory in tx position; typically,
2D. SOC since HD8. pregnant women not eligible because
–– Cervical/supraclav: extends from base of CT and PET are avoided.
the skull to clavicles. Patient positioned in • INRT alone acceptable for NLPHL,
mark. Oral cavity block placed if tumor though CTV is more generous
coverage not compromised. (~3–5 cm along vessel).
–– Axilla: C5/6, tip of scapula or 2 cm below • INRT is the SOC for early-stage fav
LNs, ipsi transverse process, flash axilla. HL per ILROG, if pre-treatment
–– Mediastinum: imaging in the treatment position is
• Upper border should be at C5–6. If available.
sclav is involved, then upper border –– INRT contours (Specht, IJROBP, 2014)
should be top of the larynx. (A) Prechemotherapy GTVCT on preche-
• Lower border is 5 cm below carina or motherapy CT scan.
2 cm below inf extent of pre-chemo dz. (B) Prechemotherapy GTVPET on preche-
• Lat borders are 1.5 cm margin on post- motherapy CT scan.
chemo volume. (C) Postchemotherapy GTVCT on post-
–– PA field: chemotherapy CT scan.
• Top of T11 or 2 cm above pre-chemo (D) Postchemotherapy GTVPET on post-
volume to aortic bifurcation (bottom chemotherapy CT scan.
L4) or 2 cm below pre-chemo volume. (E) Clinical target volume, created by mod-
• Encompass lateral transverse processed ifying GTVCT and GTVPET, on the post-
of L-spine and 2 cm from post-chemo chemotherapy CT scan.
volume. –– Take into account tumor shrinkage, ana-
–– Pelvic field: tomic changes.
• Includes external iliac, inguinal, femoral –– CTV encompasses all initial lymphoma
nodes, middle SI joint to 5 cm below lesser volume while still respecting normal struc-
trochanter; lateral border includes greater tures (e.g., lungs, CW, muscles).
trochanter, medial border at obturator fora- –– ITV – CTV + margin, create by using 4D
men, including 2 cm beyond involved CT or fluoro to account for motion.
nodes –– ITV + setup error margin = PTV. Use
• ISRT: 3D; 2008+; pre-chemo GTV deter- DIBH if involving mediastinum.
mines CTV (carve out bone, lung); becom- –– Commence 3–4w after completing
ing the SOC. chemo,
–– Supine, head extended, alpha cradle,
arms akimbo.
–– Pure ISRT: routine in the USA; optimal
pre-tx imaging not available. Not pos-
sible to reduce CTV to same extent as
INRT. GTV + ~1 cm = CTV. CTV + 1
cm = PTV.
Hematologic Cancers 401
chemo if CR.
–– Laskar, Tata Memorial, 2004. Mostly MC HL, –– 1: No uptake
a better prognosis subtype. ABVD x6c. If CR, –– 2: Uptake ≤ mediastinum
rando to obs vs IFRT 21 Gy + 10 Gy boost to –– 3: Mediastinum < uptake ≤ liver
X. EFS 88% vs 76% favoring RT. OS 100% vs –– 4: ↑Moderately compared to liver
89% favoring RT. 15% pts. did receive EFRT –– 5a: ↑Markedly compared to liver
and 115 got inverted Y. 50% pts. <15 yo and –– 5b: New FDG avid site
half stages I–II. –– 1–2 are neg; 4–5 are positive; 3 can be either,
–– German, HD15, Engert, 2012. Eval different depending on goal of study (e.g., esc vs de-
chemo. Only residual dz. >2.5 cm and PET+ esc). Usually negative.
txd w/ 30 Gy IFRT. –– Restaging PET/CT is 2 weeks after day 15 of
last cycle of ABVD. If Deauville 4 on interim
↓Field Size, ↓Dose, ↓Chemo PET-CT, the ast PET-CT is 6–8 wk. after
–– GPMC, German HD8, Milan, EORTC H8U chemo or 8–12 wk. after RT.
all ↓ FS. –– Do not perform 7-10d after chemo to avoid
–– German HD8, Engert, 2010. Early unfav. false positive from inflammation.
COPPx2 and ABVDx2, then rando to 30 Gy –– Interim PET is + in 14% F and 24% UF
EFRT vs 30 Gy IFRT. X dz. boost +10 Gy. 5-y patients.
Hematologic Cancers 407
Protons: best for mediastinum, spleen, PA –– Lhermitte’s: 20% in HL, likely from chemo.
LNs. No correlation w/ dose. Should resolve w/
time.
Constraints (per ILROG): –– Adriamycin: cardiomyopathy (limit is 450 mg
–– Lung: V20 <20%; mean lung <12 Gy. w/ RT, 550 w/o RT).
–– Heart: mean <15 Gy. –– Bleomycin: pulmonary fibrosis.
–– Bone growth affected at 8 Gy. –– Vinblastine: neuropathy and hair loss.
–– Breast tissue growth affected at 5–10 Gy.
–– Thyroid: >15 Gy assoc. w/ 30% risk Late Effects Monitoring (After 5 Years)
insufficiency. –– Optimize cardiovascular health, baseline
–– TD 5/5: echo, and stress test q10y, especially if cardiac
• Testes: 2 Gy (permanent sterility), 0.5 Gy RT.
(temporary azoospermia). –– Carotid ultrasound at 10 y if neck RT.
• Oocytes. Sterilization by 2 Gy. Failure by –– If splenic RT → pneumo, meningo, flu vac-
6–10 Gy (single fx), 12–15 Gy fractionated cines q5y.
in age <40. –– Chest XR/CT qyr if at risk for lung ca.
–– Female age >25: Start breast mammo 10 years
Follow-Up after treatment or at 40, whichever earlier if
–– Need one PET-CT to confirm response after chest or axillary RT.Get annual breast MRI at
RT; otherwise, no role for routine PET-CT in age 40 or 10 yrs. after RT.
follow-up. • Cumulative risk of breast cancer in 30y
–– q3 mo x 2y, q6 mo x3y, then annually with after treatment for HL 19% (~0.7% per
H&P, labs including TSH. year) and 26% for women <21 yo.
–– CXR or CT q6 mo x5y (alternate PET-CT
with CT yearly). Outcomes
–– Early stage favorable/unfavorable and
Toxicity NLPHL: 10-yr. OS = 90% – 10-yr. EFS = 85%.
–– Dermatitis, post scalp hair loss, xerostomia, –– Advanced stage:
dental caries (fluoride x 2 years after RT), • 10-yr. OS = 80% – 10-yr. EFS = 75%
esophagitis, thyroid dysfunction, Lhermitte’s,
herpes zoster, pneumonitis, and pericarditis.
Hematologic Cancers 409
Intermediate-High-Grade Lymphoma
NHL/DLBCL [51–55]
Revised Age-adjusted IPI for pts <60 yo, Schipp, 1993
International Prognostic Index (IPI) IPI (ritux (“APLES w/o the vowels”)
(APLES or LAKES) adds 10%) ECOG ≥2 Risk group #
Age >60 Risk group # 5 yr. OS 5 yr. OS LDH > ULN Low risk 0–1
ECOG ≥2/ Low risk 0–1 73% 94% Stage III/IV Int risk 2
KPS <70 Low/int risk 2 51% 79% Int/high risk 3
LDH >ULN Int/high risk 3 43% 55%
>1 extranodal High risk 4–5 26%
Stage III/IV
Isolated
The 4 Any local local Distant
RCTs Patients Arms Outcomes Criticisms relapse relapse relapse
GELA >60 y/o, CHOP 4c 5 yr. EFS and OS RT started >5w after 63% vs 47% vs 53% vs
LNH93–4 low risk, +/− 40 Gy (~70%) same chemo in 50% pts. 34% 21% 79%
(elderly) age- IFRT Only 88% pts. in Criticized
Bonnet, adjusted CMT arm got RT. In for this hi
2007 IPI 0, field recurrence 34%, value
stages which is >10% than
I–II. 8% the other GELA study
X.56% E and >15% than the
two US studies. This
was a very specific
population
GELA ≤ 60 yo, AVCBP x3c + AVCBP w/o RT won. Effect of RT difficult 62% 41% vs 59% vs
LNH93–1 low risk consolidation 5y EFS 82% vs 74%. to interpret given the (AVCBP) 23% 77%
(young) (IPI 0) (e.g. MTX, OS 90% vs 81% different chemos + vs 28%
Reyes, stages leucovorin, consolidation.
2005 I–II (E or etoposide, ifos, AVCBP is toxic
X cytarabine) vs (150% intensity of
allowed) CHOP x3c + CHOP; needs
40 Gy IFRT hospitalization, hi rate
of AML, lung ca)
ECOG I–IIE, int CHOP 8c For the pts. w/ CR, RT More X pts. on RT 48% 52%
1484Horn- grade If CR, then won over obs. Primary arm; not powered for (15/31) (16/31)
ing, 2004 NHL 30 Gy vs obsIf endpoint was DFS. OS vs 17% vs 82%
PR, 40 Gy for 6 yr. DFS 56 → 73%, (3/17) (14/17)
all pts OS numerically better
but NSS. Conversion
from PR → CR did
not influence DFS
SWOG I–IIE int CHOP 8c vs 5 yr. results favored (1) Differing # 0% with Relapses not
8736, grade CHOP CRT, but 8 yr. chemos; (2) Stage RT reported in
Miller, NHL 3c + 40–55 overlapped. In-field adjusted IPI = 0 in primary
1998 IFRT LRR 0%. Inc. rates of 90% of pts., low risk publication or
cardio tox in 8c of of failure. (3) Relapses abstracts.
chemo vs 3 c + IFRT. not reported in Note that
As advanced stage is primary pub or DLBCL I-II IPI
factor in original IPI, abstracts. Anecdotal 2–4 pts. felt to
investigators modified reports state excess drive the late
index to make late relapses out of out-of-field
“stage-adjusted IPI” field recurrences;
to exclude >1 E sites thus, felt to
and stages III– benefit from
IV. IFRT felt to more chemo
compensate for
reduction in chemo
Hematologic Cancers 413
Treatment –– RT technique:
• Tape penis up.
DLBCL, General • Measure scrotum separation.
–– I-II (non-X, i.e., <7.5 cm): • Use en face electrons (flash lat and inf.);
• For IPI 0–1, RCHOP x 3c preferred. Then 9–12 MeV.
PET-CT. Then ISRT 30–36 Gy. Boost • No bolus.
FDG-avid residual dz. to 40–46 Gy. • Want ≥80% of dose from 0.5 cm depth to
• Reduces risk of relapse by 50–60%. 0.5 cm from posterior.
• Alternative (cat 2b) is R-CHOP x 6c and no
further treatment. DLBCL of Bone (See Separate Section)
• For IPI 2–4, RCHOP x6c. –– R-CHOP x three cycles + RT 45 G to GTV
• Then ISRT 30–36 Gy. +2–3 cmy. Cover larger volume of bone.
• If unable to tolerate chemo, then 36–45 Gy –– Using less chemo but more RT as local
ISRT (depending on response to chemo). therapy.
• Boost FDG-avid residual dz. after chemo
to 40–46 Gy. DLBCL of the Breast
–– I-II (X, i.e., >7.5 cm): RCHOP x6c +/− ISRT –– R-CHOP x six cycles + RT 36 Gy to whole
30–36 Gy. X dz. (RICOVER-noRTh) breast (use tangents).
–– Using more chemo but less RT to minimize
–– III/IV: dose to breast.
• If high IPI, clinical trial for high-dose
chemo with autologous stem cell rescue. Mantle Cell
• R-CHOP x 4 → restage w/ PET. –– Most often advanced stage. Median OS
• CR→ R-CHOP x 2 → observe. Consider 3–4 years.
ISRT only to initial bulk (36 Gy), per –– I–II: chemo+RT.
German group. • RT for stages I–II: 30–36 Gy at 1.5 Gy/fx.
• PR → R-CHOP x 2→ restage w/ PET. –– III+: chemo.
• If PET negative: ISRT only to initial bulk –– Palliation: 2 Gy x 2 fx (“Boom boom”), can
(36 Gy). repeat PRN.
• If PET positive: refractory, go to
transplant. Primary Cutaneous B-Cell Lymphoma,
Leg Type
–– Aggressive histology (e.g., double/triple hit, –– Complete skin exam; labs w/ LDH, HBV, CT
Burkitt’s lymphoma): CAP and or PET, BM bx.
• da-R-EPOCH or R-CHOP x 6c, then –– Leg type has poor prognosis.
PET-CT, then 30 Gy ISRT for PR. –– NCCN: R-CHOP and local RT 30 Gy.
–– Palliation: 2 Gy × 2.
Gastric DLBCL
–– Same treatment as above [R-CHOP x 3 → Which DLBCLs Need CSF
ISRT (30 Gy)]. Chemoprophylaxis?
–– Testicular, paranasal sinus, epidural, BM
Testicular DLBCL involvement.
–– Orchiectomy→ R-CHOP x 6 w/ IT-MTX → –– Give 4–8c of IT MTX.
scrotal RT to 30 Gy (high contralateral relapse
rate w/o RT).
Hematologic Cancers 415
Workup
Same as NHL: LDH, CBC, urate, PET CT, BM
biopsy, get IPI, HBV, echo or MUGA, pregnancy
testing.
Studies
–– Dunleavy, 2013: NCI regimen of
DA-EPOCH-R. n = 51. No RT. Untreated. At
5-y, EFS is 93%, OS 97%. CR rate is 96%.
• These outcomes are unprecedented in a
phase II study.
Treatment
–– Tx w/ DA-EPOCH-R x 6c (Dunleavy, 2013).
Chemo requires inpt stay for 1 w.
–– Then PET-CT.
–– No RT unless persistent focal disease.
–– Less preferred: R-CHOP x6 w/ ISRT 30 Gy;
or R-CHOP x4, then ICE x3c, +/− ISRT
30 Gy.
416 N. G. Zaorsky et al.
Overview
–– 15,000 cases/yr. in UA.
–– Workup: H&P, CBC w/ diff, CMP, LDH, Ca/albumin, B2micro (tells how bulky tumor is), SPEP
with immunofixation, UPEP with immunofixation, 24 hr urine for total protein, serum-free light-
chain assay, skeletal survey (not bone scan) biopsy lesion AND unilateral BMbx; bone marrow
MRI upstages 30%.
–– PET-CT can be helpful but no bone scan (lytic lesions).
–– M-protein: a “monoclonal protein,” aka paraprotein, aka M-component. It is not IgM.
• Note: Compared to MM, “solitary” plasmacytoma RT dose different; plasmacytoma (bone met)
tx in setting of MM is palliative; SPs are not staged.
• SBP 40% more common than SEP.
Dx criteria Progress
for SPs, all Location to MM
Tumor required (usually) LN+ at 10y DDx Treatment RT technique LC Follow-up
SEP SEP/SBP HN 30– 25% NHL, RT or Mass + primary LC = q3–6 m:
lesion by 40% MALToma surgery LNs. 88–100%. CBC
skeletal Head and neck: Tsang, CMP
survey consider treating 2001: SPEP
(negative first echelon LN mostly UPEP
on bone GTV + 2–3 cm SBP, some FLC
scan!) to 50.4 Gy, ENI SEP. 8y PRN:
Plasmacy- to 40 Gy. LC 83%, LDH
toma by Keep GTV OS 65%, B2 micro
biopsy, ≥45 Gy DFS 44% BM Bx
SBP <5% Axial Rare 75% Mets, MM RT or Bone GTV + Body MRI, CT,
plasma skeleton surgery 2-3 cm PET
cells Spine: whole VB Skel surv
No Standard tx is (or q1y)
end-organ RT, 40–50 Gy.
damage No ENI. In
(CRAB - cases of surgical
calcium, resection,
renal, adjuvant RT still
anemia, recommended
bone)
SBP/SEP + +/- - - -
MGUS +/- + - - -
Smoldering MM + + + + -
MM + + + + +
Hematologic Cancers 423
Durie Salmon
- criteria staging for MM
Hgb (>12 or 13.5 is WNL) No end organ damage, so all >10 g/dL <8.5 g/dL
Serum Ca (8.5-10.2 is WNL) should be WNL ≤12 mg/dL >12 mg/dL
Bone xray WNL, or SBP only Advanced lesions
M protein production Low High
IgG <5 g/dL IgG >7 g/dL
IgA <3 g/dL IgA >5 g/dL
Light chain loss < 4g/24h Light chain loss > 12g/24h
N0 Workup
N1 Dutch G1 or NCI LN0-2
N2 Dutch G2 or NCI LN3 –– Skin biopsy: Immunophenotype commonly
N3 Dutch G3-4 or NCI LN4
CD2+, 3+, 4+, 5+, CD 8-, 30-, PCR for T-cell
M0
M1 Visceral involvement receptor gene rearrangement
B0 No Sezary cells, <5% circulating –– Biopsy only cLN+, those that are firm, irregu-
of lymphocytes
B1 Sezary cells, >5%; or neither B0 8
lar, clustered, fixed, or >1.5 cm diameter
or B2 –– If palpable but biopsy negative, still N1
B2 ≥ 1000 Sezary cells/uL
–– CBC with Sezary screen, CMP, LDH
–– Flow cytometry, PCR for T-cell receptor gene
IA T1 N0M0 B0-1. 10y OS 93%
rearrangement
IB T2 N0M0 B0-1. 10y OS 93%
–– PET/CT for stage IB+
IIA T1-2 N1-2 M0 B0-1
IIB T3 N0-2 M0 B0-1
IIIA T4 N0-2 M0 B0. 10 y OS 50%
IIIB T4 N0-2 M0 B1 CD30+ Lymphoproliferative
IVA1:T1-4 N0-2 M0 B2. 10y OS 24% Disorders
IVA2:T1-4 N3 M0 B0-2
IVB: T1-4 N0-3 M1 B0-2 –– Lymphomatoid papulosis: indolent, chronic,
recurrent, self-healing. Disappears in
Primary cutaneous Incidence 3–12 weeks. Histologically indistinguishable
lymphoma type Cell Behavior (%)
MF T Indolent 50
from cutaneous ALCL,
Anaplastic large cell T Indolent 8 –– ALCL: cutaneous lymphoma. Persistent and
(ALCL) (CD30+) progressive. ISRT 30–40 Gy. 99% CR to RT,
Lymphomatoid papulosis T Indolent 12 but 40% relapse. If multifocal, consider MTX,
CD4+ pleomorphic T Indolent 2 systemic retinoids, brentuximab, pralatrexate,
Sezary T Aggressive 3 observation if asymptomatic. If cutaneous
NK/T T Aggressive <3
ALCL within regional LNs, then MTX +/−
Primary MZL B Indolent 7
Primary FCL, t(14;18) B Indolent 11 RT, pralatrexate+/− RT, brentuximab +/− RT,
neg CHOP +/− RT.
Primary DLBCL, leg type B Aggressive 4
426 N. G. Zaorsky et al.
Day of week
Mon Tues Weds Thurs Fri
1 fraction
RPO AP LPO
RPO LAO RPO LAO
1 week AP PA AP PA OFF
LPO RAO LPO RAO
LAO PA RAO
Hematologic Cancers 427
References 15. Longo DL, Glatstein E, Duffey PL, et al. Radiation
therapy versus combination chemotherapy in the
treatment of early-stage Hodgkin's disease: seven-
1. Hartman AR, Williams SF, Dillon JJ. Survival,
year results of a prospective randomized trial. J Clin
disease-free survival and adverse effects of condi-
Oncol. 1991;9:906–17.
tioning for allogeneic bone marrow transplantation
16. Noordijk EM, Carde P, Dupouy N, et al. Combined-
with busulfan/cyclophosphamide vs total body irra-
modality therapy for clinical stage I or II Hodgkin's
diation: a meta-analysis. Bone Marrow Transplant.
lymphoma: long-term results of the European
1998;22:439–43.
Organisation for Research and Treatment of Cancer
2. Clift RA, Buckner CD, Appelbaum FR, et al.
H7 randomized controlled trials. J Clin Oncol.
Allogeneic marrow transplantation in patients with
2006;24:3128–35.
chronic myeloid leukemia in the chronic phase: a
17. Press OW, LeBlanc M, Lichter AS, et al. Phase III
randomized trial of two irradiation regimens. Blood.
randomized intergroup trial of subtotal lymphoid irra-
1991;77:1660–5.
diation versus doxorubicin, vinblastine, and subtotal
3. Clift RA, Buckner CD, Appelbaum FR, et al.
lymphoid irradiation for stage IA to IIA Hodgkin's
Allogeneic marrow transplantation in patients with
disease. J Clin Oncol. 2001;19:4238–44.
acute myeloid leukemia in first remission: a ran-
18. Carde P, Karrasch M, Fortpied C, et al. Eight Cycles
domized trial of two irradiation regimens. Blood.
of ABVD Versus Four Cycles of BEACOPPescalated
1990;76:1867–71.
Plus Four Cycles of BEACOPPbaseline in Stage III
4. Bylund A, Stattin P, Widmark A, et al. Predictive
to IV, International Prognostic Score >/= 3, High-
value of bcl-2 immunoreactivity in prostate cancer
Risk Hodgkin Lymphoma: First Results of the Phase
patients treated with radiotherapy. Radiother Oncol.
III EORTC 20012 Intergroup Trial. J Clin Oncol.
1998;49:143–8.
2016;34:2028–36.
5. Cook B DT: Wikibooks radiation oncology. https://
19. Diehl V, Franklin J, Pfreundschuh M, et al. Standard
en.wikibooks.org/wiki/Radiation_Oncology. Accessed
and increased-dose BEACOPP chemotherapy com-
April 2018.
pared with COPP-ABVD for advanced Hodgkin's
6. Edge S, Byrd D, Compton C, et al. AJCC cancer stag-
disease. N Engl J Med. 2003;348:2386–95.
ing manual. New York: Springer; 2010.
2 0.
Eichenauer DA, Bredenfeld H, Haverkamp
7. Hansen ERM. Handbook of evidence-based radiation
H, et al. Hodgkin's lymphoma in adolescents
oncology. 2nd ed: Springer; 2010.
treated with adult protocols: a report from the
8. Hristov BLS, Christodouleas JP. Radiation oncology -
German Hodgkin study group. J Clin Oncol.
a question based review. 2nd ed: LWW; 2014.
2009;27:6079–85.
9. Aleman BM, Raemaekers JM, Tirelli U, et al.
21. Engert A, Diehl V, Franklin J, et al. Escalated-
Involved-field radiotherapy for advanced Hodgkin's
dose BEACOPP in the treatment of patients with
lymphoma. N Engl J Med. 2003;348:2396–406.
advanced-stage Hodgkin's lymphoma: 10 years of
10. Engert A, Franklin J, Eich HT, et al. Two cycles of
follow-up of the GHSG HD9 study. J Clin Oncol.
doxorubicin, bleomycin, vinblastine, and dacarba-
2009;27:4548–54.
zine plus extended-field radiotherapy is superior to
22. Hasenclever D, Diehl V. A prognostic score for
radiotherapy alone in early favorable Hodgkin's lym-
advanced Hodgkin's disease. International Prognostic
phoma: final results of the GHSG HD7 trial. J Clin
Factors Project on Advanced Hodgkin's Disease. N
Oncol. 2007;25:3495–502.
Engl J Med. 1998;339:1506–14.
11. Engert A, Plutschow A, Eich HT, et al. Reduced treat-
23. Hutchings M, Loft A, Hansen M, et al. Position emis-
ment intensity in patients with early-stage Hodgkin's
sion tomography with or without computed tomogra-
lymphoma. N Engl J Med. 2010;363:640–52.
phy in the primary staging of Hodgkin's lymphoma.
12. Engert A, Schiller P, Josting A, et al. Involved-field
Haematologica. 2006;91:482–9.
radiotherapy is equally effective and less toxic com-
24. Specht L, Yahalom J, Illidge T, et al. Modern radia-
pared with extended-field radiotherapy after four
tion therapy for Hodgkin lymphoma: field and dose
cycles of chemotherapy in patients with early-stage
guidelines from the international lymphoma radiation
unfavorable Hodgkin's lymphoma: results of the HD8
oncology group (ILROG). Int J Radiat Oncol Biol
trial of the German Hodgkin's Lymphoma Study
Phys. 2014;89:854–62.
Group. J Clin Oncol. 2003;21:3601–8.
25. Steidl C, Lee T, Shah SP, et al. Tumor-associated
13.
Ferme C, Eghbali H, Meerwaldt JH, et al.
macrophages and survival in classic Hodgkin's lym-
Chemotherapy plus involved-field radiation in
phoma. N Engl J Med. 2010;362:875–85.
early-stage Hodgkin's disease. N Engl J Med.
26. Herbst C, Rehan FA, Skoetz N, et al. Chemotherapy
2007;357:1916–27.
alone versus chemotherapy plus radiotherapy for
14. Laskar S, Gupta T, Vimal S, et al. Consolidation
early stage Hodgkin lymphoma. Cochrane Database
radiation after complete remission in Hodgkin's dis-
Syst Rev. 2011:Cd007110.
ease following six cycles of doxorubicin, bleomycin,
27. Raemaekers JM, Andre MP, Federico M, et al.
vinblastine, and dacarbazine chemotherapy: is there a
Omitting radiotherapy in early positron emission
need? J Clin Oncol. 2004;22:62–8.
tomography-negative stage I/II Hodgkin lymphoma
428 N. G. Zaorsky et al.
is associated with an increased risk of early relapse: 40. Horning SJ, Weller E, Kim K, et al. Chemotherapy
Clinical results of the preplanned interim analysis of with or without radiotherapy in limited-stage d iffuse
the randomized EORTC/LYSA/FIL H10 trial. J Clin aggressive non-Hodgkin's lymphoma: Eastern
Oncol. 2014;32:1188–94. Cooperative Oncology Group study 1484. J Clin
28. De Bruin ML, Sparidans J, van't Veer MB, et al. Oncol. 2004;22:3032–8.
Breast cancer risk in female survivors of Hodgkin's 41.
Miller TP, Dahlberg S, Cassady JR, et al.
lymphoma: lower risk after smaller radiation vol- Chemotherapy alone compared with chemotherapy
umes. J Clin Oncol. 2009;27:4239–46. plus radiotherapy for localized intermediate- and
29. Chen RC, Chin MS, Ng AK, et al. Early-stage,
high-grade non-Hodgkin's lymphoma. N Engl J Med.
lymphocyte- predominant Hodgkin's lymphoma: 1998;339:21–6.
patient outcomes from a large, single-institution series 42. Friedberg JW, Byrtek M, Link BK, et al. Effectiveness
with long follow-up. J Clin Oncol. 2010;28:136–41. of first-line management strategies for stage I follicu-
30. Al-Mansour M, Connors JM, Gascoyne RD, et al. lar lymphoma: analysis of the National LymphoCare
Transformation to aggressive lymphoma in nodular Study. J Clin Oncol. 2012;30:3368–75.
lymphocyte-predominant Hodgkin's lymphoma. J 43. Friedberg JW, Taylor MD, Cerhan JR, et al. Follicular
Clin Oncol. 2010;28:793–9. lymphoma in the United States: first report of
31. Bonadonna G, Bonfante V, Viviani S, et al. ABVD the national LymphoCare study. J Clin Oncol.
plus subtotal nodal versus involved-field radiotherapy 2009;27:1202–8.
in early-stage Hodgkin's disease: long-term results. J 44. Hoskin PJ, Kirkwood AA, Popova B, et al. 4 Gy ver-
Clin Oncol. 2004;22:2835–41. sus 24 Gy radiotherapy for patients with indolent lym-
32. Donaldson SS, Link MP, Weinstein HJ, et al. Final phoma (FORT): a randomised phase 3 non-inferiority
results of a prospective clinical trial with VAMP trial. Lancet Oncol. 2014;15:457–63.
and low-dose involved-field radiation for children 45. Pfreundschuh M, Trumper L, Osterborg A, et al.
with low-risk Hodgkin's disease. J Clin Oncol. CHOP-like chemotherapy plus rituximab versus
2007;25:332–7. CHOP-like chemotherapy alone in young patients
33. Dorffel W, Ruhl U, Luders H, et al. Treatment of chil- with good-prognosis diffuse large-B-cell lymphoma:
dren and adolescents with Hodgkin lymphoma with- a randomised controlled trial by the MabThera
out radiotherapy for patients in complete remission International Trial (MInT) Group. Lancet Oncol.
after chemotherapy: final results of the multinational 2006;7:379–91.
trial GPOH-HD95. J Clin Oncol. 2013;31:1562–8. 46. Phan J, Mazloom A, Medeiros LJ, et al. Benefit of
34. Engert A, Haverkamp H, Kobe C, et al. Reduced- consolidative radiation therapy in patients with dif-
intensity chemotherapy and PET-guided radiotherapy fuse large B-cell lymphoma treated with R-CHOP
in patients with advanced stage Hodgkin's lymphoma chemotherapy. J Clin Oncol. 2010;28:4170–6.
(HD15 trial): a randomised, open-label, phase 3 non- 47. Lowry L, Smith P, Qian W, et al. Reduced dose
inferiority trial. Lancet. 2012;379:1791–9. radiotherapy for local control in non-Hodgkin lym-
35. Friedman DL, Chen L, Wolden S, et al. Dose-intensive phoma: a randomised phase III trial. Radiother Oncol.
response-based chemotherapy and radiation therapy 2011;100:86–92.
for children and adolescents with newly diagnosed 48. Dunleavy K, Pittaluga S, Maeda LS, et al. Dose-
intermediate-risk hodgkin lymphoma: a report from adjusted EPOCH-rituximab therapy in primary
the Children's Oncology Group Study AHOD0031. J mediastinal B-cell lymphoma. N Engl J Med.
Clin Oncol. 2014;32:3651–8. 2013;368:1408–16.
36. van Nimwegen FA, Schaapveld M, Cutter DJ, et al. 49. Solal-Celigny P, Roy P, Colombat P, et al. Follicular
Radiation Dose-Response Relationship for Risk of lymphoma international prognostic index. Blood.
Coronary Heart Disease in Survivors of Hodgkin 2004;104:1258–65.
Lymphoma. J Clin Oncol. 2016;34:235–43. 50. Haas RL, Poortmans P, de Jong D, et al. High
37. Wolden SL, Chen L, Kelly KM, et al. Long-term response rates and lasting remissions after low-dose
results of CCG 5942: a randomized comparison involved field radiotherapy in indolent lymphomas. J
of chemotherapy with and without radiotherapy Clin Oncol. 2003;21:2474–80.
for children with Hodgkin's lymphoma--a report 51. International Non-Hodgkin’s Lymphoma Prognostic
from the Children's Oncology Group. J Clin Oncol. Factors Project. A predictive model for aggressive
2012;30:3174–80. non-Hodgkin’s lymphoma. N Engl J Med 329:987–
38.
Littrup PJ, Williams CR, Egglin TK, et al. 94, 1993.
Determination of prostate volume with transrec- 52. Hoppe BS, Moskowitz CH, Filippa DA, et al.
tal ultrasound for cancer screening. Radiology. Involved-field radiotherapy before high-dose therapy
1991;179:49–53. and autologous stem-cell rescue in diffuse large-cell
39. Bonnet C, Fillet G, Mounier N, et al. CHOP alone lymphoma: long-term disease control and toxicity. J
compared with CHOP plus radiotherapy for localized Clin Oncol. 2008;26:1858–64.
aggressive lymphoma in elderly patients: a study by 53. Pugh TJ, Ballonoff A, Rusthoven KE, et al. Cardiac
the Groupe d'Etude des Lymphomes de l'Adulte. J mortality in patients with stage I and II diffuse large
Clin Oncol. 2007;25:787–92. B-cell lymphoma treated with and without radiation: a
Hematologic Cancers 429
surveillance, epidemiology, and end-results analysis. the local control. Int J Radiat Oncol Biol Phys.
Int J Radiat Oncol Biol Phys. 2010;76:845–9. 2006;64:1013–7.
54. Aviles A, Neri N, Delgado S, et al. Residual dis- 61. Tsang RW, Gospodarowicz MK, Pintilie M, et al.
ease after chemotherapy in aggressive malignant Solitary plasmacytoma treated with radiotherapy:
lymphoma: the role of radiotherapy. Med Oncol. impact of tumor size on outcome. Int J Radiat Oncol
2005;22:383–7. Biol Phys. 2001;50:113–20.
55. Held G, Murawski N, Ziepert M, et al. Role of
62. Greipp PR, San Miguel J, Durie BG, et al. International
radiotherapy to bulky disease in elderly patients staging system for multiple myeloma. J Clin Oncol.
with aggressive B-cell lymphoma. J Clin Oncol. 2005;23:3412–20.
2014;32:1112–8. 63. Steensma DP, Gertz MA, Greipp PR, et al. A high bone
56. Christie D, Dear K, Le T, et al. Limited chemotherapy marrow plasma cell labeling index in stable plateau-
and shrinking field radiotherapy for Osteolymphoma phase multiple myeloma is a marker for early disease
(primary bone lymphoma): results from the trans- progression and death. Blood. 2001;97:2522–3.
Tasman Radiation Oncology Group 99.04 and 64. Rades D, Hoskin PJ, Stalpers LJ, et al. Short-course
Australasian Leukaemia and Lymphoma Group radiotherapy is not optimal for spinal cord compres-
LY02 prospective trial. Int J Radiat Oncol Biol Phys. sion due to myeloma. Int J Radiat Oncol Biol Phys.
2011;80:1164–70. 2006;64:1452–7.
57. Pellegrini C, Gandolfi L, Quirini F, et al. Primary 65. Salazar OM, Rubin P, Hendrickson FR, et al.
bone lymphoma: evaluation of chemoimmunotherapy Single-dose half-body irradiation for palliation of
as front-line treatment in 21 patients. Clin Lymphoma multiple bone metastases from solid tumors. Final
Myeloma Leuk. 2011;11:321–5. radiation therapy oncology group report. Cancer.
58. Schechter NR, Portlock CS, Yahalom J. Treatment 1986;58:29–36.
of mucosa-associated lymphoid tissue lymphoma 66. Olsen E, Vonderheid E, Pimpinelli N, et al. Revisions
of the stomach with radiation alone. J Clin Oncol. to the staging and classification of mycosis fungoides
1998;16:1916–21. and Sezary syndrome: a proposal of the International
59. Fischbach W, Goebeler ME, Ruskone-Fourmestraux Society for Cutaneous Lymphomas (ISCL) and the
A, et al. Most patients with minimal histological cutaneous lymphoma task force of the European
residuals of gastric MALT lymphoma after success- Organization of Research and Treatment of Cancer
ful eradication of Helicobacter pylori can be managed (EORTC). Blood. 2007;110:1713–22.
safely by a watch and wait strategy: experience from 67. Quaglino P, Pimpinelli N, Berti E, et al. Time
a large international series. Gut. 2007;56:1685–7. course, clinical pathways, and long-term hazards risk
60. Tournier-Rangeard L, Lapeyre M, Graff-Caillaud
trends of disease progression in patients with clas-
P, et al. Radiotherapy for solitary extramedullary sic mycosis fungoides: a multicenter, retrospective
plasmacytoma in the head-and-neck region: A dose follow-up study from the Italian Group of Cutaneous
greater than 45 Gy to the target volume improves Lymphomas. Cancer. 2012;118:5830–9.
Soft Tissue Cancers
11
Nicholas G. Zaorsky, Daniel M. Trifiletti,
and Heath B. Mackley
Abstract
This chapter discusses the general manage-
ment of patients with sarcomas, particularly
soft tissue sarcomas, with special focus on
principles that guide radiotherapy manage-
ment. Several key components of trimodality
care and radiation field design are discussed.
0 10 20 30 40 50 60 70 80 90 100
Points
>50
Age
<=50
>5cm
Size
<=5cm
Close or Positive
Margin
Negative
High
Grade
Low
Others
Histology
Atypical Lipoma or well Diff
Total Points
0 50 100 150 200 250 300 350 400 450
Nomogram to predict the rate of local recurrence of extremity soft tissue sarcoma, after resection, without radiation
therapy. Adapted from [1]
0 10 20 30 40 50 60 70 80 90 100
Points
Age
85 5 75 70 65 60 55 50 45 40 35 30 25 20 15
Total Points
0 20 40 60 80 100 120 140 160 180 200 220 240 260
3-Year LRFS
0.95 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1
5-Year LRFS
0.95 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.05
7-Year LRFS
0.95 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.05
Nomogram to predict the rate of local recurrence of desmoid after surgery, without adjuvant therapy. Adapted from [2]
434 N. G. Zaorsky et al.
Recurrence
Can try surgery and RT again
Soft Tissue Cancers 439
• T4 – >15 cm II T1 2/3 IIA T1 2/3 Pre-op RT (cat 1) then surg, then +/- RT boost +/- chemo
doxo/ifos. PORT for marginal surg, R+, more late fibrosis with post-op. Trial closed
LR. No difference in 5-year DFS (~54%) or early because of higher rates of wound com-
OS (~65%), but not powered for this. plications w pre-op, esp. in ant thigh (45% vs
38%).
Post-op RT • Secondary endpoints: Same LC (93%) and
–– NCIC / Yang 1998: WLE w 1–2 cm margin R0 DM (25%). Pre-op had slight benefit for
+/-EBRT (45 Gy wide field +18 Gy boost to OS (85% vs 72%) but washed out after
bed = 63 Gy). n = 91. G3 tumors got chemo. 5 years, and it was underpowered. Pre-op
RT improved LC for all grades (70% vs RT had higher acute wound complications
100%), but no change in OS or DMFS (similar (35% vs 17%), but lower rate of G2+ fibro-
to MSKCC). For G1 patients, LC favored RT sis (32% vs 48%), edema (15% vs 23%),
arm, 95% vs 67%, NSS. RT worsened joint and joint stiffness (18% vs 23%). Overall,
motion and edema. supports use of pre-op RT in STS.
–– Pisters 1996: 1982–1994. WLE+/−brachy –– CAN-NCIC-SRS (Davis, 2005): compared
(45 Gy LDR Ir-192). Tx volume was 2 cm late RT morbidity from O’Sullivan study,
sup/inf and 1.5–2 cm med/lat. Brachy →↑LC more detailed metrics.
for G3 (89% vs 66% at 5 years), but not for
G1–2. Pre-op RT +/– Boost
• Subset analysis on 20% of pts.: BT –– Toronto, Al Yami, 2010. N = 216. All treated w
improved LC if R0 but not if R2. pre-op RT and have R+. Retrospective review.
–– Higher LR if age >50, recurrent at presenta- 50/25 for 52 pts.; 41 pts. received 16 Gy boost.
tion, R+, fibrosarcoma or malignant periph- No difference in OS 11.5% w/o boost, 22% w
eral nerve tumor. Higher DM if large tumor, boost. 5-year LC 90% vs 74% (NSS).
G3, deep location, recurrent dz, and –– MGH, Pan, 2014. N = 67. Similar to above.
leiomyosarcoma. No difference in LC for boost. Thus, do not
–– MGH (Delaney, 2007): n = 154. Retrospective. boost.
All R+. At 5 years, LC 76%, DFS 48%, and
OS 65%. LC highest if dose >64 Gy (85% vs Hyperthermia
66%). –– EORTC 62961, Issels, ASCO 2007. 341 pts. w
5+ cm G2–3, deep and extracompartmental
Amputation Versus LSS STS get NA EIA +/− regional hyperthermia.
–– NCI, Rosenberg 1982: STS, G3 → amputa- Hyperthermia won, improved LRC 3.8 years
tion vs WLE + RT (boost to 60–70 Gy). vs 2 years and median DFS 2.6 years vs 1.4
Randomization favored LSS, 2:1. No differ- years.
ence in OS (83% at 5 years). Increased LR w
LSS, 20% vs 0%. No difference in 5-year DFS Toxicity
88% vs 83% (NSS) –– MGH (Baldini, 2013): n = 103. STS of
extremity or trunk pre-op RT median dose
Pre-op Versus Post-op RT 50 Gy. Twenty-three had BT boost, median
–– Pollack 1998: pre-op (50 Gy) vs post-op (60– 18 Gy in 2–4 Gy fractions BID. 90%
66 Gy). Same LC (81%). LC. Major wound complications (MWC) rate
–– O’Sullivan 2002: pre-op (50 Gy) vs post-op 35%, and 25% of these required reoperation.
(66–70 Gy). If +margin, pre-op got 16 Gy MWC higher if diabetes, size >10 cm, prox-
boost, and 10/88 pts. received this boost. imity to the skin <3 mm, use of vascular flap
PTV1 expansion was 5 cm prox and distal to or STSG closure. Authors conclude they favor
GTV; PTV2 was reduced to 2 cm around tar- pre-op RT, but 35% MWC rate is
get. Primary endpoint, more temporary wound concerning.
healing problems with pre-op (35 vs 15%) but
Soft Tissue Cancers 443
IGRT Dose
–– RTOG 0630 (Wang, JCO, 2015). Multi- –– 50 Gy in 25 fractions
institutional assessing late tox of STS of
extremity w pre-op IGRT. In pts. with G3 Planning
tumors ≥8 cm, the CTV = GTV + 3 cm longi- –– IMRT or 3D.
tudinal and 1.5 cm radial. For G1 tumors or –– 6 MV beam energy
tumors <8 cm, CTV = GTV + 2 cm longitudi- –– Daily KV imaging
nal and 1 cm radial. 50 Gy/25 to 95% of
PTV. Primary endpoint was toxicity, plan to Technique (for Both Pre- and Post-op)
test 20% abs improvement in G2+ tox at –– Spare >1 cm of limb cross section to decrease
2 years from 37% in the pre-op RT arm of lymphedema risk.
CAN-NCIC-SRT2 study. G2 late tox (subQ –– Treat ½ bone circumference if possible.
fibrosis, joint stiffness, edema) was 10.5% vs –– Minimize joint treated.
37% in CAN-NCIC-SR2. Fibrosis 5.4% vs –– >60 Gy to femur – risk of osteonecrosis.
31%. Joint stiffness 3.5% vs 18%. Edema –– When to treat scar:
15% vs 5.3%. 2-year LC was 94%, all failures • Traditionally, it was always covered.
in CTV. • Haas IJROBP, 2012 review recommenda-
tions: (1) CTV large (>5 cm); (2) G2–3; (3)
R1.
Treatment Paradigm • In low-risk pts. (G1, wide R0), coverage
to drain sites can be omitted. Tx will
–– Stage I: surgery alone, aim for >1 cm SMs, increase risk of late morbidity.
consider RT for +margin (cat 2b) or close SMs
444 N. G. Zaorsky et al.
Constraints Brachytherapy
(RTOG 0630) –– CTV: Tumor bed plus 2–3 cm sup/inf &
–– Joints V50 <50% 1–1.5 cm circumferentially.
–– Bone, weight-bearing V50 <50% –– Afterloading catheters placed 1 cm apart.
–– Skin + subq tissue, longitudinal V20 <50% –– Catheters loaded with iridium-192 loaded
–– Anus V30 <50% after POD 7–14 to permit wound healing.
–– Femoral head: V60% <5% –– Time >7 days can lead to higher infection risk.
–– Kidney V14 <50% –– Dose:
–– Lungs V20 <20% –– 15–25 Gy at 40–50 cGy/hr. given as boost
tx plus 45–50 Gy with EBRT
Additional Constraints, to Prevent Bone –– 45 Gy over 4–6 days, which is equivalent to
Fracture, Pre-op (Dickie, 2009) 60 Gy EBRT, can be given for definitive
–– Bone V40 <64% post-op tx or for tx of recurrent disease
–– Mean bone dose <37 Gy –– HDR-BT: 3.4 Gy BID × 10 fx over 5 days
–– Max bone dose <59 Gy
Indications Preferred over post-op for G2-3 at G1-3 with R+ Preferred over post-op, as <70% amenable
MGH LR s/p prior surgery alone to R0.
Amputation, NV bundle sacrifice, Location not amenable to salvage surgery Pre-op will give smaller field, less chance to
Unavoidable + margin have to escalate near bowel
Timing 3-6 weeks. >6 w assoc w > late fibrosis. 3-8 weeks 3-6 weeks
Pros/cons Smaller field, lower dose (50 Gy), ↑ Bigger fields (~330 cm2 vs. ~420 cm2), N/A
(O’Sullivan, acute wound comps (35 vs 17%), ↓- higher dose (60-66 Gy),
Lancet, 2002) stage; reduced surgical seeding; ↓ acute wound comps,
improved tumor oxygenation (?); may ↑ long-term fibrosis,
improve resectability and chance ↓ ROM;
for R0; <15% LR <15% LR; scar bolus; possible hypoxia in
toxicities are temporary tumor bed
toxicities are permanent
Volumes (Haas, GTV = MR T1 post-gadolinium GTV = MR T1 post-gadolinium (if present) (per IEP) CTV = iGTV + 1.5 cm symmetric
IJROBP, 2012; CTV = GTV CTV = GTV post op bed margins. 4DCT for sim w some form of
White, 2005; + T2 peri-tumoral edema + 4 cm + 4 cm prox/distal, respiratory control if tumor >1 cm. Trim
Wang, 2011; prox/distal, +1.5 cm radial, edited at bone, scar from uninvolved bone, kidney, liver,
Bahig, 2013; +1.5 cm radial, edited at bone bowel/air cavity, extension below uninvolved
Wang, 2015) [59,60] (White). Reasonable to decrease to 3 Haas IJROBP drain site coverage skin, expansion beyond RP compartment. If
cm prox/distal (Wang). recommendations: (1) CTV large (>5 cm); 4D unavailable for upper abd tumor, then
PTV = CTV + 5-10 mm (depending (2) G2-3; (3) R1. CTV is 2-2.5 cm cephalon-caudal, and 1.5-
on IGRT use) PTV = CTV + 5-10 mm 2.0 cm radially.
PTV1 = 50 Gy (per RTOG S-0124) CTV= 5 cm
PTV = 50 Gy PTV2 (cone down to GTV) = 16-20 Gy circumferential margin, trim at OARs down
60-66Gy for R0 to 3 cm.
RTOG 0630 volume (Wang):
66-68Gy for R1
G2/3 tumor AND≥8 cm, CTV sup/inf 70-76Gy for R2
margins 3 cm, radial 1.5 cm. Also Consider SIB PTV 57.5 Gy @ 2.3 Gy/fx
cover edema on T2 MR
ALL OTHERS, CTV sup/inf margins
2 cm, radial 1 cm. Also cover edema
on T2 MR
If R+, consider boost:
10-16 Gy IORT
12-20 Gy BT
10-14 or 16-20 or 20-26 Gy EBRT
Outcomes Likely no difference in outcomes, esp. when control for SM status: LC (O’Sullivan,
Lancet, 2002; Pollack, IJROBP, 1998); OS (O’Sullivan, ASCO, 2004)
LC, DFS, LNM, DM rates (Zagars, IJROBP, 2003). Trend toward improved OS,
LC on meta-analysis (Al-Absi, Ann Surg Onc, 2010)
Pt QOL No difference in 3-year QOL; QOL is likely dependent on tumor characteristics (Davis, JCO, 2002)
448 N. G. Zaorsky et al.
Kaposi Sarcoma [62, 63] Greater dose a/w higher response rate, lower
incidence of residual pigmentation, longer
Overview duration of tumor control
–– Limited data –– South Africa (Radiother Oncol,
–– Treatment not recommended unless cosmetic 2008;88:211). PRT, n = 60. Inclusion criteria,
or functional impairment, not responding to epidemic KS. Arms, 20/5 vs 24/12. Results,
HAART. arms equivalent
Kaposi Sarcoma
–– Four types: Treatment
• AIDS assoc –– Good life expectancy: 40 Gy / 20 fractions,
• Iatrogenic, from immune suppression 30 Gy/15 fx.
• Endemic to sub-Saharan Africa –– Poor life expectancy: Palliate 20 Gy /5 frac-
• Classic – in elderly men w Mediterranean tions, 8–12 Gy/1fx.
or East European heritage –– Other considerations:
–– HHV8 infection • 15 Gy for oral lesions.
• 20 Gy for the eyelids, conjunctiva, and
Studies genitals.
–– Kirova 1998: 30 Gy (15–10 for face/groins). • TSEB given in 4 Gy fractions weekly for
92% response 6–8 weeks has also been used.
–– Stelzer (IJROBP 27,;1993). PRT. Arms: 8 Gy
/ 1 vs 20 Gy / 10f x vs 40 Gy / 20 fx. Results:
Soft Tissue Cancers 449
–– Dose response at ≥55 Gy a/w better LC in RR • Low-dose cytotoxic chemo, e.g., MTX and
(Pediatr Blood Cancer, 2011). doxo
–– MDACC recommends 56 Gy based on their • Targeted tx, e.g., imatinib
experience (IJROBP 2008) because higher
doses have more toxicity. Treatment Planning
–– Nonsurgical approaches: –– CTV = GTV + 3–5 cm longitudinal and 2 cm
• Hormone ablation, e.g., w tam in all other directions
• NSAIDs, e.g., sulindac
Soft Tissue Cancers 451
References 17.
Rosenberg AE, Nielsen GP, Keel SB, et al.
Chondrosarcoma of the base of the skull: a clini-
copathologic study of 200 cases with emphasis on
1. Cahlon O, Brennan MF, Jia X, Qin LX, Singer S,
its distinction from chordoma. Am J Surg Pathol.
Alektiar KM. A postoperative nomogram for local
1999;23:1370–8.
recurrence risk in extremity soft tissue sarcomas after
18. Fernandez-Miranda JC, Gardner PA, Snyderman CH,
limb-sparing surgery without adjuvant radiation. Ann
et al. Clival chordomas: A pathological, surgical, and
Surg. 2012;255(2):343–7. https://doi.org/10.1097/
radiotherapeutic review. Head Neck. 2014;36:892–906.
SLA.0b013e3182367aa7.
19. Carpentier A, Polivka M, Blanquet A, et al.
2. Crago AM, Denton B, Salas S, Dufresne A, Mezhir JJ,
Suboccipital and cervical chordomas: the value of
Hameed M, Gonen M, Singer S, Brennan MF. A prog-
aggressive treatment at first presentation of the dis-
nostic nomogram for prediction of recurrence in des-
ease. J Neurosurg. 2002;97:1070–7.
moid fibromatosis. Ann Surg. 2013;258(2):347–53.
20. Catton C, O'Sullivan B, Bell R, et al. Chordoma:
https://doi.org/10.1097/SLA.0b013e31828c8a30.
long-term follow-up after radical photon irradiation.
3. Bylund A, Stattin P, Widmark A, et al. Predictive
Radiother Oncol. 1996;41:67–72.
value of bcl-2 immunoreactivity in prostate cancer
21. Yamada Y, Laufer I, Cox BW, et al. Preliminary
patients treated with radiotherapy. Radiother Oncol.
results of high-dose single-fraction radiotherapy
1998;49:143–8.
for the management of chordomas of the spine and
4. Cook B DT. Wikibooks radiation oncology. https://
sacrum. Neurosurgery. 2013;73:673–80; discussion
en.wikibooks.org/wiki/Radiation_Oncology.
680.
Accessed April 2018.
22. Hug EB, Loredo LN, Slater JD, et al. Proton radiation
5. Edge S, Byrd D, Compton C, et al. AJCC cancer stag-
therapy for chordomas and chondrosarcomas of the
ing manual. New York: Springer; 2010.
skull base. J Neurosurg. 1999;91:432–9.
6. Hansen EK, Long JD. Handbook of Evidence-Based
23. Fagundes MA, Hug EB, Liebsch NJ, et al. Radiation
Radiation Oncology. 2nd ed: Springer; 2018. ISBN-
therapy for chordomas of the base of skull and cervi-
13: 9783319626413
cal spine: patterns of failure and outcome after relapse.
7. Hristov B, Lin SH, Christodouleas JP. Radiation
Int J Radiat Oncol Biol Phys. 1995;33:579–84.
oncology – a question based review. 2nd ed: LWW;
24. DeLaney TF, Spiro IJ, Suit HD, et al. Neoadjuvant
2014.
chemotherapy and radiotherapy for large extremity
8. Eilber F, Giuliano A, Eckardt J, et al. Adjuvant che-
soft-tissue sarcomas. Int J Radiat Oncol Biol Phys.
motherapy for osteosarcoma: a randomized prospec-
2003;56:1117–27.
tive trial. J Clin Oncol. 1987;5:21–6.
25. Kirova YM, Belembaogo E, Frikha H, et al.
9. Link MP, Goorin AM, Miser AW, et al. The effect of
Radiotherapy in the management of epidemic
adjuvant chemotherapy on relapse-free survival in
Kaposi's sarcoma: a retrospective study of 643 cases.
patients with osteosarcoma of the extremity. N Engl
Radiother Oncol. 1998;46:19–22.
J Med. 1986;314:1600–6.
26. Kraybill WG, Harris J, Spiro IJ, et al. Phase II study
10. Ozaki T, Flege S, Kevric M, et al. Osteosarcoma of the
of neoadjuvant chemotherapy and radiation therapy in
pelvis: experience of the Cooperative Osteosarcoma
the management of high-risk, high-grade, soft tissue
Study Group. J Clin Oncol. 2003;21:334–41.
sarcomas of the extremities and body wall: Radiation
11. Noel G, Feuvret L, Ferrand R, et al. Radiotherapeutic
Therapy Oncology Group Trial 9514. J Clin Oncol.
factors in the management of cervical-basal chor-
2006;24:619–25.
domas and chondrosarcomas. Neurosurgery.
27. Mendenhall WM, Zlotecki RA, Hochwald SN,
2004;55:1252–60; discussion 1260–2.
et al. Retroperitoneal soft tissue sarcoma. Cancer.
12. Amichetti M, Cianchetti M, Amelio D, et al. Proton
2005;104:669–75.
therapy in chordoma of the base of the skull: a sys-
28.
O'Sullivan B, Davis AM, Turcotte R, et al.
tematic review. Neurosurg Rev. 2009;32:403–16.
Preoperative versus postoperative radiotherapy in
13. Kano H, Lunsford LD. Stereotactic radiosurgery of
soft-tissue sarcoma of the limbs: a randomised trial.
intracranial chordomas, chondrosarcomas, and glo-
Lancet. 2002;359:2235–41.
mus tumors. Neurosurg Clin N Am. 2013;24:553–60.
29. Pervaiz N, Colterjohn N, Farrokhyar F, et al. A sys-
14. Bakst R, Wolden S, Yahalom J. Radiation therapy for
tematic meta-analysis of randomized controlled trials
chloroma (granulocytic sarcoma). Int J Radiat Oncol
of adjuvant chemotherapy for localized resectable
Biol Phys. 2012;82:1816–22.
soft-tissue sarcoma. Cancer. 2008;113:573–81.
15. Machak GN, Tkachev SI, Solovyev YN, et al.
30. Pisters PW, Harrison LB, Leung DH, et al. Long-term
Neoadjuvant chemotherapy and local radiotherapy
results of a prospective randomized trial of adjuvant
for high-grade osteosarcoma of the extremities. Mayo
brachytherapy in soft tissue sarcoma. J Clin Oncol.
Clin Proc. 2003;78:147–55.
1996;14:859–68.
16. Souhami RL, Craft AW, Van der Eijken JW, et al.
31. Pollack A, Zagars GK, Goswitz MS, et al. Preopera-
Randomised trial of two regimens of chemotherapy
tive vs. postoperative radiotherapy in the treatment of
in operable osteosarcoma: a study of the European
soft tissue sarcomas: a matter of presentation. Int J
Osteosarcoma Intergroup. Lancet. 1997;350:911–7.
Radiat Oncol Biol Phys. 1998;42:563–72.
Soft Tissue Cancers 453
Abstract
This chapter discusses the general management of patients with bone and spine tumors, with special
focus on principles that guide radiotherapy and radiosurgery management. Several key components
of trimodality care and palliative care are discussed.
D. M. Trifiletti (*)
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
N. G. Zaorsky
Penn State Cancer Institute, Hershey, PA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu
D. E. Spratt
University of Michigan, Ann Arbor, MI, USA
Cortex (rim)
Cancellous
Body
Pedicle
Transverse
process
a
in
m
la
Sup articular
process NICHOLAS G ZAORSKY, MD
Spinous process
Bone/Spine Cancers 457
Goals of Care
1. Preserve/restore neurologic function
2. Reduce pain
3. Provide local control
Epidemiology
• 300,000 patients have osseous metastatic disease/year in the USA.
• The spine is the most common location of bone metastases (60% spine).
• 10% of patients with spine metastases will develop MSCC (~20,000 per year).
• Usually in >50-year-olds.
• Most common cancers causing MSCC are lung cancer, prostate cancer, multiple myeloma, breast
cancer, and lymphoma.
• 20% of MSCC cases lack history of cancer – usually from non-Hodgkin lymphoma, myeloma, and
small-cell lung cancer.
Anatomy
• Most commonly arises from the vertebral body (80%)
• Most commonly in the thoracic spine (60%)
Presentation
• Most common: Pain (70–95%)
• Also common: Sensory deficit (45–90%), weakness (61–91%), and autonomic dysfunction
(40–57%)
Pain Types
• Biologic pain: cancer-related pain and night and morning pain. Non-positional. Inflammatory.
Steroids often help.
• Mechanical: from mechanical strain, not n ecessary cancer-related, positional pain.
• Radicular: radiating pain from osseous or cancer compressing neural foramen, often positional.
• Funicular: radiating pain from compression of the cord itself, non-positional.
Workup
• H&P, full neuro exam assessing for sensation, motor, bowel/bladder, and gait/proprioception
• Steroids (dexamethasone 10 mg IV × 1 and then 4–6 mg Q6H). Careful if no cancer diagnosis is
known (e.g., lymphoma)
• Surgical consult for stability, decompression, fixation
• MRI total spine (20% have additional tumors), emergent bx if needed if no cancer diagnosis
• CT myelogram if MRI contraindicated
• Surgery = 360 degree decompression with s tabilization (not laminectomy)
• CT appearance:
• Lytic: hypodense. Lung, breast, GI, thyroid, melanoma, myeloma, renal cell, urothelial, and ovarian
• Blastic: hyperdense. Lung, breast, GI, prostate, and nasopharynx
458 D. M. Trifiletti et al.
Steroids
• Vecht 1989: loading dose 10 mg IV vs 100 mg IV. Both went on to 16 mg daily. No difference.
• Sorenson, 1994: Randomize to (1) 96 mg IV × 1, then 24 mg QID ×3, then taper over 10 d vs (2)
no dex. Gait function following tx: 81% vs 63%. Ambulatory at 6 m: 59% vs 33%.
NCCN
• Dexamethasone 8–10 mg dexamethasone bolus, then 4 mg Q6H minimum
• Debulking/fixation if solitary site with >3 m life expectancy, paraplegia <24 hrs, not hematologic
cancer, or if unstable
• Post-op RT 1–3 weeks post-op
• Chemo for hematologic cancers
• SBRT not for MSCC
• OAR: thecal sac or cord PRV + 1.5 mm
Radioresponsiveness
Intermediate
Very Favorable Unfavorable Resistant
Lymphoma Prostate HPV-squamous cell Melanoma
Multiple myeloma Breast NSCLC Sarcoma
Germinoma HPV+ squamous cell Renal cell
cEBRT Sim/Planning
• Cervical spine: laterals, pull shoulders down.
• Thoracic/lumbar: Usually AP/PA. Make sure spine is straight.
• Add +/− 1 vertebral body.
• 30/10, 20/5, and 8/1.
MNOP Algorithm for Patient with MSCC (Spratt DE et al. Lancet Onc 2017)
M Unstable Stable
Radioresponsiveness (Histology)
cEBRT Surgery →
Surgically cEBRT or SBRT
P
Stabilize Surgery →
cEBRT or SBRT
Simplified
Radiosensitive Radioresistant
Low-grade MSCC cEBRT SBRT
High-grade MSCC cEBRT Surgery, then
SBRT
462 D. M. Trifiletti et al.
Overview
–– Sites: spine > pelvis > ribs > femur > skull
Fracture Risk
–– Femur: 65%; subtrochanteric > fem neck > peritrochanteric
–– Acetabulum: 9%
–– Tibia: 8%
–– Humerus: 17%
–– Forearm: 2%
Surgery
–– Mirels 1989: scoring system for fracture from mets to long bones. Mean score 7 in nonfracture
group. >8 should get surgical evaluation for prophylactic internal fixation prior to RT.
Mirels points 1 2 3
Site UE LE Peritroch
Pain Mild Mod Mechanical
Radiograph Blastic Mix Lytic
% of shaft <33% 34–67% >68%
–– VanderLinden 2004: femur cortical involvement >30 mm and/or circumferential >50% predict
for fx
Algorithm
–– (1) Fracture: surgery and then post-op RT.
–– (2) No fracture: if painless, observe; if painful, then assess for impending fracture. If no impending
fracture, then RT. If yes, then surgery.
EBRT
–– RTOG 97–14 (Harstell 2005): breast and prostate bone mets → 8 × 1 vs 30/10. Overall response
66%. Pain CR same: 15–18%. At 3 mos, 33% no longer required narcotics. More G2+ tox with
30/10 (17% vs 10% G2-4 tox). More retreatment with 8 × 1 (9 vs 18%).
–– TROG 96.05: 1996–2002, RCT of 8 Gy x 1 vs 20 Gy x 5. 8 × 1 not effective as 20/5, but not SS
worse.
–– Bone pain trial working party: 8 × 1 vs 20/5 vs 30/10. Same effectiveness. More reRT with 8 × 1
(23 vs 10%).
–– Chow 2007: meta-analysis. No differences except 2.5× increase in retreatment if 8 × 1.
–– ASTRO guidelines: Essentially 8 Gy x 1 for everything.
Bone/Spine Cancers 463
Radiopharmaceuticals
–– Strontium-89 (β): Sciuto 2002 and Porter 1993
–– Samarium-153 (β and γ): Sartor 2004 and Oosterhof 2003
–– Radium-223 (α):
• ALSYMPCA: Ra223 improved OS (11 → 14.9 m) over placebo for symptomatic mCRPC
patients
PORT
–– Townsend 1994: PORT reduced need for 2nd surgery (15 → 2%) and improved function
SRS/SBRT
General concept – SRS/SBRT often used for radioresistant histologies (sarcoma, melanoma, RCC)
and oligometastatic disease. However, for simple pain control, ASTRO recommends 8 Gy x 1.
–– Gerstzen 2007: Retrospective, median 20Gy x1. Improved pain in 86%, LC 90%. More durable
than historical series of 8 Gy x 1 or 30 Gy in 10 fx.
464 D. M. Trifiletti et al.
blind, randomised placebo-controlled, phase 3 trial. Canadian Cancer Centers. Int J Radiat Oncol Biol
Lancet Oncol. 2015;16:1463–72. Phys. 2009;75:193–7.
30. Howell DD, James JL, Hartsell WF, et al. Single- 3 2. Loblaw DA, Wu JS, Kirkbride P, et al. Pain flare in
fraction radiotherapy versus multifraction radio- patients with bone metastases after palliative radio-
therapy for palliation of painful vertebral bone therapy--a nested randomized control trial. Support
metastases-
equivalent efficacy, less toxicity, more Care Cancer. 2007;15:451–5.
convenient: a subset analysis of Radiation Therapy 33. Gomez-Iturriaga A, Cacicedo J, Navarro A, et al.
Oncology Group trial 97–14. Cancer. 2013;119: Incidence of pain flare following palliative radiother-
888–96. apy for symptomatic bone metastases: multicenter
31. Hird A, Chow E, Zhang L, et al. Determining the inci- prospective observational study. BMC Palliat Care.
dence of pain flare following palliative radiotherapy 2015;14:48.
for symptomatic bone metastases: results from three
Physics and Radiobiology
13
Daniel M. Trifiletti, Nicholas G. Zaorsky,
and David J. Schlesinger
Abstract
This chapter discusses the basics of clinical radiation physics and radiation biology. Special
emphasis is placed on the key aspects of physics and radiobiology that guide clinical management
and preparation for board exams and maintenance of certification exams.
Radiation Physics
This chapter discusses the basics of clinical radiation physics and radiation biology. Special emphasis
is placed on the key aspects of physics and radiobiology that guide clinical management and prepara-
tion for board exams and maintenance of certification exams.
Units
Measurement Common units Official (SI) unit
Energy Joules (J), mega-electron volts (MeV) Joules (J)
Activity: disintegrations per unit time Curie (Ci) Becquerel (Bq)
Exposure: ionization Roentgen (R) Coulombs/kg (C/kg)
Absorbed dose: energy deposited in tissue Rad Gray (Gy)
1 Gy = 1 J/kg
Dose equivalent: biological effect Rem Sievert (Sv)
D. M. Trifiletti (*)
Mayo Clinic, Jacksonville, FL, USA
e-mail: Trifiletti.daniel@mayo.edu
N. G. Zaorsky D. J. Schlesinger
Penn State Cancer Institute, Hershey, PA, USA University of Virginia, Charlottesville, VA, USA
e-mail: nzaorsky@pennstatehealth.psu.edu e-mail: djs9c@hscmail.mcc.virginia.edu
Useful Constants
• λ = “lambda” is the inverse of the mean lifetime, aka decay rate, or how much decays in a given
amount of time. Units is disintegrations / (time period you are using).
( (
t1/ 2 = -.693* t / ln A(t ) / A0 ))
Given activities, need to find the half-life:
I x = I0e- m x
Dose Ratios
q = 90° - j / 2
Field-Matching Problems
Additional requiredrotation = 2* ϴ
Physics and Radiobiology 471
For MV ElectronBeams
D Q w = M * Pgr Q* k ¢R 50* kecal * N 60Co D ,w
pion = éë(1 - VH / VL ) / ( M H / M L - VH / VL ) ùû
• VH = high bias voltage (usually 300 V), ML = low bias voltage (usually 150 V)
• MH = measurement under high bias, ML = measurement under low voltage
• Pion >1 since MH should be greater than ML
472 D. M. Trifiletti et al.
Film Analysis
Optical density = log (Io/It).
• Adjusts absorbed dose for the biological effectiveness of the type of radiation
• D = absorbed dose (Gy)
• WR = radiation weighting factor
• Adjusts absorbed dose to account for the sensitivity of each organ to each type of radiation
• T = organ
• WT = weighting factor for organ
• HT = equivalent dose for organ T
• ∑T *WT = 1
Physics and Radiobiology 473
Brachytherapy
Primary Barrier
B = Pd 2 / WUT
# requiredTVLs = n = −log(B)
Required barrier thickness = X = TVL1 + (n−1)TVLe
Radiobiology
• n = number of fractions
• d = dose per fraction
• α/β = alpha/beta ratio for target
( nd / n1d1 ) = (a / b + d1 ) / ( a / b + d )
• n = standard # fractions
• n1 = equivalent # fractions needed for altered schedule
• d = standard dose / fraction
• d1 = desired dose / fraction
EQD 2 = éë BED / (1 + 2 / (a / b ) ) ùû
Translocation Cancer
t(2:13) and t(1:13) Alveolar rhabdomyosarcoma
t(8:14) and t(8:22) Burkitt’s, B-cell All
t(11:14) Mantle cell (BCL1, cyclin D1)
t(11:22) Ewings, PNET
t(12:22) Clear cell sarcoma
t(14:18) Follicular, DLBCL (BCL2)
t(14:19) CLL (BCL3)
t(X:18) Synovial cell sarcoma
Cancer CD testing
All lymphoid 45+
B cells 19+, 20+, 22+
T cells 2+, 3+, 5+, 7+, 4 + (helper), 8+ (cytotoxic)
NK cells 16+, 56+, 57+
Follicular 5-, 10+, 43-
Mantle cell 5+, 23−, 43+
MALT 5−, 10−, 23−
Hodgkin 15+, 30+
Physics and Radiobiology 477
478 D. M. Trifiletti et al.
Physics and Radiobiology 479
A B
Abdominoperineal Resection (APR), 293 Barcelona Clinic Liver Cancer (BCLC), 287
Active surveillance (AS), 318 Basal cell carcinoma (BCC), 184
Adenocarcinoma, 166 anatomic areas, 184
Adenoid cystic carcinoma (ACC), 169, 170 chemo, 184
Adjuvant chemo, 151 electron background, 185
Adjuvant ipilimumab, 182 general, 184
Adjuvant radiation therapy, 155, 187 local recurrence, high-risk factors for, 185
Adjuvant therapy, 180, 182 orthovoltage background, 185
Adriamycin, 180 retrospective reports, 184
Aggressive palliation, 189 surgery background, 184
Alveolar ridge, 157 workup, 184
Amphotericin B, 145 Benadryl, 145
Anal cancer Bevacizumab, 97
AJCC, 300 Bilateral neck, 153
anorectal contouring, 305, 306 Bilateral Wilms’ approach, 53
Australasia GI Trials, 304 Biliary tract cancer, 282, 283
dose constraints, 307 Bladder cancer, 333–337
follow-up, 302 Bone metastases, 55
HGSIL, 300 algorithm, 462
management, 302 EBRT, 462
outcomes, 302 PORT, 463
overview, 300 radiopharmaceuticals, 463
RT vs. CRT, 301 risk of, 462
simulation, 304 SRS/SBRT, 463
toxicity, 302 surgery, 462
treatment, 301 Brachial plexopathy, 146
Anaplastic carcinoma, 179 Brachial plexus, 141, 202, 205
Anaplastic thyroid cancer, 179 Brachytherapy, 474–475
Anterior border, 175 Brain metastases, 55
Arnold’s nerve, 162, 173 asymptomatic brain metastases, 107
Arteriovenous malformation (AVM) diagnosis, 107
clinical presentation, 119 postoperative radiation, 109, 110
diagnosis, 119 prognostic models evolution, 108
follow-up, 120 SRS
grading system, 119 dose, 109
natural history, 119 multi-fraction SRS for larger targets, 109
outcomes, 119 multiple metastases, 109
pathophysiology, 119 SRS +/− WBRT, 108, 109
SRS toxicity, 120 targetable mutations, 109
treatment, 119 WBRT +/− SRS, 108
Atypical teratoid rhabdoid tumor (ATRT), 67 symptomatic brain metastases, 107
Auriculotemporal nerve, 169, 173 Brainstem glioma, 78
C D
Carboplatin, 156, 164 Dabrafenib, 183
Carcinoid DeCIDE trial, 164
diagnosis, 218 Desmoid tumor (DT), 449, 450
H/P, 218 Dexamethasone, 156
imaging, 218 Differentiated thyroid cancer (DTC), 179
neuroendocrine carcinoid, 218 Diffuse anaplasia (DA), 52
Cavernous sinus, 150, 152 Diffuse intrinsic pontine glioma (DIPG), 78
Central nervous system (CNS) tumors Dihydropyridine dehydrogenase (DPD) deficiency, 293
AVM (see Arteriovenous malformation) Dorsal vagal complex (DVC), 140
brain metastases (see Brain metastases) Doxepin mouthwash, 145
brain pathology, 87 Dysphagia/aspiration-related structure (DARS), 140
clinical history, 88
conformity index, 89
CSF flow, 88 E
diagnosis, 88 Early-stage non-small cell lung cancer
ependymoma (see Ependymoma) brachytherapy, 206
gliomas (see High-grade glioma (HGG)) (see contours, 207
Low-grade glioma (LGG)) conventional RT alone, 206
gradient index, 89 during RT, 208
hemangiopericytoma, 121 follow-up, 208
intradural intramedullary spinal cord inoperability, 206
tumors, 88, 89 RFA, 206
meningioma (see Meningioma) risks, 206
MRI, 88 SBRT, 206, 207
optic nerve sheath meningioma, 106 eligibility, 207
PCNSL (see Primary CNS lymphoma (PCNSL)) high-risk factors, failure after, 208
pituitary tumors (see Pituitary tumors) ongoing trials, 207
primary ocular DLBCL, 102 peripheral regimens, 208
radionecrosis, 89 requirements in Pulm function, 207
single-fraction SRS, 87 simulation, 207
standard fractionation, 87 surgery, 206
TN (see Trigeminal neuralgia) Ear pain, 162, 173
TO (see Thyroid ophthalmopathy) Elective nodal irradiation, 170
VS/AN (see Vestibular schwannoma/acoustic Electrons, 183
neuroma) Endocrine remission, 113, 114
Cetuximab, 163, 165 Endometrial cancer
Chemoradiation, 165 adjuvant chemo, 378
Children’s Oncology Group (COG) approach, 66 adjuvant RT, 377
craniopharyngioma, 73 adjuvant VCBT, 377
ependymoma, 68, 69 adjuvant WPRT, 377
germinoma, 72 fertility preservation, 381
NGGCT, 72 follow-up, 382
pineoblastoma, 66 general dosing, 380
pineocytoma, 66 hysterectomy types, 376
Chlorpromazine, 145 imaging, 375
Cipro, 146 indications, 381
Cisplatin, 160, 164, 165 malignancy, 378
Cisplatinum, 156 pathology, 375
Clark method, 182 salvage, 381
Clinical target volume (CTV) expansions, 139, 175 stage I, 375, 381
Clivus, 152 stage III/IV, 375, 381
Clodronate, 146 surgery, 376, 377
Cochlea, 140 toxicity, 382
Colon cancer, 299 treatment, 379
Corticotroph adenomas (Cushings), 113, 114 vaginal relapse, 378
Index 483
suprasellar mass, 29 R
treatment comparison, 33–39 Radiation physics
PEG tube, 140 brachytherapy
PENTOCLO regimen, 146 patient release regulations, 474–475
PET-CT, 139, 167 sources, 474
Pharyngeal constrictors, 140 constants, 468
Photons, 183, 187 corrected ion chamber reading, 471
Pineal tumors, 70 decay equations and calculations, 468
Pineoblastoma, 66 divergence of parallel–opposed fields, 470
Pineocytoma, 66, 72 dose ratios, 469
Pituitary tumors electron beams, 470
clinical presentation, 112 film analysis, 472
corticotroph adenomas (Cushings), 113, 114 ion recombination correction, 471
DDx sellar mass, 112 linear accelerator calibration
diagnosis, 112, 113 MV electron beams, 471
differential diagnosis, 112 MV photon beams, 471
lactotroph adenomas (prolactinoma), 113 organ weighting factors, 473
nonsecretory adenomas, 113 photon attenuation equations, 468
outcomes, 114 photon SAD setups, 470
RT, 114 photon SSD setups, 470
somatotroph adenomas (growth hormone), 114 polarity correction, 472
toxicity/follow-up, 114 radiation linear energy transfer and weighting factors,
treatment, 113 473–474
Plaque technique, 149 radiation safety and dose limits, 472
Platinum-monotherapy, 158 radiation weighting factors, 472
Pleural mesothelioma radiotherapy structural shielding equations, 475
adj RT, 227 temperature and pressure correction, 471
advanced stage/unresectable, 227 tumor markers, 476–478
AJCC7 staging, 226 typical external beam parameters, 473
AJCC8 staging, 226 units, 467–468
chemotherapy, 227 wedge angle and hinge angle, 470
constraints, 227 Radiation weighting factors, 472
diagnosis, 226 Radical neck dissection (RND), 135
NCCN guidelines, 227 Radioactive iodine, 180
radiation, 227 Radiobiology, 475
surgery, 226 Radionecrosis, 89
techniques, 227 Radiotherapy structural shielding equations, 475
Posterior border, 175 Rectal cancer
Posterior fossa syndrome, 29 adj chemo/XRT studies, 294
Posterior fossa tumors (Ddx), 63 anatomy, 293
Prednisone, 146 APR, 293
Primary barrier, 475 chemo, 293
Primary CNS lymphoma (PCNSL) follow-Up, 297
chemo + hyper-fractionated RT, 101 IMRT, 297
chemo + low-dose WBRT, 100 LAR, 294
chemo +/– RT, 100 local recurrence, 296
diagnosis, 100 NCCN, 296
extranodal NHL, 100 neoadj chemo/XRT studies, 294
imaging, 100 outcomes, 297
prognosis, 100 overview, 293
salvage WBRT, 101 post-op RT, 294
simulation/planning, 102 preop CRT, 295, 296
treatment, 101, 102 preop RT, 295
Primary mediastinal B-cell lymphoma requirements, 296
(PMBCL), 415 3D-CRT, 297
Primary ocular DLBCL, 102 TME, 294
Pterygoid fossa, 152 transanal vs. APR/LAR, 296
Pterygopalatine fossa, 152 transanal excision, 294
PTV expansions, 139 Recursive partitioning analysis (RPA), 108
490 Index
U
T Upper Urothelial Tract (UUT) Cancer, 354
Taxol, 180 Urothelial Cell Carcinoma (UCC), 338
Taxotere, 164 Use factor (U), 475
T-cell acute lymphoblastic leukemia Uterine cancer
(T-ALL), 40 adjuvant chemo, 378
Thymoma and thymic carcinoma adjuvant RT, 377
adj chemo, 224 adjuvant VCBT, 377
AJCC 8th edition staging, 223 adjuvant WPRT, 377
diagnosis, 223 fertility preservation, 381
inoperable surgery, 224 follow-up, 382
masaoka staging, 223 general dosing, 380
natural history, 224 hysterectomy types, 376
neoadjuvant chemo, 224 imaging, 375
paraneoplastic syndrome, 223 indications, 381
PORT, 224 malignancy, 378
posttreatment, 224 pathology, 375
surgery, 224 salvage, 381
treatment, 224 stage I, 375, 381
WHO grading, 223 stage III/IV, 375, 381
Thyroid cancer, 166, 179 surgery, 376, 377
adjuvant therapy, 180 toxicity, 382
after RAI, 180 treatment, 379
anaplastic thyroid cancer, 179 vaginal relapse, 378
differentiated, 179 VCBT, 380
NCCN, 180 WPRT, 380, 381
pathology, 179 Uveal melanoma (see Ocular melanoma)
post-op, 180
primary therapy, 180
radioactive iodine, 180 V
RT, 180 Vaginal cancer, 390–391
work-up, 179 VA Larynx Trial, 173
Thyroid dysfunction, 146 Vemurafenib, 183
Thyroid ophthalmopathy (TO) Vertical hemilaryngectomy, 175
492 Index