You are on page 1of 308

PERS PE C T IV E Where Americans Die

From the Veterans Affairs Boston Health 1. Cross SH, Warraich HJ. Changes in the Los Angeles Times. February 16, 2020
Care System, Section of General Internal place of death in the United States. N Engl J (https://www​.­latimes​.­com/​­opinion/​­story/​­2020​
Medicine (M.W.W.), Brigham and Women’s Med 2019;​381:​2369-70. -­02​-­16/​­doctor​-­patients​-­send​-­home​-­to​-­die).
Hospital (M.W.W., R.S.S., J.S.W.), and the 2. Gerrard R, Campbell J, Minton O, et al. 5. Evans WG, Cutson TM, Steinhauser KE,
Dana–Farber Cancer Institute, Department Achieving the preferred place of care for Tulsky JA. Is there no place like home?
of Psychosocial Oncology and Palliative Care hospitalized patients at the end of life. Pal- Caregivers recall reasons for and experi-
(M.W.W.) — all in Boston; and the Institute liat Med 2011;​25:​333-6. ence upon transfer from home hospice to
for Health, Health Care Policy, and Aging Re- 3. Barnato AE, Anthony DL, Skinner J, inpatient facilities. J Palliat Med 2006;​ 9:​
search and the Department of Family Medi- Gallagher PM, Fisher ES. Racial and ethnic 100-10.
cine and Community Health, Rutgers Uni- differences in preferences for end-of-life
versity, New Brunswick, NJ (E.A.L.). treatment. J Gen Intern Med 2009;​24:​695- DOI: 10.1056/NEJMp2112297
701. Copyright © 2022 Massachusetts Medical Society.
Where Americans Die

This article was published on March 12, 4. Gray N. Think you want to die at home?
2022, at NEJM.org. You might want to think twice about that.
The Patient Resident

The Patient Resident


Natasha Z. Rabinowitz Steele, M.D., M.P.H.​​

I slide open the door and slip


into my patient’s room. Mr. C.
is a 64-year-old army veteran
biopsy,” I say gently. Our eyes
meet, and my body language gives
me away. His eyes begin welling
called my father, a gentle, kind
family doctor. “Dad . . . do I
have cancer?” I asked, catching
who presented several days ago with tears. my breath — the words sounded
with massive hemoptysis. I say “It’s cancer, isn’t it?” I nod. surreal as I heard them out loud.
those words aloud each morn- His eyes slowly close as the My hands held my eyes shut as
ing on rounds, but they hardly news sinks in. A monitor alarms he promised he’d be on the next
describe him. as his heart rate soars. I silence flight.
I learned intimate details about it — I don’t need technology to Earlier on the day of my diag-
his body from his laboratory, im- tell me he’s terrified. I gaze down nosis, I’d rounded as a new in-
aging, and pathology reports. at his trembling hands — the tern on the cardiology service. I
But I only came to know him hands that once held his chil- had brushed off a nagging cough
through the hours at his bedside. dren, wiped tears from his wife’s and weakness: I’d expected to
The woman he loved was work- face, that traced the sign of the feel run down after my cesarean
ing in the artichoke fields and cross each Sunday at Mass. delivery, so I pushed through. I
struggling to learn English. He We sit in silence. It’s a loud was doubled over in pain by noon
was proud of his three sons, silence, full of racing fears of the conference, but it passed. I learned
scattered around the country. Af- unknown. It’s that moment when in medical school to work through
ter a career in combat, he was a everything has changed for him: discomfort, prioritize patient care
God-fearing man, but he feared he has gone from before to after. over my own body, and see pa-
death most of all. I know this moment all too well. tients and doctors as inherently
When I first examined him, I I look at my own hands — different. It didn’t dawn on me
suspected he had cancer. He was hands that a year prior first held that I could be gravely ill. Then a
cachectic, and I could feel the my newborn daughter. Hands that, rubbery supraclavicular node ap-
uneven contours of his organs soon after, clutched my phone as peared, and I couldn’t ignore it.
beneath my fingers. Now, I’ve I read my own radiology report: Just 2 weeks into my intern-
come to share the results of the “Mediastinal lymphadenopathy ship, the disparate identities of
biopsy. consistent with lymphoma. Bilat- physician, mother, and cancer
“There she is,” says Mr. C. eral pleural effusions.” I remem- patient had collided inside me. I
with a smile, pulling himself up ber the room started to spin but didn’t know who I should be at
in bed. As his intern, I am a famil- I felt paralyzed; then paralysis gave any given moment. I had expect-
iar face amidst the ever-changing way to sobs. “I don’t want to ed to learn medicine by observing
personnel. leave her,” I repeated to my hus- and caring for patients; instead, I
“I came by to talk about your band over and over. Panicked, I learned about chemotherapeutics,

1010 n engl j med 386;11  nejm.org  March 17, 2022

The New England Journal of Medicine


Downloaded from nejm.org by Salvador Macias-Díaz on March 23, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
PE R S PE C T IV E The Patient Resident

antiemetics, and granulocyte col- Over the next year, I under- ized in the same room. I caught
ony-stimulating factors firsthand. went 12 chemotherapy treatments, a glimpse of myself in the mirror
At a time when I was just begin- two hospitalizations, two pericar- where I’d once stared at my
ning a life of healing by day and diocenteses, four PET scans, and sunken face; now a rounder face
parenting by night, I suddenly innumerable blood tests, echo- looked back at me. I felt my fear
became a patient in my own hos- cardiograms, and x-rays. Being return. Would I get sick again? I
pital, at the mercy of my mercu- sick was a full-time job. Each day had shed the cloak of cancer, yet
rial biology. felt like free soloing a cliff: at I still carried its weight.
I was shaking as I removed any moment, bad news from a lab I stopped rushing through my
my crisp white coat and climbed or scan could hasten the abyss. ward work. When patients spoke,
into a hospital bed, donning a On my last day of treatment, I slowed to listen. I pledged to be
patient gown for the first time. there was no bell to ring, no their ally in a journey through
The radiology report had prompt- celebratory singing. My hospital the unknown, a journey I share.
ed an echocardiogram that re- bracelet was clipped off, and I Slowly, my identities as cancer
vealed cardiac tamponade — my was discharged with instructions patient, doctor, and parent began
heart was straining to pump to return in a few weeks for a fi- to complement one another, their
blood past an overwhelming tu- nal scan. The results came swiftly sum greater than the parts.
mor burden in my chest. That in a phone call. I was cancer- The alarm sounds again and
evening, I lay awake, remember- free. But being cured was only startles me out of my reverie. I
ing the patients I’d rounded on the start of healing. squeeze Mr. C.’s hand, and he
that morning. It struck me that I The following day, the most squeezes back. We sit that way
was sicker than any of them. On glaring remnant of my disease, for a long time.
the verge of cardiac collapse, I the long central catheter to my Later that night I’m at the din-
felt terrifyingly mortal. right atrium, was removed. My ner table with my family, marking
After I left the hospital, time body was returned to me, no lon- the first anniversary of my diag-
slowed. For years, I had been in a ger merely a vehicle for fighting nosis. My husband puts his arm
rush — working to get into a malignant force. But was it even around me, and I begin to cry.
medical school; withstanding the still me? My oncologist cleared Our daughter, just learning to
pressure to survive classes; tran- me to return to residency. No walk, comes to me. She reaches
sitioning to wards while trying to longer sick but far from well, I for my face, gently touching my
maintain my marriage and start a stumbled through my first steps tears in wonderment. Tears that
family; prepping for board exams of survivorship. fall because I’m still here, because
and residency interviews. Now, I was eager to be on the wards I’m well, because I can witness
the cadence of my life followed a in this time of Covid — I had my daughter just beginning her
series of 2-week cycles centered patiently awaited the chance to journey. I think about Mr. C., re-
around chemotherapy. My hair fell match the tempo and bravado of calling the tears and the touch of
out in fistfuls, my postpartum my coresidents, chasing the diag- someone who is nearing the end
skin hung off my aching bones, noses and prognoses of others. of his own. Though all of our
my breast milk vanished. I feared But internally, I was struggling journeys will have beginnings and
holding my daughter too close, with the now-porous partition endings, our lives are what we
wanting to keep her safe from between doctor and patient. Every choose to do with the precious,
the war raging in my body. When patient reminded me of some as- unpredictable, terrifying, and
I did hold her, I felt mind-expand- pect of my own illness. Their pri- beautiful moments in between.
ing love laced with an over- mal fear was familiar, and the Identifying details have been changed to
protect the patient’s privacy.
whelming fear of loss. She’d scent of their hospital gowns Disclosure forms provided by the author
scrunch up her face and give me evoked memories of my own. are available at NEJM.org.
a smile identical to mine at her The hospital was full of mean- From Stanford Medicine, Stanford, CA.
age, evoking a time when I was ingful landmarks. One day I This article was published on March 12,
naive to this pain. She was just paused at the doorway of a pa- 2022, at NEJM.org.
starting her life, and I was fight- tient with breast cancer, remem- DOI: 10.1056/NEJMp2116289
ing for mine. bering when I’d been hospital- Copyright © 2022 Massachusetts Medical Society.
The Patient Resident

n engl j med 386;11  nejm.org  March 17, 2022 1011


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on March 23, 2022. For personal use only. No other uses without permission.
Copyright © 2022 Massachusetts Medical Society. All rights reserved.
Radiología. 2014;56(3):193---205

www.elsevier.es/rx

RADIOLOGÍA HOY

RECIST y el radiólogo
J. Cervera Deval

Servicio de Radiología, Fundación Instituto Valenciano de Oncología, Valencia, España

Recibido el 25 de septiembre de 2011; aceptado el 12 de marzo de 2012


Disponible en Internet el 16 de agosto de 2012

PALABRAS CLAVE Resumen Mediante las imágenes biomédicas podemos diagnosticar, estadificar, controlar y
RECIST; valorar la efectividad de los procedimientos terapéuticos a los que son sometidos los pacientes
Respuesta del tumor; oncológicos. RECIST (Response Evaluation Criteria In Solid Tumors) es el método para monitori-
Estudios de zar el tratamiento mediante medidas unidimensionales de los tumores, obtenidas con técnicas
seguimiento; de imagen reproducibles como son la TC, la RM y la PET. Los cambios metabólicos inducidos por
Oncología; los nuevos tratamientos modifican la biología y comportamiento del tumor, con discordancia
Imagen diagnóstica; ocasional entre el estado clínico del paciente y la respuesta medida con RECIST, lo que apunta
PET; a la necesidad de incluir pruebas funcionales en la valoración de la respuesta al tratamiento.
Técnicas de El objetivo es revisar los criterios RECIST conociendo la aportación de la imagen funcional
diagnóstico para valorar correctamente la eficacia y la repercusión del tratamiento en los pacientes con
molecular; tumores sólidos.
Tumores sólidos © 2011 SERAM. Publicado por Elsevier España, S.L. Todos los derechos reservados.

KEYWORDS RECIST and the radiologist


RECIST;
Tumor response; Abstract Biomedical imaging makes it possible not only to diagnose and stage cancer, but also
Follow-up studies; to follow up patients and evaluate the response to treatment. RECIST (Response Evaluation
Oncology; Criteria In Solid Tumors) provides a method to monitor the response to treatment based on one
Diagnostic imaging; dimensional measurements of tumors obtained with reproducible imaging techniques like CT,
PET; MRI, and PET. The metabolic changes induced by new treatments modify the biology and beha-
Molecular diagnostic vior of the tumor; occasionally, there is a discrepancy between the patient’s clinical condition
techniques; and the response measured by RECIST, which indicates that functional tests need to be included
Solid tumors in the evaluation of the response to treatment.
The objective is to review the RECIST criteria to include the contribution of functional imaging
to enable the efficacy and effects of the treatment in patients with solid tumors.
© 2011 SERAM. Published by Elsevier España, S.L. All rights reserved.

Correo electrónico: jcervera@Fivo.Org

0033-8338/$ – see front matter © 2011 SERAM. Publicado por Elsevier España, S.L. Todos los derechos reservados.
http://dx.doi.org/10.1016/j.rx.2012.03.010
194 J. Cervera Deval

Introducción
La RAE define «respuesta» como «efecto que se pretende
conseguir con una acción»1 . En el ámbito de la oncología
y la radiología, se trata de medir la respuesta a los tra-
tamientos con biomarcadores de imagen y, así, mantener
un tratamiento o suspenderlo para evitar posibles toxicida-
des y gastos innecesarios. La respuesta al tratamiento de
los tumores sólidos clásicamente se basa en los cambios
de tamaño, elemento base de los criterios RECIST (Res-
ponse Evaluation Criteria In Solid Tumors)2 . Estos cambios
intentan representar el comportamiento biológico de los
tumores, lo que ocasionalmente crea controversia porque,
en los pacientes tratados con nuevos fármacos, no siempre
un aumento del tamaño significa progresión3 .
En ensayos clínicos se han usado diferentes criterios con
el objetivo de normalizar la valoración de la respuesta al tra-
tamiento, elemento fundamental para poder comparar los
efectos de los tratamientos. Más aún cuando se desarrollan
nuevos fármacos y terapias costosas para mejorar la super-
vivencia, y cuyos objetivos son 1) la mejoría clínica, por la
reducción de la carga tumoral, o 2) la curación, por la desa-
parición completa del tumor. El propósito de esta revisión
es repasar los criterios RECIST 1.1 e introducir los métodos
de valoración con imagen funcional en los tratamientos con
los nuevos quimioterápicos.

Criterios RECIST 1.1

A finales de los 90, la OMS4 unifica la valoración de la res-


Figura 1 Imagen axial de TC sin contraste endovenoso.
puesta y de la recidiva, el intervalo libre de enfermedad, y
Adenocarcinoma de pulmón. (A) Medida de los diámetros
establece una graduación de la toxicidad del tratamiento.
perpendiculares máximos, OMS. (B) Medida unidimensional
En el año 2000 aparecen los criterios RECIST2 como modifi-
máxima, RECIST. La suma de los diámetros máximos de hasta 5
cación de los criterios de la OMS, actualizados en 2009 a la
lesiones y solo 2 por órgano (RECIST 1.1) define la carga tumoral
versión 1.1 (http://www.recist.com) (fig. 1)5 , que incluye
del paciente.
la resonancia magnética (RM) y la PET con 18 F-FDG como
modalidades de imagen aceptadas para valorar la respuesta6
(tabla 1). Modificaciones importantes que la actualización Para aplicar los criterios RECIST, primero hay que defi-
1.1 aporta son la reducción del número de lesiones diana nir en el estudio inicial las lesiones diana representativas y
(máximo de 5, solo 2 por órgano); el límite de 5 mm, por reproducibles durante el seguimiento, teniendo en cuenta
debajo del cual las lesiones no se consideran medibles; la que su gran variabilidad (confluencia, fragmentación, mala
inclusión de las adenopatías como lesiones diana cuando su definición, técnica de imagen, apreciación), y la dificultad
eje corto sea mayor de 15 mm7 ; y la mención especial a las para medirlas, como ocurre en los órganos móviles (ovario,
lesiones óseas, a las lesiones quísticas y a las lesiones previa- tracto gastrointestinal), originan discrepancias intraobser-
mente irradiadas. El rastreo óseo, la PET o las radiografías no vador e interobservador8 . La valoración de la respuesta se
son adecuados para medir las lesiones óseas, aunque pueden realizará con la misma técnica empleada en el estudio ini-
ser útiles para confirmarlas o determinar su desaparición. cial, constatando la duración de la respuesta. La suma de las
Las metástasis líticas o mixtas líticas-blásticas son medibles lesiones diana del estudio basal se usará para seguir y valo-
cuando el componente de partes blandas cumpla criterios rar objetivamente la respuesta. Cuando una lesión diana se
de lesión medible. Las blásticas no son medibles (fig. 2). fragmente durante el tratamiento se medirán sus partes, se
Las lesiones quísticas con criterios radiológicos de quiste sumarán, y se considerarán como una única lesión (fig. 3).
simple no son medibles porque no se consideran malignas. Aun con sus limitaciones y excepciones, RECIST es el
Las que correspondan a metástasis quísticas pueden consi- método radiológico sencillo, rápido y fiable más usado8 ,
derarse medibles si cumplen los criterios de lesión medible. para valorar el tamaño tumoral, aunque en ocasiones no se
No obstante, cuando existan otras lesiones no quísticas serán correlacione bien con la situación clínica del paciente.
estas las seleccionadas como lesiones diana. RECIST no con-
templa los cambios de densidad (TC) o de intensidad de señal
(RM) durante el seguimiento. Las lesiones localizadas en Tipos de respuesta
áreas previamente irradiadas o sometidas a cualquier tera-
pia locorregional, deben considerarse lesiones no medibles, Se considera respuesta completa (RC) cuando desaparecen
a menos que se demuestre su progresión. la enfermedad medible y la evaluable, sin que aparezcan
RECIST y el radiólogo
Tabla 1 Resumen comparativo de los parámetros de evaluación según OMS, RECIST 1.0, RECIST 1.1 y PERCIST
OMS RECIST 1.0 RECIST 1.1 PERCIST
Enfermedad medible Como medir Medida bidimensional: Medida unidimensional: Medida unidimensional: diámetro SUV máximo al menos 1,5
producto de los diámetros diámetro máximo máximo veces el de hígado o > 2
perpendiculares máximos veces un ROI en la aorta
torácica descendente
Tamaño mínimo No se especifica tamaño ≥ 10 mm con TC helicoidal ≥ 10 mm con TC helicoidal o RM Determinada por RECIST 1.1
mínimo (no menor que el doble del
espesor de corte)
≥ 20 mm con técnicas ≥ 20 mm en radiografía de tórax
convencionales bien definida y rodeada de aire
Número total de Todas las medibles Máximo 10 lesiones y hasta 5 Máximo 5 lesiones y hasta 2 por Máximo 5 lesiones y hasta 2
lesiones por órgano órgano. por órgano, seleccionando
las de máxima captación
Se incluyen metástasis quísticas,
lesiones óseas líticas con
componente de partes blandas.
Ganglios linfáticos No se especifica No se especifica Normales los ganglios de eje corto
< 10 mm
Ganglios con eje corto ≥ 15 mm
pueden ser lesión diana.
No medibles los de 10 a < 15 mm
Enfermedad no medible Linfangitis, masas abdominales Lesiones óseas, enfermedad Las lesiones inferiores a 10 mm.
palpables no medibles y leptomeníngea, ascitis,
afectación dérmica derrame pleural y pericárdico,
carcinoma inflamatorio de la
mama, linfangitis
carcinomatosa de piel o
pulmón y las masas u
organomegalias abdominales
identificadas en la exploración
clínica no medibles por
métodos de imagen
Adenopatías con eje corto ≥ 10 y
≤ 15 mm.
Lesiones blásticas.
Quistes simples, indeterminados o
complejos.
Lesiones previamente tratadas o
irradiadas a menos que progresen

195
196 J. Cervera Deval

Tabla 2 Criterios de respuesta OMS, RECIST, EORTC-PET, PERCIST 1.0


Tipos OMS RECIST 1.1 EORTC-PET PERCIST 1.0 Choi
RC Desaparición de Desaparición de Ausencia de captación Desaparición completa Desaparición de todas
18
todas las lesiones todas las lesiones F-FDG, indistinguible de captación de 18 F-FDG las lesiones. Sin nuevas
y gánglios del tejido circundante en lesiones diana lesiones
patológicos medibles con actividad
menor a la hepática e
indistinguible del fondo
vascular
RP >50% disminución ≥ 30% de la suma Disminución del 15-25% Disminución al menos Disminución del tamaño
de la suma de las de los diámetros de SUV después de un del 30% del SUV máximo ≥10% o disminución de
áreas máximos ciclo y más del 25% de las lesiones diana la densidad ≥15% de
después de más ciclos medibles Unidades Hounsfield.
Sin nuevas lesiones.
Ausencia de progresión
de la enfermedad no
mensurable
EE No respuesta No respuesta Incremento de < 25% o Ni respuesta metabólica No cumple criterios de
parcial ni parcial ni disminución de < 15% del completa ni respuesta RC, RP o PE.
progresión progresión SUV, sin incremento de metabólica parcial ni
la extensión de la progresión metabólica
captación.
No hay deterioro
sintomático atribuible a
progresión tumoral
PE ≥ 25% de aumento ≥20% incremento Aumento del SUV > 25%. Aumento del SUV > 30% Aumento de tamaño
de la suma de las de la suma de los en lesiones diana. ≥10% y sin criterios de
áreas o aparición diámetros y RP en la densidad del
de una nueva ≥ 5 mm de tumor.
incremento
absoluto en la
suma de diámetros
Aumento > 20% de la Aumento de la extensión Nuevas lesiones.
extensión de la de la captación del
captación. tumor.
Nuevas captaciones Nuevas captaciones Nódulos nuevos
patológicas no intratumorales o
explicables por efecto aumento de los nódulos
del tratamiento o existentes o un aumento
infección de la parte tisular de
una lesión hipodensa.
Fuente: adaptada de Wahl26 y Choi32 .
EE: enfermedad estable; EORTC: European Organisation for Research and Treatment of Cancer; PE: progresión enfermedad; RC: respuesta
completa; RP: respuesta parcial; SUV: valor estandarizado de captación.

nuevas lesiones, con ausencia de síntomas y normalización rar progresión. Así, si, por ejemplo, en el estudio basal una
de marcadores, durante al menos 4 semanas. Hablamos de lesión diana mide 30 mm y en el primer control se reduce a
respuesta parcial (RP) cuando se reduce al menos un 30% la 10 mm (reducción del 67%) se considera RP. Si en el siguiente
suma de los diámetros de las lesiones diana. Hay progresión control mide 13 mm se consideraría EE, ya que además de
de la enfermedad (PE) cuando aumenta el 20%, se pro- aumentar el 20% se requiere el aumento absoluto de 5 mm.
duce un incremento absoluto de al menos 5 mm en la suma Si en el seguimiento posterior aumenta a 16 mm se conside-
de los diámetros de las lesiones diana, o aparecen nuevas raría PE por incrementar su tamaño más de 5 mm y más del
metástasis o progresan las lesiones no diana. La enferme- 20% respecto al menor de los tamaños registrados (10 mm)9 .
dad estable (EE) significa que no hay suficiente reducción
o suficiente incremento para considerarla RP o PE, respec-
tivamente. Se define como respuesta global (RG) a la mejor Volumen tumoral
respuesta después de iniciado el tratamiento antes de que
recidive o progrese la enfermedad (tablas 2---4). Se ha propuesto utilizar el volumen tumoral10,11 para evaluar
Es importante tener presente el criterio de aumento la respuesta, ya que bastantes neoplasias no son esféri-
absoluto de 5 mm en la medida de una lesión para conside- cas, y crecen o se reducen de modo irregular12 , y porque
RECIST y el radiólogo 197

Tabla 3 Combinación de las posibles respuestas en pacientes con lesiones medibles y su mejor respuesta global
Lesiones diana Lesiones no diana Nuevas lesiones Respuesta global Confirmación de mejor
respuesta global
RC RC No RC Sí ≥ 4 semanas
RC No RC/No PE No RP Sí ≥ 4 semanas
RC No evaluable No RP
RP No RC/No PE/No evaluable No RP
EE No RC/No PE/No evaluable No EE Sí. Al menos una vez ≥ 4
semanas desde el estudio basal
PE Cualquiera Sí o No PE No previa EE, RP o RC
Cualquiera PE Sí o No PE
Cualquiera Cualquiera Sí PE
Fuente: adaptada de Van Persijn van Meerten8 .
EE: enfermedad estable; PE: progresión enfermedad; RC: respuesta completa; RP: respuesta parcial.

el volumen proporciona una información de la carga tumo- anti-EGFR [epithelial growth factor receptor], anti Her2,
ral más fiable13 , con una predicción de la respuesta más inhibidores de la vía PI3k/Akt/mTOR, cKIT, terapia hor-
exacta, temprana14 y fácilmente reproducible15,16 . Aunque monal) no se reflejan directamente en la forma y, por
en RECIST no se ha incluido la valoración volumétrica de los tanto, no se pueden valorar adecuadamente con los cri-
tumores, sí contempla las medidas coronales de imágenes terios RECIST17 . Existe una amplia variedad de técnicas
isotrópicas5 . de imágenes funcionales que permiten medir los cambios
fisiopatológicos, metabólicos o bioquímicos18 mediante bio-
Valoración funcional marcadores de imagen médica12,19 , los cuales proporcionan
información, tanto para el diagnóstico como para evaluar la
respuesta20 .
Los cambios metabólicos que inducen en los tumores
los nuevos tratamientos (antiangiogénicos, antivasculares,
Difusión con resonancia magnética

Las técnicas de difusión con RM, basadas en el movi-


miento browniano del agua, han demostrado ser un buen
indicador temprano de la respuesta tumoral21 . General-
mente los cambios en el coeficiente de difusión aparente
(ADC) se correlacionan inversamente con la celularidad. Las
imágenes potenciadas en difusión mejoran la detección y
caracterización de los tumores malignos, ya que estos son
hipercelulares y, por tanto, la difusión está restringida20 .
Un incremento del ADC puede reflejar un incremento en la
movilidad del agua, bien por la pérdida de la integridad de la
membrana, o bien por un incremento en la proporción total
de líquido extracelular, con la correspondiente disminución
del tamaño o número de células, como ocurre en la apoptosis
o necrosis. Por el contrario, una disminución del ADC refleja
una disminución del agua extracelular por un incremento

Tabla 4 Combinación de las posibles respuestas en pacien-


tes con lesiones no medibles y su mejor respuesta global
Lesiones no diana Nuevas lesiones Respuesta global
RC No RC
No RC/No PE No No RC/No PE
Figura 2 Carcinoma de células renales con lesión lítica (flecha
No evaluable No No evaluable
negra) en el hueso ilíaco derecho y con línea de medida inclu-
PE inequívoca Sí o No PE
yendo el componente de partes blandas en la imagen axial de
Cualquiera Sí PE
TC con contraste intravenoso y ventana de partes blandas (A).
Lesiones blásticas en ambos huesos ilíacos y L4 por adenocarci- Fuente: adaptada de Van Persijn van Meerten8 .
noma de próstata en la imagen axial de la pelvis con ventana EE: enfermedad estable; PE: progresión enfermedad; RC: res-
ósea (B). puesta completa; RP: respuesta parcial.
198 J. Cervera Deval

Figura 3 Medida del diámetro máximo (1) en una lesión diana (A) que tras el tratamiento se fragmenta (B). Para su valoración debe
sumarse el diámetro máximo de cada uno de las partes escindidas (en B, 1 + 2 + 3). En el caso contrario, de producirse coalescencia
se mide el máximo de la lesión resultante (1 en A).

Figura 4 Glioblastoma multiforme, estudio basal y tras quimioterapia con fármaco antiangiogénico (bevacizumab) y radioterapia
esteroatáxica fraccionada. En el estudio basal se aprecia la tumoración parafalciana en la imagen axial potenciada en T1 con
contraste intravenoso (A). En la imagen del coeficiente de difusión aparente (B) se aprecia una restricción de la difusión en la parte
sólida, y en el estudio de espectroscopia por RM 1 H muestra un cociente colina/N-acetil aspartato (Cho/NAA) de 3,7 (C), propio de
una tumoración glial de alto grado. El estudio axial T1 con contraste tras el tratamiento muestra una reducción de la tumoración
(D), aumento del ADC respecto al estudio basal (E) ----nótese las áreas de desmielinización debidas a la radioterapia---- y el cociente
Cho/NAA es 0,86 (F). Todo ello sugiere respuesta al tratamiento con disminución del metabolismo y el aumento de producción de
membranas celulares (principal responsable del metabolito Cho en el estudio de espectroscopia).
RECIST y el radiólogo 199

del número o del tamaño de las células, como ocurre en la


fibrosis, en el edema o en la progresión del tumor, lo que
la hace una herramienta muy útil para valorar precozmente
los efectos moleculares y, celulares de los tratamientos17,22
(fig. 4).

Perfusión

La perfusión tumoral19 permite observar cómo el contraste


inyectado se difunde al espacio extravascular extracelular,
lo que aporta información de la permeabilidad endote-
lial y, del volumen del espacio intersticial intercambiable,
y permite valorar la fisiología tisular y microvascular.
Con esta técnica se evalúa la respuesta basada en la
neoangiogénesis (fig. 5), considerando como respuesta la
reducción de los valores relativos de volumen (rCBV)
y flujo tumoral (rCBF). Así, por ejemplo, para dife-
renciar una radionecrosis de la persistencia tumoral en
las metástasis cerebrales, se estima que un rCBV > 2
respecto a la sustancia blanca cerebral sugiere permanencia
tumoral.

Espectroscopia con resonancia magnética

La espectroscopia con resonancia magnética (eRM) de pro-


tón 1 H es una técnica robusta, para uso clínico en el cerebro,
la mama y la próstata17 . Se basa en detectar los metaboli-
tos presentes o aumentados en el tumor y, su disminución
o desaparición durante el seguimiento. Los metabolitos con
importancia biológica son el ATP, la colina (Ch), la creatina
(Cr), el lactato, el citrato (Ci) y el N-acetil-aspartato (NAA).
En el cerebro, la combinación de RM y eRM puede ayu-
dar a evaluar el tipo de tumor cerebral y el grado, así como
a diferenciar el realce tumoral de otras causas de realce.
El criterio metabólico de la eRM de 1 H se basa en que en
las neoplasias malignas cerebrales hay un aumento de Ch
y descenso de NAA, con aumento del cociente Cho/NAA.
Cuando el tumor responde, disminuye este cociente meta-
bólico, tanto en los tumores cerebrales primarios como en
las metástasis.
En la mama, la disminución precoz de Ch es un buen indi-
cador de respuesta. En la próstata, la Ch, Cr y Ci pueden
predecir la existencia de cáncer en la zona periférica, así
como de recidiva tras la prostatectomía radical, radiotera-
pia o crioterapia.

Tomografía por emisión de positrones Figura 5 Estudio de perfusión de un tumor de la base de len-
gua en el plano axial, antes (A) y después (B) de ser tratado con
La 18 F-FDG es un excelente biomarcador metabólico de la cetuximab. Existe una disminución del producto del área de per-
actividad tumoral23 . El aumento de captación de 18 F-FDG se meabilidad tumoral de 38 ml/100 ml/min a 33 ml/100 ml/min, y
relaciona con el aumento de actividad glicolítica cuantifica- del flujo sanguíneo de 86,8 ml/100 ml/min a 77 ml/100 ml/min,
ble de la mayoría de los tumores malignos, lo que posibilita considerados como enfermedad estable, por descenso de solo
evaluar la respuesta (fig. 6) e identificar precozmente a los el 11,29%.
pacientes que responderán o no24 . Además de los criterios
de respuesta con 18 F-FDG de la European Organisation for Casos especiales de respuesta
Research and Treatment of Cancer (EORTC)25 , hay que des-
tacar a los criterios de respuesta con la PET (PERCIST 1.0), Clásicamente, se ha considerado que una reducción de
para valorar semicuantitativamente la actividad glicolítica tamaño tumoral representaba un signo precoz de mejoría
tumoral mediante el valor estandarizado de captación (SUV) clínica, aun con los errores subjetivos de medida condi-
de 18 F-FDG26 . cionados por la irregularidad del margen tumoral27 , los
200 J. Cervera Deval

realces heterogéneos y, los diferentes protocolos de imagen.


Algunos autores28 consideran que la reducción del tamaño
tumoral no representa siempre respuesta y, no predice la
supervivencia global de la enfermedad, por lo que propo-
nen emplear la no progresión como predictor de respuesta.
Así, el carcinoma de pulmón no microcítico, responde
ocasionalmente cambiando la densidad del tumor sin modi-
ficar sustancialmente el tamaño, lo que en RECIST no se
consideraría como respuesta29 .
Las nuevos quimioterápicos y los procedimientos inter-
vencionistas de las lesiones focales30 se dirigen a detener
el crecimiento. En ocasiones, estos tratamientos provo-
can que los tumores puedan aumentar de volumen y, en
cambio, tener una excelente respuesta clínica31 . Así, por
ejemplo, las metástasis del tumor del estroma gastrointes-
tinal tratado con imatinib pueden aumentar de tamaño y
disminuir la densidad (fig. 7), lo que es un criterio de res-

Figura 6 (A) Imagen axial potenciada en T1 con contraste y


saturación grasa donde se aprecia una metástasis cerebelosa
izquierda; (B) estudio PET- F18 FDG donde se muestra la acti-
vidad metabólica en esta localización. Tras la radiocirugía se
aprecia un tamaño similar de la lesión en RM, aunque con cam-
bios internos (C) y desaparición de la actividad metabólica en Figura 7 GIST rectal que en el estudio basal presenta 2 metás-
el estudio de PET (D). tasis, la mayor de ellas con 49 UH de densidad (A), que en un
control 6 meses después (B) presenta aumento de tamaño y dis-
minución de densidad a 29 UH, considerándose como criterio de
respuesta propuesto por Choi32 ya que disminuye más del 15%.
RECIST y el radiólogo 201

puesta aceptado, descrito por Choi32 (tabla 2). Criterios primera indicación para valorar la respuesta, este no es
similares se aplican en metástasis hepáticas tratadas con necesario si al paciente se le ha hecho antes un estudio de
radioembolización33,34 , a sarcomas de partes blandas tra- PET37 .
tados con quimioterapia y radioterapia preoperatoria, y al Para el glioma cerebral se aplicaban los criterios de res-
carcinoma de células renales tratado con inhibidores de la puesta de MacDonald et al.38 en 1990 (tabla 5), los cuales
tirosin-cinasa35,36 . presentaban numerosas limitaciones39,40 en los pacientes
En el cáncer de esófago, la PET tiene un papel importante tratados con radioterapia y temozolamida, o con antiangio-
en el diagnóstico y la estadificación, y, además, puede pre- génicos que afectan la permeabilidad vascular del tumor,
decir qué tumor responderá al tratamiento. De este modo, especialmente los de mecanismo de acción sobre el factor
las guías de la National Comprehensive Cancer Network, de crecimiento del endotelio vascular (VEGF: bevacizu-
aunque recogen que el estudio de TC con contraste es la mab) y en el receptor VEGF (cediranib). Estos tratamientos

Tabla 5 Criterios de respuesta en gliomas


RECIST 1,1 Macdonald (2D) RANO
RC Desaparición de todos los Desaparición de todos los realces Desaparición completa de todas las lesiones
realces tumorales. tumorales. Confirmado a las 4 que captan contraste, sean medibles o no,
Confirmado a las 4 semanas semanas. No nuevas lesiones. No mantenidas al menos 4 semanas. No nuevas
corticoides y estabilidad o mejoría lesiones. Estabilidad o reducción de las
clínica. lesiones hiperintensas en T2/FLAIR que no
captan contraste. Ausencia de corticoides y
mejoría clínica*
RP ≥ 30% disminución de la ≥ 50% disminución en la suma de los ≥ 50% disminución de la suma de productos de
suma de los diámetros productos de los diámetros máximos los diámetros transversos de las lesiones
máximos. Confirmado a las perpendiculares de todas las lesiones medibles que capten contraste, mantenido al
4 semanas que realzan. Confirmado a las 4 menos 4 semanas.
semanas. No nuevas lesiones.
Reducción o estabilidad de dosis de
corticoides y estabilidad o mejoría
clínica.
No hay nuevas lesiones. Estabilidad o
reducción de las lesiones hiperintensas en
T2/FLAIR que no captan contraste.
Mejoría o estabilidad clínica con menor dosis
de corticoides.
No hay progresión de lesiones no medibles
EE Ausencia de RC, RP, PE Ausencia de RC, RP, PE y estabilidad No cumple los criterios de RC, RP o PE.
clínica Estabilidad o reducción de las lesiones
hiperintensas en T2/FLAIR que no captan
contraste.
Dosis de esteroides igual o inferior a la del
estudio basal.
PE ≥ 20% incremento en la ≥ 25% incremento en la suma de los Aumento ≥ 25% de tamaño de la suma de
suma de los diámetros productos de diámetros productos de los diámetros perpendiculares de
máximos. Confirmado a las perpendiculares de lesiones que las lesiones que captan contraste. Aumento de
4 semanas realzan, o nuevas lesiones o las lesiones no medibles
deterioro clínico
Dosis de esteroides igual o superior a la que
tomaba al realizar la RM basal.
Aumento significativo de las lesiones no
captantes
Aparición de alguna lesión nueva.
Deterioro clínico no atribuible a otras causas
que no sea directamente al tumor o a cambios
en la dosis de esteroides
Fallecimiento o deterioro clínico severo
*Pacientes sin lesiones medibles la mejor
respuesta es EE
Fuente: adaptado de Wen39 .
202 J. Cervera Deval

pueden producir una marcada disminución de la capta-


ción de contraste en la fase temprana del tratamiento (1
o 2 días), sin que esto signifique respuesta radiológica-
farmacológica. Los cambios deben permanecer al menos 4
semanas para considerarlos como verdadera respuesta. Con
el fin de soslayar estas limitaciones y estandarizar la valo-
ración de la respuesta en ensayos clínicos, se ha propuesto
una actualización por parte de un amplio grupo de exper-
tos de los criterios de respuesta conocidos como «Response
Assessment in Neuro-Oncology Working Group» (Criterios
RANO)39 . Su aportación es definir las lesiones medibles como
imágenes cuantificables y bien delimitadas, con diámetros
perpendiculares máximos de al menos 10 mm de diámetro en
2 o más imágenes (espesor de corte: 5 mm); y como lesio-
nes no medibles, a las mal definidas o predominantemente
quísticas, visibles solo en un eje, o menor de 10 mm (fig. 8)
y, a las cavidades posquirúrgicas.
En el seguimiento de linfomas se recomienda la PET por
discriminar mejor que la TC a los pacientes que puedan
recaer y a los que tienen posibilidades de remisión41 (fig. 9).

Figura 9 Linfoma de Hodgkin, esclerosis nodular, con adeno-


patías mediastínicas (A) hipermetabólicas en el estudio basal de
PET (B), que tras 6 ciclos de quimioterapia tiene una respuesta
completa (C).

El estudio de PET debe realizarse de 8 a 12 semanas des-


pués de la radioterapia o quimioterapia, ya que procesos
inflamatorios pueden observarse hasta 2 semanas después
de la quimioterapia.
En el carcinoma hepatocelular los criterios de medida
anatómicos pueden ser erróneos cuando se aplican tera-
pias moleculares, tales como el sorafenib o tratamientos
locorregionales, por lo que se propuso una modificación
Figura 8 Lesión medible con diámetros transversales (A) aun- de los criterios RECIST (mRECIST)42,43 . En ella se define una
que presenta componente necrótico en su interior. Nódulo (B) lesión como evaluable si mide al menos 1 cm en una dimen-
dentro de una cavidad quística mayor de 10 mm (lesión medi- sión, demostrable en la fase arterial del estudio dinámico
ble). La lesión quística debe controlarse como no medible. de TC o RM. Recientemente se ha propuesto un criterio
RECIST y el radiólogo 203

Tabla 6 Criterios mRECIST y RECICL para valoración del hepatocarcinoma


mRECIST RECICL
Lesión evaluable Lesiones clasificadas como medibles Lesiones diana (un máximo de 5)
con criterio RECIST (1 cm o más en
una dimensión)
Método de evaluación Medida unidimensional del tumor Suma de todas las lesiones diana. Medida
viable (lesión realzada en fase bidimensional (las lesiones no realzadas en
arterial) y que sea posible su medida TC dinámico o con depósito de lipiodol son
en el seguimiento medidas como necrosis)
Respuesta completa Desaparición de cualquier realce Necrosis del 100% del tumor o 100% de
arterial en lesiones diana reducción del tamaño
Respuesta parcial Al menos 30% de disminución de la Efecto necrótico o reducción del tamaño
suma de los diámetros de las lesiones del 50% a < 100%
diana viables (realzadas en fase
arterial), tomando como referencia
la suma basal de los diámetros de las
lesiones diana
Enfermedad estable No respuesta parcial ni progresión No respuesta parcial ni progresión
Progresión enfermedad ≥ 20% suma de diámetros de lesiones ≥ 25 de aumento del tumor respecto al
diana viables, tomando como efecto necrotizante o aparición de una
referencia la suma más pequeña de nueva lesión (categorizada en 3 grupos:
los diámetros de lesiones viables lesión solitaria intrahepática, lesiones
recogidas desde el inicio del intrahepáticas múltiples, invasión vascular
tratamiento. Desarrollo de una nueva o diseminación extrahepática)
lesión
Fuente: adaptado de Lencioni42 y Kudo44 .
mRECIST: criterios RECIST modificados; RECICL: criterios de evaluación de respuesta en carcinoma hepático.

de valoración de respuesta (Response Evaluation Criteria Confidencialidad de los datos. Los autores declaran que en
in Cancer of the Liver [RECICL]) (tabla 6), que cuantifica la este artículo no aparecen datos de pacientes.
combinación de las medidas en 2 dimensiones de las lesio- Derecho a la privacidad y consentimiento informado. Los
nes diana44 . Además, incluyen marcadores tumorales y un autores han obtenido el consentimiento informado de los
protocolo de valoración en los casos tratados con qui- pacientes y/o sujetos referidos en el artículo. Este docu-
mioembolización arterial, así como recomendaciones de mento obra en poder del autor de correspondencia.
seguimiento según el tipo de tratamiento.
Conflicto de intereses
Conclusión El autor declara no tener ningún conflicto de intereses.

Aunque el parámetro más usado para valorar la respuesta


al tratamiento es la reducción tumoral, no siempre una Bibliografía
reducción del volumen tumoral se acompaña de una mejora
1. Real Academia Española Diccionario de la Lengua Española. 22.a
de la supervivencia. Las discrepancias entre los criterios
ed. Madrid: Espasa-Calpe; 2001.
anatómicos y metabólicos de respuesta, los fallos de pre- 2. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,
dicción, la variabilidad interobservador y la utilidad de Rubinstein L, et al. New guidelines to evaluate the response to
valoraciones volumétricas, hacen que quizás deban revisarse treatment in solid tumors. European Organization for Research
los criterios RECIST. Probablemente, la combinación de bio- and Treatment of Cancer, National Cancer Institute of the Uni-
marcadores de imagen anatómica y funcional refleje mejor ted States, National Cancer Institute of Canada. J Natl Cancer
la respuesta tumoral a los tratamientos. La utilización Inst. 2000;92:205---16.
clínica de biomarcadores funcionales requiere una estan- 3. Benjamin RS, Choi H, Macapinlac HA, Burgess MA, Patel SR,
darización, una vez hayan podido ser validados en ensayos Chen LL, et al. We should desist using RECIST, at last in GIST. J
Clin Oncol. 2007;25:1760---4.
multicéntricos.
4. World Health Organization WHO Handbook for Reporting
Results of Cancer Treatment Offset Publication. Geneva.
Switzerland, 1979 [consultado 3 Sep 2011]. Disponible en:
Responsabilidades éticas http://whqlibdoc.who.int/publications/9241700483.pdf.
5. Eisenhauer EA, Therase P, Bogaerts J, Schwartz LH,
Protección de personas y animales. Los autores declaran Sargent D, Ford R, et al. New response evaluation crite-
que para esta investigación no se han realizado experimen- ria in solid tumours: revised RECIST guideline (version 1.1). Eur
tos en seres humanos ni en animales. J Cancer. 2009;45:228---47.
204 J. Cervera Deval

6. Wahl RL, Jacene H, Kasamos Y, Lodge MA. From RECIST to PER- histopatholgically responding and non responding tumors. J Nucl
CIST: evolving considerations for PET response criteria in solid Med. 2008;49:1038---46.
tumors. J Nucl Med. 2009;50 Suppl. 1:S122---30. 25. Young H, Baum R, Cremerius U, Herholz K, Hoekstra O,
7. Schwartz LH, Bogaerts J, Ford R, Shanka L, Therasse P, Lammertsma AA, et al. Measurement of clinical and sub-
Gwyther S, et al. Evaluation of lymph nodes with RECIST 1.1. clinical tumour response using[18 F]-fluorodeoxyglucose and
Eur J Cancer. 2009;45:261---7. positron emission tomography: Review and1999 EORTC recom-
8. van Persijn van Meerten EL, Gelderblom H, Bloem JL. RECIST mendations reproducibility of quantitative measurements for
revised: implications for the radiologist. A review article on the evaluating 2-[F-18]-fluoro-2-deoxy-d-glucose uptake at PET. Eur
modified RECIST guideline. Eur Radiol. 2010;20:1456---67. J Cancer. 1999;35:1773---82.
9. Nishino M, Jagannathan JP, Ramaiya N, Van den 26. Wahl RL, Jacebe H, Kasamon Y, Lodge MA. From RECIST to PER-
Abbeele AD. Revised RECIST guideline version 1.1: what CIST: evolving considerations for PET response criteria in solid
to know and what radiologist need to know. AJR Am J tumors. J Nucl Med. 2009;50:122S---50S.
Roentgenol. 2010;195:281---9. 27. Erasmus JJ, Gladish GW, Broemeling L, Sabloff BS,
10. Lee SM, Kim SH, Lee LM, Im SA, Bang YJ, Kim WH, et al. Use- Truong MT, Herbst RS, et al. Interobserver and intraob-
fulness of CT volumetry for primary gastric lesions in predicting server variability in measurement if non-small-cell carcinoma
pathologic response to neoadjuvant chemotherapy in advanced lung lesions: implications for assessment of tumor response. J
gastric cancer. Abdom Imaging. 2009;34:430---40. Clin Oncol. 2003;21:2574---82.
11. Baghi M, Mack MG, Hambek M, Bisdas S, Muerthel R, 28. Tuma RS. Somtimes size doesn’t matter: reevaluating RECIST
Wagenblast J, et al. Usefulness of MRI volumetric evaluation in and tumor response rate endpoints. J Natl Cancer Inst.
patients with squamous cell cancer of the head and neck treated 2006;98:1272---4.
with neoadjuvant chemotherapy. Head Neck. 2007;29:104---8. 29. Pallardó Calatayud Y, Cervantes Ruipérez A, Revert Ventura AJ.
12. Yaghmai V, Miller FH, Rezai P, Benson AB, Salem R. Response Estadificación del cáncer y valoración de la respuesta al tra-
to treatment series: Part 2, tumor response assessment- tamiento. En: Actualizaciones SERAM: Imagen en Oncología.
using new and conventional criteria. AJR Am J Roentgenol. Madrid: Ed. Médica Panamericana; 2009. p. 1-12.
2011;197:18---27. 30. Park MH, Rhim H, Kim YS, Choi D, Lim HK, Lee WJ. Spectrum
13. Tran LN, Brown MS, Goldin JG, Yan X, Pals RC, McNitt-Gray MF, of CT findings after radiofrequency ablation of hepatic tumors.
et al. Comparison of treatment response classifications between Radiographics. 2008;28:379---92.
unidimensional, bidimensional and volumetric measurements of 31. Riaz A, Miller FH, Kulil LM, Nicolaidis P, Yaghmai V,
metastatic lung lesions on chest computed tomography. Acad Lewandowski RJ, et al. Imaging response in the primary index
Radiol. 2004;11:1355---60. lesion and clinical outcomes following transarterial locorre-
14. Curran SD, Muellner AU, Schwartz LH. Imaging response assess- gional therapy for hepatocellular carcinoma. JAMA. 2010;303:
ment in oncology. Cancer Imaging. 2006;6:S126---30. 1062---9.
15. Buerke B, Puesken M, Müter S, Weckesser M, Gerss J, Heindel 32. Choi H, Charnsangavej C, de Castro Faria S, Tamm EP,
W, et al. Measurement accuracy and reproducibility of semiau- Benjamín RS, Johnson MM, et al. CT evaluation of the response
tomated metric and volumetric lymph node analysis in MDCT. of gastrointestinal stromal tumors after imatinib mesylate
AJR Am J Roentgenol. 2010;195:979---85. treatment a quantitative analysis correlated with FDG PET fin-
16. Jaffe CC. Measures of response: RECIST, WHO, and new alter- dings. AJR Am J Roentgenol. 2004;183:1619---28.
natives. J Clin Oncol. 2006;24:3245---51. 33. Lewandowski RJ, Geschwind JF, Liapi E, Salem R. Transcathe-
17. García Figueiras R, Padhani AR, Vilanova JC, Goh V, Villalba ter intraarterial therapies: rationale and overview. Radiology.
Martín C. Imagen funcional. Parte 2. Radiología. 2010;52: 2011;259:641---57.
208---20. 34. Sato KT, Lewandowski RJ, Mulcahy MF, Atassi B, Ryu RK,
18. Sullivan DC, Gatsonis C. Response to treatment series: Part I and Gates VL, et al. Unresectable chemorefractory liver metasta-
introduction, measuring tumor response-challenges in the era ses: radioembolization with 90Y microspheres- safety, efficacy,
of molecular medicine. AJR Am J Roentgenol. 2011;197:15---7. and survival. Radiology. 2008;247:507---15.
19. Martí-Bonmatí L, Sanz Requena R, Alberich Bayarri A. Nue- 35. Katabathina VS, Lassau N, Pedrosa I, Ng CS, Prasad SR. Eva-
vos horizontes en la imagen en oncología. En: Actualizaciones luation of treatment response in patients with metastatic renal
SERAM: Imagen en Oncología. Madrid: Ed. Médica Panameri- cell carcinoma: role of state-of-the-art cross-sectional imaging.
cana; 2009. p. 13-22. Curr Urol Rep. 2012;13:70---81.
20. García Figueiras R, Padhani AR, Goh VJ, Vilanova JC, Baleato 36. Motzer RJ, Michaelson MD, Redman BG, Hudes GR, Wilding G,
González S, Villalba Martín C, et al. Novel oncologic drugs: Figlin RA, et al. Activity of SU11248, a multitargeted inhibitor of
what they do and how they affect images. Radiographics. vascular endotelial growth factor receptor and platelet-derived
2011;31:2059---91. growth factor receptor, in patients with metastatic renal cell
21. Malayeri AA, El Khouli RH, Zaheer A, Jaobs M, Corona- carcinoma. J Clin Oncol. 2006;24:16---24.
Villalobos CP, Kamel IR, et al. Principles and applications of 37. Kwee RM. Prediction of tumor response to neoadjuvant therapy
diffusion-weighted imaging in cancer detection, staging, and in patients with esofageal cancer with use of 18 F FDG PET: a
treatment follow-up. Radiographics. 2011;31:1773---91. systematic review. Radiology. 2010;254:707---17.
22. Hamstra DA, Rehemtulla A, Ross BD. Diffusion magnetic reso- 38. MacDonald DR, Cascino TL, Schold SC, Caimcross JG. Response
nance imaging: a biomarker for treatment response in oncology. criteria for phase II studies of supratentorial malignant glioma.
J Clin Oncol. 2007;25:4104---9. J Clin Oncol. 1990;8:1277---80.
23. Shankar LK, Hofman JM, Bacharach S, Grahan MM, Karp J, 39. Wen PY, MacDonald DR, Reardon DA, Cloughesy TF, Sorensen AG,
Lammertsma AA, et al. Consensus recommendations for the Galanis E, et al. Updated response assessment criteria for high-
use of 18 F-FDG PET as an indicator of therapeutic response grade gliomas: response assessment in neuro-oncology working
in patients in National Cancer Institute trials. J Nucl Med. group. J Clin Oncol. 2010;28:1963---72.
2006;46:1059---66. 40. Henson JW, Ulmer S, Harris GJ. Brain tumor imaging in clinical
24. Benz MR, Evilevitch V, Allen-Auerbach MS, Eilber FC, trials. AJNR Am J Neuroradiol. 2008;29:419---24.
Phelps ME, Czernin J, et al. Treatment monitoring by 18 F-FDG 41. Juweid ME, Stoobants S, Hoekstra OS, Mottaghy FM,
PET/CT in patients with sarcomas: Interobserver variability Dietlein M, Guermazi A, et al. Use of positron emission tomo-
of quantitative parameters in treatment-induced changes in graphy for response assessment of lymphoma: consensus of the
RECIST y el radiólogo 205

Imaging Subcommittee of International Harmonization Project therapies in hepatocellular carcinoma: are response evaluation
in lymphoma. J Clin Oncol. 2007;25:571---8. criteria in solid tumors reliable. Cancer. 2009;115:616---23.
42. Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment 44. Kudo M, Kubo S, Takayasu K, Sakamoto M, Tanaka M, Ikai I, et al.
for hepatocellular carcinoma. Semin Liver Dis. 2010;30:52---60. Response evaluation criteria in cancer of the liver (RECICL) pro-
43. Forner A, Ayuso C, Varela M, Rimola J, Hessheimer AJ, de posed by the liver cancer study group of Japan (2009 revised
Lope CR, et al. Evaluation of tumor response after locorregional version). Hepatol Res. 2010;40:686---92.
European Journal of Cancer xx (2016) 1e6

Available online at www.sciencedirect.com

ScienceDirect

journal homepage: www.ejcancer.com

Current Perspective

RECIST 1.1dUpdate and clarification: From the


RECIST committee

Lawrence H. Schwartz a,b,*, Saskia Litière c, Elisabeth de Vries d,


Robert Ford e, Stephen Gwyther f, Sumithra Mandrekar g,
Lalitha Shankar h, Jan Bogaerts c, Alice Chen i, Janet Dancey r,
Wendy Hayes j, F. Stephen Hodi k, Otto S. Hoekstra l, Erich P. Huang m,
Nancy Lin k, Yan Liu c, Patrick Therasse n, Jedd D. Wolchok o,p,q,
Lesley Seymour r

a
Department of Radiology, Columbia University Medical Center, New York, NY, USA
b
New York Presbyterian Hospital, New York, NY, USA
c
EORTC HQ, Brussels, Belgium
d
Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
e
Clinical Trials Imaging Consulting, NJ, USA
f
Department of Medical Imaging, East Surrey Hospital, Redhill, Surrey, UK
g
Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
h
Clinical Trials Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute,
Bethesda, MD, USA
i
Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute,
Bethesda, MD, USA
j
Exploratory Clinical & Translational Research, Bristol-Myers Squibb, Princeton, NJ, USA
k
Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
l
Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
m
National Cancer Institute, National Institutes of Health, Rockville, MD, USA
n
Laboratoires Servier, Paris, France
o
Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
p
Weill Cornell Medical and Graduate Colleges, New York, NY, USA
q
Ludwig Institute for Cancer Research, New York, NY, USA
r
Canadian Cancer Trials Group, Queen’s University, Kingston, Canada

Received 25 March 2016; accepted 27 March 2016

* Corresponding author: CUMC, 180 Fort Washington Avenue, New York, NY 10032, USA. Tel.: þ1 212 305 8994; fax: þ1 212 385 4835.
E-mail address: LSchwartz@columbia.edu (L.H. Schwartz).

http://dx.doi.org/10.1016/j.ejca.2016.03.081
0959-8049/ª 2016 Published by Elsevier Ltd.

Please cite this article in press as: Schwartz LH, et al., RECIST 1.1dUpdate and clarification: From the RECIST committee, European Journal
of Cancer (2016), http://dx.doi.org/10.1016/j.ejca.2016.03.081
2 L.H. Schwartz et al. / European Journal of Cancer xx (2016) 1e6

KEYWORDS Abstract The Response Evaluation Criteria in Solid Tumours (RECIST) were developed
RECIST; and published in 2000, based on the original World Health Organisation guidelines first pub-
Clarifications; lished in 1981. In 2009, revisions were made (RECIST 1.1) incorporating major changes,
Tumour response including a reduction in the number of lesions to be assessed, a new measurement method
to classify lymph nodes as pathologic or normal, the clarification of the requirement to
confirm a complete response or partial response and new methodologies for more appropriate
measurement of disease progression. The purpose of this paper was to summarise the ques-
tions posed and the clarifications provided as an update to the 2009 publication.
ª 2016 Published by Elsevier Ltd.

1. Introduction an adjunct to the gold standard end-points of survival


and quality of life, the RECIST Working Group con-
World Health Organisation response guidelines were tinues to build data warehouses (e.g. data from clinical
first published in 1981 [1,2]. The Response Evaluation trials involving targeted agents and immunotherapies,
Criteria in Solid Tumors (RECIST) criteria were based FDG-positron emission tomography [PET]/computed
on those criteria and were themselves updated in a 2009 tomography [CT] data) to review the criteria, update
in the European Journal of Cancer (RECIST 1.1) [3]. The them periodically as required and validate any changes
revised guidelines incorporated major changes to the in a standardised, methodical manner in response to
original RECIST criteria [2], including a reduction in the both therapeutic and imaging technology advances.
number of lesions to be assessed, a new measurement Critically, the global oncology community must be able
method to classify lymph nodes as pathologic or to implement and adopt any changes proposed to
normal, the clarification of the requirement to confirm a RECIST in a timely and cost-effective manner.
complete response (CR) or partial response (PR) and Since the original publication, users have submitted
new recommendations for the assessment of disease questions on the use and interpretation of the guidelines
progression. Supplementary information provided through the RECIST website (http://www.eortc.org/
included imaging guidelines, which better defined image recist/). These questions are reviewed by the Steering
acquisition and interpretation. Committee and replies provided based on the 2009 pub-
The RECIST criteria have gained widespread adop- lication. If not addressed in the publication, the Working
tion and are widely used in oncology clinical trials. The Group is consulted to prepare appropriate replies. Rele-
RECIST 1.1 paper has been cited 3881 times as of vant and/or frequently asked questions (FAQs) are posted
December 2015 Web of Science. End-points categorised on a regular basis on the RECIST website.
by the RECIST criteria have been used as either primary Here we summarise the questions and clarifications
or supportive data for regulatory approval of new posed as an update to the 2009 publication.
therapeutics by both the Food and Drug Administration
and European Medicines Agency (EMA) [4]. RECIST 2. Commonly asked questions regarding RECIST 1.1
provides a standardised set of rules for response
assessment using tumour shrinkage, based upon imaging 1. What is the frequency of tumour evaluation?
modalities that are globally available and interpretable
by most clinicians. This standardisation, and the rules The schedule and frequency of tumour response re-
and criteria established, provide a framework for evaluation is protocol specific and is based on the
reproducible analysis and reporting of changes in therapeutic, the disease, the anticipated time to response
tumour size. The reproducibility of these criteria and the and progression, as well as practical considerations,
correlations with historical trial results serve an impor- such as cost and patient convenience.
tant purpose in drug discovery. However, in the context of phase II studies where the
Despite the widespread acceptance of RECIST, the beneficial effect of therapy is not known, follow-up
RECIST Working Group continues its work. RECIST every 6e8 weeks (timed to coincide with the end of a
and RECIST 1.1 were developed and tested using data cycle) is reasonable. Shorter or longer time intervals
from clinical trials testing cytotoxic drugs. In the last than these could be justified in specific regimens or cir-
decade, there have been substantial changes in the cumstances. The protocol should specify which areas
mechanism of action of cancer therapeutics (targeted (chest, abdomen) and organs are to be evaluated at
agents, immunotherapies), as well as advances in imag- baseline (usually those most likely to be involved with
ing and clinical trial design and end-points. Although metastatic disease for the tumour type under study) and
the majority of clinical trials continue to use RECIST as how often evaluations should be repeated [2]. The

Please cite this article in press as: Schwartz LH, et al., RECIST 1.1dUpdate and clarification: From the RECIST committee, European Journal
of Cancer (2016), http://dx.doi.org/10.1016/j.ejca.2016.03.081
L.H. Schwartz et al. / European Journal of Cancer xx (2016) 1e6 3

method of assessment should be stated and, ideally, lesions. Other involved lymph nodes should be assessed
repeated using the same imaging technique, equipment and followed as non-target lesions. The analysis of data
and assessor each time. Additional assessments should from the RECIST warehouse was performed in this
be performed if there is suspicion of new site of metas- manner.
tasis or of disease progression based upon clinical
symptoms. The presence of a new lesion(s) should be 6. Should double the slice thickness/interval be applied to lymph
documented on an imaging study. All potential sites of nodes as well when a CT slice thickness of 10 mm is used?
metastases should be evaluated at each time point rather
than following only sites of disease identified at baseline. It is strongly recommended that CT slice thickness of
5 mm be used. Although RECIST 1.1 (2009) recom-
2. Should lesions smaller than 5 mm be reported as the actual mends the following: ‘As is described in Appendix II,
size or reported as a default value of 5 mm? when CT scans have slice thickness greater than 5 mm,
the minimum size for a measurable lesion should be
All lesions, both nodal and non-nodal, must be twice the slice thickness’, in 2016 contemporary CT
evaluated, accurately measured (if measurable/target) scanners globally should be able to acquire images with
and recorded at all time points. If a lesion is no longer a slice thickness of 5 mm or less. This is recommended
seen, it should be recorded as zero (or absent if non- by the Quantitative Imaging Biomarkers Alliance
measurable/non-target). If a lesion is smaller than 5 mm guidelines for standardising image acquisition for CT
and the radiologist believes the lesion can be accurately (http://rsna.org/QIBA_.aspx) [5]. There are many dis-
measured, then the actual size should be recorded. advantages and no real advantages in obtaining 10-mm
It is recognised that lesions become small and ill slices. Not only are lesions more difficult to measure, but
defined on a CT scan such that the radiologist cannot new lesion conspicuity is significantly less at 10 mm [6,7].
accurately measure them. Some radiologists use the term
‘too small to measure’ to describe this phenomena. 7. When lymph nodes coalesce forming a conglomerate mass,
When this occurs, the radiologist may decide that the which axis should be measured to assess the response: short
lesion is present but he/she does not feel comfortable or long axis?
providing the oncologist with a precise measurement. If
this occurs, the radiologist may assign the lesion a value The short axis of lymph nodes should always be
of 5 mm by default. This default value is nominally measured. As nodal lesions coalesce, a plane between
derived from the 5-mm CT slice thickness (but should them may be maintained that would aid in obtaining
not be changed with varying CT slice thickness). The maximal short-axis diameter measurements of each in-
measurement of this type of lesion is potentially non- dividual lesion. If the nodal lesions have truly coalesced
reproducible, and providing this default value will pre- such that they are no longer separable, the vector of the
vent a false assessment of response or progressive dis- longest diameter in this instance should be used to
ease due to measurement error. determine the perpendicular vector for the maximal
short-axis diameter of the coalesced lesion (Fig. 1). Non-
3. If there are three or more measurable lesions in one organ, nodal lesions that coalesce should similarly be assessed
and we select two of them as target lesions, how should the by the longest diameter.
third lesion be considered?

The third lesion should be considered a non-target


lesion and should be recorded and followed as part of
the non-target disease.

4. Is a single-target lesion measurable if a patient has multiple


non-target non-measurable disease?

Yes, a single-target lesion is considered measurable


disease, provided the lesion meets the definition of
measurability as described in RECIST 1.1.

5. How should the limitation of two target lesions per organ be


Fig. 1. Note optimal manner for measuring the short axis of
applied to lymph nodes? Can individual chains/regions be
coalescing lymph nodes (blue arrow). At baseline, there are two
considered one organ or are lymph nodes (all locations
distinct nodes; therefore, the short axis is measured for each (red
included) a single organ?
and yellow lines) and at cycle 6 the single-short axis of the coa-
lesced node is now measured as a single line (red) (For interpre-
Lymph nodes are considered one organ. Only two tation of the references to colour in this figure legend, the reader is
lymph nodes should be measured per patient as target referred to the web version of this article.).

Please cite this article in press as: Schwartz LH, et al., RECIST 1.1dUpdate and clarification: From the RECIST committee, European Journal
of Cancer (2016), http://dx.doi.org/10.1016/j.ejca.2016.03.081
4 L.H. Schwartz et al. / European Journal of Cancer xx (2016) 1e6

8. Clarification of the definition of stable disease. criteria for PD must be met as well, such as the
appearance of new lesions or a sum measurement of
The definition of stable disease (SD) is clarified as target lesions that has increased more than 20%.
follows: ‘Neither sufficient shrinkage (compared to It is important to remember that lymph nodes must
baseline) to qualify for CR or PR nor sufficient increase meet the criteria for malignancy (defined pragmatically
(taking as reference the smallest sum of diameters at as 10 mm) in order to be considered to have reap-
baseline or while on study, whichever is smallest) to peared. A node which is less than 10 mm is considered
qualify for progressive disease (PD)’. benign and is not PD. In a subject with CR, a new node
It is important to recognise that the classification of a that meets the size criteria for a pathologic node is
response (either CR or PR) occurs in comparison to the considered a new site of disease and is, therefore,
sum of diameters at baseline, while progression is based consistent with PD.
on a comparison to the smallest of the sum of diameters In subjects with PR or SD:
at baseline or the smallest sum of diameters during the a. A previously abnormal target node that became
trial (nadir). Most protocols require the criteria for SD normal and subsequently enlarged in size meeting the
for a specified period (e.g. at least 4 weeks) before SD criteria for a pathologic and measurable node (a short
can be concluded. Thus, if imaging is conducted at 2 and axis of 15 mm) should be added to the sum of the
4 weeks on-study and at 2 weeks, the criteria for CR, diameters to determine if the criteria for PD are met
PR, or PD are not met, but at 4 weeks meets the criteria based on target lesions.
for PD, the best overall response is PD, not SD as the b. A previously abnormal non-target node that
subject was not on-study long enough to qualify for SD. became normal and subsequently recurred must meet
the criteria for PD based on NT lesions to call
9. If an abnormal lymph node (or non-nodal disease) ‘disap- progression.
pears’ but then ‘reappears’ should this considered to be PD? c. A normal node at baseline that subsequently be-
comes pathologic is considered a new lesion and results
In general, significant lesions that completely regress in PD.
and then reappear are indicative of PD. However, it is In the circumstances illustrated above, where a single
important to consider the patient’s entire tumour pathologic node is driving the progression event,
burden in order to make certain that the patient is not continuation of treatment/follow-up and confirmation
falsely classified as having PD based upon a single by a subsequent examination should be contemplated. If
measurement or lesion, especially when those lesions are it becomes clear that the ‘new node’ has not resolved, or
small or there is a change in optimal imaging assess- has significantly increased in size, and truly represents
ment. This holds true for all types of metastases (Fig. 2). PD, the date of PD would be the date the new node was
If the response was previously considered to be CR, first documented.
with resolution of all sites of disease, then the reappear-
ance of any lesion, or the development of a new lesion 10. How should patients be classified who have had surgery/
considered to be malignant, would generally be considered radiotherapy during trials for which they are being fol-
PD (see comments below regarding lymph nodes). lowed by RECIST?
In the case of PR or SD, if a previously resolved
lesion reappears, then PD should not be assigned purely For most clinical trials using RECIST, surgery or
based on the reappearance of the lesion; rather, other radiotherapy after trial inclusion and prior to disease

Fig. 2. Patient with colorectal cancer. Liver metastases at baseline (red circle) appear to be resolved at cycle 3 with ‘reappearance’ at cycle 5 (red
circle). However, notice that the imaging techniques are different and the cycle 3 is of poor image quality to visualise these metastases.
Therefore, these lesions have not truly reappeared and the patient should not be considered to have progressive disease at cycle 5. There is true
progression at cycle 7 (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.).

Please cite this article in press as: Schwartz LH, et al., RECIST 1.1dUpdate and clarification: From the RECIST committee, European Journal
of Cancer (2016), http://dx.doi.org/10.1016/j.ejca.2016.03.081
L.H. Schwartz et al. / European Journal of Cancer xx (2016) 1e6 5

progression is a protocol deviation, and if a target lesion only be assessed on the existing lesions and their change
has been surgically removed or treated with radio- from baseline to follow-up on CT or MRI. Providing
therapy, then the patient’s response is not evaluable. If the scan was evaluable and all lesions visible/measur-
surgery or radiotherapy is part of the clinical trial, then able, the response would, therefore, be CR, PR or SD as
the protocol must define in advance how response and no new lesions were seen on CT/MRI.
progression will be handled and ‘censored’ in the ana-
lyses. If the treatment has resulted in inoperable lesions 13. Can the CT information from PET-CT be used as the
being suitable for resection, the researcher may wish to basis of CT assessments? Is the technical quality of such
capture that separately as an indicator of ‘activity’. images sufficient for quantification as required in
RECIST?
11. The section on FDG-PET mentions that correlation with
CT is warranted for new lesions and that PD should be At present, the low dose or the attenuation correction
declared if the hot spot on PET corresponds to a pro- CT portion of a combined PET-CT is not of optimal
gressing lesion on CT. How should this correlation be diagnostic CT quality for use with RECIST measure-
made if the hot spot on PET corresponds to a target lesion ments. However, if the site has documented that a CT
that has increased in size but the sum of the measurements with appropriate radiation dose for diagnostic quality
does not show an increase that is sufficient for PD (other and intravenous and oral contrast was used (if not
target lesions have not enlarged or have actually medically contraindicated), the CT portion of the PET-
decreased). Is this PD? CT can be used for RECIST measurements.
The RECIST 1.1 guidelines state ‘a. Negative FDG- 14. Can we use coronal or sagittal imaging to measure lesions
PET at baseline, with a positive FDG-PET at follow- in CT if the largest diameter is in a plane other than axial?
up is a sign of PD based on a new lesion. b. No FDG-
PET at baseline and a positive FDG-PET at follow-up: It is recommended that the axial imaging plane be
If the positive FDG-PET at follow-up corresponds to a used in all cases on CT scans for consistency and for
new site of disease confirmed by CT, this is PD. If the ease of measurement especially since reconstructions or
positive FDG-PET at follow-up is not confirmed as a advanced workstations are not always available glob-
new site of disease on CT, additional follow-up CT ally. It is recognised that the other planes may represent
scans are needed to determine if there is truly pro- the true long axis of the tumour but depending on the
gression occurring at that site (if so, the date of PD will CT acquisition parameters across time points, and this
be the date of the initial abnormal FDG-PET scan). If may be difficult to consistently and reproducibly
the positive FDG-PET at follow-up corresponds to a measure.
pre-existing site of disease on CT that is not pro-
gressing on the basis of the anatomic images, this is
3. Conclusions
not PD’.
Therefore, the scenario is not PD. Currently, PET
The RECIST Working Group is currently testing the
scanning is considered a complementary modality pri-
Criteria for their applicability with modern therapeutics,
marily to assess for new tumour lesions. Therefore, if a
imaging techniques and trial end-points. The current
target lesion is FDG avid and even if the standardised
RECIST 1.1 criteria remain widely used. A number of
uptake value on PET has increased, the patient would
users have asked questions regarding the interpretation
only be considered to be PD if the CT metrics for pro-
of the 2009 Criteria and we have attempted to summa-
gression were met, including a greater than 20% increase
rise those questions, and the answers, in this update. For
in the sum measurement of lesions, non-target un-
further updates on the criteria, please visit the RECIST
equivocal PD or new lesions on CT (regardless of their
website at http://www.eortc.org/recist/
presence or SUV on PET).

12. If there is a hot spot on FDG-PET (baseline PET is not Conflict of interest statement
available) that is not associated with a new CT (or
magnetic resonance imaging [MRI]) lesion, the article E. de Vries: Research grants from Roche/Genentech,
states that PD should not be declared. What should the Amgen, Novartis, Pieris and Servier to the institute,
response be, not evaluable? data monitoring committee Biomarin, advisory board
Synthon. R. Ford: Dr. Ford is not a stockholder in any
FDG-PET is a complementary modality and should pharmaceutical company and holds no options, grants
be used in adjunct to the CT or MRI. Therefore, in this or patents. He is currently or was previously a consul-
example, the patient’s response should be based solely tant for the following companies (either directly through
upon the measurements and findings on CT and/or Clinical Trials Imaging Consulting, LLC., or indirectly
MRI. Since there is no PET at baseline and no corre- through other pharmaceutical service companies for
sponding new lesion on CT or MRI, then the patient can which he consults) including ACR Image Metrix,

Please cite this article in press as: Schwartz LH, et al., RECIST 1.1dUpdate and clarification: From the RECIST committee, European Journal
of Cancer (2016), http://dx.doi.org/10.1016/j.ejca.2016.03.081
6 L.H. Schwartz et al. / European Journal of Cancer xx (2016) 1e6

AbbVie, Amgen, Aptiv, Aragon, Bioclinica, Biomedical References


Systems, Bristol Myers Squibb, Celldex, Celgene, Cel-
sion, Covance, DNAtrix, Eisai, Exelixis, Genentech/ [1] Miller AB, Hoogstraten B, Staquet M, Winkler A. Reporting re-
Roche, ICON Medical Imaging, Image Endpoints, sults of cancer treatment. Cancer 1981;47:207e14.
[2] Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,
Janssen, Kyowa, Merck, Mirati Therapeutics, Novartis,
Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT,
Novocure, Oncothyreon, Ono, Optimer, OrbiMed Ad- Christian MC, Gwyther SG. New guidelines to evaluate the
visors, Pfizer, Radiant Sage, Quintiles, Red Hill Pharma, response to treatment in solid tumors. European Organization for
Sun Advanced Pharma Research Company, Tokai Research and Treatment of Cancer, National Cancer Institute of
Pharmaceuticals, Vascular Biogenics and Virtualscopics. the United States, National Cancer Institute of Canada. J Natl
Cancer Inst 2000;92:205e16.
S. Hodi: Consultant for Merck, Novartis, Genentech,
[3] Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D,
Synta; research support to institution from Bristol- Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L,
Myers Squibb. N. Lin: Research funding for clinical Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response
trials from (no direct salary support): Genentech, GSK, evaluation criteria in solid tumours: revised RECIST guideline
Novartis, Synta, Array Biopharma, Kadmon, and (version 1.1). Eur J Cancer 2009;45:228e47.
[4] Janku F, Berry DA, Gong J, Parsons HA, Stewart DJ,
Puma. J.D. Wolchok: Research grants from Bristol-
Kurzrock R. Outcomes of phase II clinical trials with single-agent
Myers Squibb; advisory board member for Brisol-Myers therapies in advanced/metastatic nonesmall cell lung cancer pub-
Squibb, Merck, Medimmune, Genentech. All authors lished between 2000 and 2009. Clin Cancer Res 2012;18:6356e63.
have no conflicts relevant to the study. LH Schwartz: [5] http://www.rsna.org/uploadedFiles/RSNA/Content/Science_and_
Research grants from Novartis, Astellas, Eli Lilly, Education/QIBA/QIBA_CT%20Vol_
TumorVolumeChangeProfile_v2.2_PubliclyReviewedVersion_
Merck, Pfizer, Consultant to GSK, Novartis.
08AUG2012.pdf.
[6] Zhao B, Tan Y, Bell DJ, Marley SE, Guo P, Mann H, Scott ML,
Acknowledgements Schwartz LH, Ghiorghiu DC. Exploring intra- and inter-reader
variability in uni-dimensional, bi-dimensional, and volumetric
measurements of solid tumors on CT scans reconstructed at
Canadian Cancer Trials Group participation was different slice intervals. Eur J Radiol 2013;82:959e68.
supported by the Canadian Cancer Society Research [7] Sullivan DC, Schwartz LH, Zhao B. The imaging viewpoint: how
Institute (grant #021039). This publication was sup- imaging affects determination of progression-free survival. Clin
ported by the EORTC Cancer Research Fund and by Cancer Res 2013;19:2621e8.
the National Cancer Institute (NCI) grant number
5U10-CA11488-45.

Please cite this article in press as: Schwartz LH, et al., RECIST 1.1dUpdate and clarification: From the RECIST committee, European Journal
of Cancer (2016), http://dx.doi.org/10.1016/j.ejca.2016.03.081
Journal Pre-proof

Impact of acetaminophen on the efficacy of immunotherapy in cancer patients.

A. Bessede, A. Marabelle, J.P. Guégan, F.X. Danlos, S. Cousin, F. Peyraud, N.


Chaput, M. Spalato, G. Roubaud, M. Cabart, M. Khettab, A. Chaibi, C. Rey, I. Nafia,
F.X. Mahon, J.C. Soria, A. Italiano
PII: S0923-7534(22)01208-X
DOI: https://doi.org/10.1016/j.annonc.2022.05.010
Reference: ANNONC 941

To appear in: Annals of Oncology

Received Date: 31 March 2022


Revised Date: 9 May 2022
Accepted Date: 24 May 2022

Please cite this article as: Bessede A, Marabelle A, Guégan J, Danlos F, Cousin S, Peyraud F, Chaput
N, Spalato M, Roubaud G, Cabart M, Khettab M, Chaibi A, Rey C, Nafia I, Mahon F, Soria J, Italiano
A, Impact of acetaminophen on the efficacy of immunotherapy in cancer patients., Annals of Oncology
(2022), doi: https://doi.org/10.1016/j.annonc.2022.05.010.

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.

© 2022 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.
ORIGINAL REPORT

Impact of acetaminophen on the efficacy of immunotherapy in cancer patients.

A Bessede*1; A Marabelle*2; JP Guégan1; FX Danlos2; S Cousin3; F Peyraud3; N Chaput4,5,6 ;

M Spalato3; G Roubaud3; M Cabart3; M Khettab7; A Chaibi3; C Rey3; I Nafia1; FX Mahon3,7; JC

Soria8; A Italiano2,3,7

* These two authors contributed equally to the work

1. Explicyte, 229 cours de l’Argonne, Bordeaux, France

2. Département d’Innovation Précoce et d’Essais Thérapeutiques (DITEP), INSERM

f
oo
U1015 & CIC1428, Université Paris Saclay, Gustave Roussy, Villejuif, France

3. Department of Medicine, Institut Bergonié, Bordeaux, France

r
-p
4. Laboratory of Immunomonitoring in Oncology, Gustave Roussy Cancer Campus,
re
CNRS-UMS 3655 and INSERM-US23, Villejuif, France
lP

5. Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France

6. Laboratory of Genetic Instability and Oncogenesis, UMR CNRS 8200, Gustave


na

Roussy, Université Paris-Saclay, Villejuif, France


ur

7. Faculty of Medicine, University of Bordeaux, Bordeaux, France


Jo

8. Department of Medicine, Gustave Roussy, Villejuif, France

Correspondence to: Dr. Antoine ITALIANO, MD, PhD

DITEP, Gustave Roussy, Villejuif, France

antoine.italiano@gustaveroussy.fr

1
ABSTRACT

Background: Acetaminophen (APAP) use has been associated with blunted vaccine immune

responses. This study aimed to assess APAP impact on immunotherapy efficacy in patients

with cancer.

Patients and Methods: Exposure to APAP was assessed by plasma analysis and was

correlated with clinical outcome in three independent cohorts of patients with advanced cancer

who were treated with immune checkpoint blockers (ICB). APAP immunomodulatory effects

were evaluated on a pre-clinical tumor model and on human peripheral blood mononuclear

f
cells (PBMCs) from healthy donors.

oo
Results: Detectable plasma APAP levels at treatment onset was associated with a significantly

r
worse clinical outcome in ICB-treated cancer patients, independently of other prognostic
-p
factors. APAP significantly reduced ICB efficacy in the pre-clinical MC38 model, as well as the
re
production of PD1 blockade-related interferon-γ secretion by human PBMCs. Moreover,
lP

reduction of ICB efficacy in vivo was associated with significantly increased tumor infiltration
na

by regulatory T cells (Tregs). Administration of APAP over 24 h induced a significant expansion

of peripheral Tregs in healthy individuals. In addition, interleukin-10, a crucial mediator of Treg-


ur

induced immune suppression, was significantly upregulated upon treatment with ICB in cancer
Jo

patients taking APAP.

Conclusion: This study provides strong pre-clinical and clinical evidence of the role of APAP

as a potential suppressor of antitumor immunity. Hence, APAP should be used with caution in

patients treated with ICB.

KEYWORDS: acetaminophen, immune checkpoint inhibitors, immunotherapy, cancer

2
HIGHLIGHTS

Acetaminophen has been shown to blunt the antibody response to vaccination

Acetaminophen may impact efficacy of immunotherapy in patients with cancer

Further research is needed to decipher the impact of acetaminophen on immunity

f
r oo
-p
re
lP
na
ur
Jo

3
INTRODUCTION

Pain is the most common symptom experienced by patients with advanced cancer.

Acetaminophen (APAP, commonly known as paracetamol) alone or in combination with a

weak opioid, such as codeine or tramadol, is usually considered as the first-line strategy to

manage mild-to-moderate pain in this setting1. Although generally considered to be safe,

evidence suggests that APAP may have negative immunomodulatory effects. Indeed,

preclinical studies demonstrated that APAP can inhibit the proliferation of immune cells and

the T cell-dependent antibody response2,3. Moreover, pioneer clinical studies have suggested

f
that APAP inhibits viral clearance and/or neutralizing antibody response in patients infected

oo
with chicken pox or rhinovirus4. More recently, randomized studies have shown that APAP has

r
a negative impact on vaccination response with decreased antibody levels in subjects
-p
receiving APAP for fever prophylaxis5,6. Given its potential to impair vaccine effectiveness, the
re
World Health Organization stated in 2015 that administration of APAP before or at the time of
lP

vaccination is not recommended7.


na

The development of immune checkpoint blockers (ICBs) was a revolutionary milestone in the

field of immuno-oncology. Anticytotoxic T lymphocyte-associated protein-4 (CTLA-4) and anti-


ur

programmed cell death (PD)-1/PD-ligand 1 (PD-L1) therapies, such as ipilimumab, nivolumab


Jo

or pembrolizumab, are now available for treating various malignancies, including non-small

cell lung cancer, melanoma, and bladder cancer8,9. The objective of our study was to

investigate whether APAP may impair the efficacy of ICB in patients with advanced cancer.

4
METHODS

Patients, treatments, and evaluation

Study design, eligibility criteria, and treatment were previously described for the CheckMate

025 trial10 (NCT01668784, sponsor: Bristol Myers Squibb). The main inclusion criteria for the

BIP (NCT02534649, sponsor: Institut Bergonié) and PREMIS studies (NCT03984318,

sponsor: Gustave Roussy) were age ≥ 18 years, histologically proven malignant tumor,

unresectable and/or metastatic disease, and at least one tumor evaluation by imaging after

immunotherapy onset. All patients included in the BIP and PREMIS studies were treated with

f
anti-PD-(L)1 either in monotherapy or combined with anti-CTLA-4 antibodies, either within

oo
clinical trials, or in the context of approved indications of the European Medicine Agency, or

r
within early access programs. The best response to treatment was evaluated according to
-p
Response Evaluation Criteria in Solid Tumors (RECIST) guidelines11. Routine follow-up and
re
treatment beyond progression therapeutic options were similar within the two studies. To
lP

investigate the pharmacodynamic impact of APAP on peripheral immune cells, four healthy
na

volunteers received 1,000 mg of APAP orally every 6 h, over a period of 24 h (total dose: 4 g).

Peripheral blood mononuclear cells (PBMCs) were collected at baseline and 2h after the last
ur

administration of APAP. This study was approved by the Institutional ethics review board of
Jo

the Institut Bergonié (Bordeaux, France). Written informed consent was obtained from all

subjects involved in the study.

Metabolomic profiling of plasma samples from the BIP study

Detection of APAP and of its metabolite APAP glucuronide was performed for the patients

included in the institutional profiling program BIP (NCT02534649; sponsor: Institut Bergonié)

via liquid chromatography–mass spectrometry. Details are provided in Supplementary

Methods.

Quantitative dosage of APAP

5
Quantitative dosage of APAP and of its metabolite APAP glucuronide was performed for the

patients included in the institutional profiling program PREMIS (NCT03984318; sponsor:

Gustave Roussy) via liquid chromatography–mass spectrometry. Details are provided in

Supplementary Methods.

Human PBMC profiling

PBMCs were isolated from whole blood of healthy donors before and 24 h after APAP dosing

using density gradient centrifugation with Lymphoprep (STEMCELL Technologies, Vancouver,

BC, Canada) as per the manufacturer’s instructions. PBMCs (5 × 105 cells) were first stained

f
oo
with Zombie NIR viability kit (Biolegend, San Diego, CA, USA) for 10 min and then blocked for

5 min with Human TruStain FcX (Biolegend) and True-Stain Monocyte Blocker (Biolegend)

r
-p
solutions. Cells were incubated for 15 min at 4 °C with cell surface marker antibodies, washed
re
in MACS buffer (2 mM EDTA, 0.5% bovine serum albumin, 1x phosphate-buffered saline),
lP

fixed for 30 min at 4 °C with Foxp3 Fixation/Permeabilization solution (eBioscience, Waltham,

MA, USA) and permeabilized with Permeabilization buffer (eBioscience) for 5 min at room
na

temperature. The cells were finally incubated with intracellular marker antibodies for 30 min at
ur

4 °C, washed twice, and resuspended in MACS buffer for analysis on the Novocyte Quanteon
Jo

flow cytometer (Agilent Technologies, Santa Clara, CA, USA). Two different antibody panels

were used for PBMC immunophenotyping: 1)

Zombie/CD45/CD3/CD4/CD8/CD19/CD25/CD56/PD1/CTLA4/TIGIT/TIM3/FoxP3/LAG3, and

2) Zombie/CD45/CD3+CD19/CD11c/CD11b/CD14/CD56/HLA-

R/CD123/PDL1/CD15/IDO1/Arginase1 (Supplementary Table 1). To normalize the signals

obtained before and after APAP dosing, PBMCs from a healthy donor were stained in parallel

of the samples and served as shared control for CytoNorm normalization12. Normalization and

cell phenotyping were both performed on FlowJo v10.8 (BD Biosciences, Franklin Lakes, NJ,

USA). Differences in cell populations and marker expression between pre- and post-treatment

samples were tested using a paired Student’s t-test. Only significant changes in marker

expression were illustrated on the heatmaps (pheatmap R package v1.0.12).

6
Plasma proteomics

Proteomic profiling of plasma samples from cancer patients included in the institutional profiling

program PREMIS (NCT03984318; sponsor: Gustave Roussy) was assessed using the Olink

Target 96 Inflammation panel (Olink Proteomics AB, Uppsala, Sweden) according to the

manufacturer's instructions. Details are provided in Supplementary Methods. Differences in

plasma collected at baseline and week 6 were assessed by paired Student’s t-test using the

limma R package (v3.48.3), and proteins with a fold change > 1.25 and a Benjamini-Hochberg-

adjusted p-value < 0.05 were extracted. Venn diagram was drawn using the ggvenn R package

f
(v0.1.9)

oo
In vitro functional assays

r
-p
PBMCs from three different healthy donors were isolated by Lymphoprep density gradient
re
centrifugation. Cells (1 × 105) were seeded in 96-well plates and treated with anti-CD3 (1
lP

µg/mL, Biolegend) with or without nivolumab (1 µg/mL, Selleckchem) along with increasing

doses of APAP (Sigma-Aldrich). After 72 h, supernatants were collected and interferon-γ


na

release was assessed by homogeneous time resolved fluorescence using the Human IFN
ur

gamma kit (Cisbio, Codolet, France).


Jo

Animal studies

All animal studies were performed under protocols approved by the Institutional Animal Care

and Use Committee of the University of Bordeaux (Bordeaux, France). Details are provided in

Supplementary Methods.

Statistical analysis

The cutoff date for statistical analysis of baseline demographic data and clinical outcome was

by 6/30/2021. Descriptive statistics were used to describe the distribution of variables in the

population. Progression-free survival (PFS) was defined as the time from the start of treatment

until disease progression, death, or last patient contact. Overall survival (OS) was defined as

the time from the start of treatment until death or last patient contact. Survival rates were

7
estimated using the Kaplan–Meier method (survival R package, v3.2-13). Patients from the

Checkmate025 and BIP studies were classified either as “High” or “Low” based on a threshold

value (1x105 mass spectrometry intensity) while patients from the PREMIS study were

classified as “Presence” or “Absence” according to the quantitation of APAP and APAP

glucuronide. Differences between groups were evaluated by chi-square test for categorical

variables and ANOVA with Tuckey tests or Wilcoxon tests for continuous variables. Prognostic

factors were planned to be identified by univariate and multivariate analyses using a Cox

regression model. Variables tested in univariate analysis included age, gender, tumor type,

f
presence of liver metastasis, presence of bone metastasis, number of metastatic sites,

oo
antibiotic use, steroid use, performance status, number of previous lines of treatment, lactate

r
dehydrogenase levels, and presence of detectable levels of APAP in the plasma. Variables
-p
associated with PFS and OS with a p-value < 0.05 in the univariate analysis were planned to
re
be included in the multivariate analysis. Analyses were performed in R using the survival
lP

analysis package (v0.2.0). All statistical tests were two-sided, and p  < 0.05 indicated statistical

significance.
na
ur
Jo

8
RESULTS

Since self-medication with APAP is highly prevalent among the general population, analysis of

medical records is not appropriate to accurately determine APAP exposure. We therefore

analyzed the publicly available serum metabolomics data from 297 patients with advanced

renal cell carcinoma and treated with nivolumab in the context of the randomized phase III trial

CheckMate 025 (NCT01668784)10. We found that patients with detectable levels of APAP or

APAP glucuronide had significantly worse OS than patients without detectable APAP levels at

treatment onset (Figure 1A, B and Supplementary Figure 1A). To note, correlations with

f
objective response rate and PFS were not analyzed as these data were not available. We then

oo
used the same untargeted mass spectrometry-based metabolomics approach to analyze

r
plasma samples of 34 patients included in the institutional profiling program BIP
-p
(NCT02534649) and treated with ICB for advanced disease. Their characteristics are
re
described in Supplementary Table 2. Significant levels of APAP or APAP glucuronide were
lP

detected in 50% of these patients. As shown in Figure 1E, patients exposed to APAP had
na

significantly lower objective response rate (0% vs. 29.4%; p = 0.015), and trended to have

worse PFS (median PFS, 1.87 vs. 4.72 months; 95% confidence interval [CI], 0.30–1.32; p =
ur

0.219; Figure 1C) and OS (median OS, 7.87 vs. 16.56 months; 95% CI, 0.3–1.63; p = 0.412;
Jo

Figure 1D) than patients without detectable APAP levels at treatment onset.

To investigate the robustness of these results, we also evaluated the levels of APAP using a

quantitative mass spectrometry approach in plasma samples collected from 297 patients

enrolled in the PREMIS study (NCT03984318). Their characteristics are described in Table 1.

We found that patients with detectable APAP levels had significantly worse PFS (median PFS:

2.63 vs. 5.03 months; 95% CI, 0.53–0.91; p = 0.009; Figure 1F) and OS (median OS: 8.43 vs.

14.93 months; 95% CI, 0.32–0.69; p < 0.0001; Figure 1G) compared with patients with APAP-

free plasma. Objective response rate was also numerically higher in the APAP-negative group

than in the APAP-positive group (28.9% vs 20.7%; p=0.106); Figure (Figure 1H), although

the difference did not reach statistical significance (except in patients with poor performance

9
status, Supplementary Figure 2). On multivariate analysis, APAP plasma levels remained

independently associated with both PFS and OS (Table 2).

To confirm the impact of APAP on ICB efficacy and provide mechanistic insights, we conducted

preclinical investigations using the MC38 colon tumor model, which has been shown to be

responsive to PD-1/PD-L1 blocking antibodies13. Tumor rejection rates were significantly lower

in mice treated with anti-PD-1/PD-L1 antibodies concomitantly with non-toxic doses of APAP

than in mice treated with anti-PD-1/PD-L1 antibodies alone, with a trend for overall survival

benefit (p=0.17), respectively (Supplementary Figure 3A, B). To investigate the mechanisms

f
oo
underlying APAP activity in vivo, we performed a flow-cytometry-based analysis of tumor-

infiltrating leukocytes 13 days after tumor inoculation (8 days after treatment initiation). An

r
-p
increase in tumor infiltration by regulatory T cells (Tregs) was observed in mice treated with

APAP and, to a higher and significant extent, in mice treated with both APAP and ICB
re
(Supplementary Figure 3D).
lP

To evaluate the direct impact of APAP on immune cells, we exposed human PBMCs from
na

healthy donors to anti-CD3 antibodies in the presence or absence of nivolumab (1 µg/mL) with
ur

increasing concentrations of APAP (0, 100, and 300 µM) for 72 h. As expected14,15, nivolumab

increased the anti-CD3-induced interferon-γ secretion, an event that was drastically limited in
Jo

the presence of APAP (Supplementary Figure 4). We then immunophenotyped PBMCs from

four healthy donors who received 1,000 mg of APAP every 6 h for a period of 24 h. As depicted

in Supplementary Figure 6A and B, APAP induced the expansion of Tregs in all donors, as

well as the expression of the coinhibitory receptors LAG3 and TIM3, which are associated with

a strong immunosuppressive phenotype16. Interestingly, APAP induced a concomitant

expansion of myeloid and plasmacytoid dendritic cells (mDCs and pDCs, respectively), the

latter being characterized by an overexpression of both indoleamine 2,3-dioxygenase 1 (IDO1)

and arginase immunosuppressive enzymes (Supplementary Figure 6C, D). Finally, to further

assess the immunomodulation effect of APAP in cancer patients treated with ICB, the major

surrogate cytokines were profiled in the plasma of patients included in the PREMIS study. In

10
total, 92 cytokines were measured using the Olink Target 96 inflammation panel. We found

that interleukin (IL)-10, a crucial mediator of immune suppression induced by Tregs17, and Flt3-

ligand, an essential growth factor for dendritic cells18–20, were significantly upregulated upon

treatment with ICB, exclusively in patients taking APAP (Supplementary Figure 7).

f
r oo
-p
re
lP
na
ur
Jo

11
DISCUSSION

In this study, we found that patients with advanced cancer taking APAP during immunotherapy

experience worse clinical outcomes, which suggests that APAP decreases T cell-mediated

antitumor immunity. It is unlikely that our data are the result of bias or unmeasured

confounding. First, exposure to acetaminophen was not inferred from medical records which

could have introduced the possibility of recall bias. Second, association with adverse outcome

was observed in three independent cohorts including two academic studies and one-industry

sponsored clinical trial which enrolled patients who were hyperselected (45 inclusion/exclusion

f
oo
criteria). Third, the multivariate analysis considered all the features that may be correlated with

both prognosis and need for taking acetaminophen including antibiotic or steroid use and

r
-p
presence of bone metastases. Fourth, in agreement with our clinical findings, we showed that

APAP reduces the efficacy of ICB in a well-characterized preclinical model of colorectal


re
cancer13. Finally, we were able to demonstrate in vitro, using human PBMCs-based functional
lP

assays, that APAP significantly limits the anti-PD-1 therapy-associated effect on interferon-γ
na

production. This observation is consistent with previous data showing that APAP decreases

the interferon-induced antiviral responses of cultured mammalian cells to influenza virus21.


ur
Jo

The first reports on APAP as an immune response modulator were published in the early 90’s4.

Nonetheless, Prymula et al. were the first to investigate this issue in detail in a randomized

study that comprised 9–16-week-old healthy infants5. They reported reduced immunogenicity

of common pediatric vaccines upon APAP use. The impact of APAP on immunogenicity was

further supported by post-hoc analyses from 10 previous trials with a reduction in antibody

concentrations that was consistent across almost all the antigens studied5. Similarly, a recent

systematic review of clinical studies comprising 2,775 patients reported that prophylactic APAP

administration negatively affected the immune response to pneumococcal conjugate vaccines

in children22.

12
Data related to the potential impact of APAP on antitumor immunity remain almost non-

existent. In a retrospective study, Kostner et al. analyzed the impact of body temperature

achieved during IL-2 infusion in patients with advanced melanoma23. Interestingly, they found

that IL-2-induced fever (peak temperature ≥ 39.5 °C) was associated with an improved survival.

However, when stratifying according to APAP use, the association between fever and

improved survival was not present among patients routinely receiving APAP, thereby

suggesting that APAP intake impairs antitumor immunity.

To date, the mechanisms involved in the immunomodulatory effect of APAP were unknown.

f
As suggested by Kostner et al.23, data from vaccination studies indicated that prevention of

oo
inflammation and fever is unlikely. Indeed, these studies showed that immune responses (and

r
-p
effect of APAP) in children with and without fever were similar5,22. Our preclinical in vivo

experiments indicated that the lower anticancer effects observed when APAP was combined
re
with ICB were associated with a significant increase in tumor infiltrating Tregs. These data are
lP

in line with a recent mice study reporting that APAP has an impact on the maternal immune
na

adaptation to pregnancy, by strongly increasing the proportion FoxP3+ Tregs in uterus-draining

lymph nodes24. FoxP3+ Tregs, which are essential for promoting maternal immune tolerance
ur

toward the fetus, are also involved in antitumor immune response suppression. Similarly, in a
Jo

mouse model of drug-induced liver injury, administration of APAP was shown to promote Treg

induction and IL-10 production via a protective feedback loop that physiologically aims to

alleviate APAP-induced liver injury, thus contributing to an immunosuppressive milieu25.

Strikingly, high-throughput proteomic analysis of the plasma allowed us to identify IL-10, a

crucial mediator of immune suppression induced by Tregs17, as being significantly upregulated

upon treatment with ICB exclusively in patients taking APAP. Moreover, immunophenotyping

of PBMCs from healthy donors before and after APAP consumption showed a significant

upregulation of Tregs, particularly of the subset expressing LAG3 and TIM3 immune

checkpoints, which are critical for Treg-mediated suppression26. In addition, APAP was also

found to promote the expression of Flt3-ligand, an essential growth factor for DCs18–20, and a

13
significant expansion of mDCs and pDCs expressing IDO1 and arginase, two subsets of

immune cell populations known to be associated with induction of immunosuppressive Tregs27.

Altogether, our data suggest that Tregs are key players that may underlie the

immunomodulatory effect of APAP, thus compromising ICB efficacy.

While some controversies still exist concerning the potential predisposing effect of

acetaminophen on cancer development, it is commonly considered as an innocuous drug28.

By combining preclinical experiments and high-throughput profiling of human clinical samples,

our study reports the most comprehensive picture of the immunomodulatory effects of APAP.

f
oo
A recent study raised concerns about the potential deleterious impact of APAP in patients with

coronavirus disease 201929. Our results confirm that more research should be performed to

r
-p
understand the impact of APAP on immunity and present a compelling case for caution in using

this drug in cancer patients treated with ICB. Whether this rule applies at immunotherapy onset
re
or all over the treatment duration, to all antipyretics, all regimens (ICB combined with cytotoxic
lP

chemotherapy or with tyrosine kinase inhibitors), and to other immuno-oncology agents


na

(antitumor vaccines, chimeric antigen receptor T cells) requires further investigations.


ur
Jo

14
ACKNOWLEDGEMENTS
The authors thank the patients who participated in this study and the investigators and
teams who conducted the study. The authors also acknowledge the contributions of
Céline Auzanneau, Maud NGO Camus and Claudio Nicotra for sample logistics.

FUNDING
This work was supported by the Conseil Regional Nouvelle Aquitaine, Fondation
Bergonié, Explicyte and Gustave Roussy. No grant number applicable.

DISCLOSURE
AI reports a consulting or advisory role with AstraZeneca, Bayer, Chugai, Deciphera,
Merck, Parthenon, Roche, Springworks; grants from AstraZeneca, Bayer, BMS, Merck,
MSD, Novartis, Pharmamar, Roche AB, JPG are employees of Explicyte JCS is

f
currently employee of Amgen. The other co-authors declare no conflict of interest.

r oo
REFERENCES
-p
1. Prescott LF. Paracetamol: past, present, and future. Am J Ther. March 2000;7(2):143–
re
147.

2. Ueno K, Yamaura K, Nakamura T, Satoh T, Yano S. Acetaminophen-induced


lP

immunosuppression associated with hepatotoxicity in mice. Res Commun Mol Pathol


Pharmacol. 2000;108(3–4):237–251.
na

3. Yamaura K, Ogawa K, Yonekawa T, Nakamura T, Yano S, Ueno K. Inhibition of the


antibody production by acetaminophen independent of liver injury in mice. Biol Pharm Bull.
February 2002;25(2):201–205.
ur

4. Graham NM, Burrell CJ, Douglas RM, Debelle P, Davies L. Adverse effects of aspirin,
Jo

acetaminophen, and ibuprofen on immune function, viral shedding, and clinical status in
rhinovirus-infected volunteers. J Infect Dis. December 1990;162(6):1277–1282.

5. Prymula R, Siegrist C-A, Chlibek R, Zemlickova H, Vackova M, Smetana J, et al. Effect of


prophylactic paracetamol administration at time of vaccination on febrile reactions and
antibody responses in children: two open-label, randomised controlled trials. Lancet.
October 17, 2009;374(9698):1339–1350.

6. Falup-Pecurariu O, Man SC, Neamtu ML, Chicin G, Baciu G, Pitic C, et al. Effects of
prophylactic ibuprofen and paracetamol administration on the immunogenicity and
reactogenicity of the 10-valent pneumococcal non-typeable Haemophilus influenzae
protein D conjugated vaccine (PHiD-CV) co-administered with DTPa-combined vaccines
in children: An open-label, randomized, controlled, non-inferiority trial. Hum Vaccin
Immunother. March 4, 2017;13(3):649–660.

7. ACIP Vaccine Administration Guidelines for Immunization | CDC [Internet]. 2022 [cited
May 5, 2022]. Available at: https://www.cdc.gov/vaccines/hcp/acip-recs/general-
recs/administration.html

15
8. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved
survival with ipilimumab in patients with metastatic melanoma. N Engl J Med.
2010;363(8):711–23.

9. Sun L, Zhang L, Yu J, Zhang Y, Pang X, Ma C, et al. Clinical efficacy and safety of anti-
PD-1/PD-L1 inhibitors for the treatment of advanced or metastatic cancer: a systematic
review and meta-analysis. Sci Rep. February 7, 2020;10(1):2083.

10. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al.
Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. New England Journal
of Medicine. November 5, 2015;373(19):1803–1813.

11. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New
response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1).
European Journal of Cancer. January 2009;45(2):228–247.

12. Van Gassen S, Gaudilliere B, Angst MS, Saeys Y, Aghaeepour N. CytoNorm: A

f
oo
Normalization Algorithm for Cytometry Data. Cytometry Part A. 2020;97(3):268–278.

13. Lau J, Cheung J, Navarro A, Lianoglou S, Haley B, Totpal K, et al. Tumour and host cell

r
PD-L1 is required to mediate suppression of anti-tumour immunity in mice. Nat Commun.
February 21, 2017;8:14572. -p
re
14. Peng W, Liu C, Xu C, Lou Y, Chen J, Yang Y, et al. PD-1 blockade enhances T-cell
migration to tumors by elevating IFN-γ inducible chemokines. Cancer Res. October 15,
2012;72(20):5209–5218.
lP

15. Dulos J, Carven GJ, van Boxtel SJ, Evers S, Driessen-Engels LJA, Hobo W, et al. PD-1
blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from
na

patients with prostate and advanced melanoma cancer. J Immunother. March


2012;35(2):169–178.
ur

16. Ma Q, Liu J, Wu G, Teng M, Wang S, Cui M, et al. Co-expression of LAG3 and TIM3
identifies a potent Treg population that suppresses macrophage functions in colorectal
Jo

cancer patients. Clin Exp Pharmacol Physiol. October 2018;45(10):1002–1009.

17. Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich J-M, et al. Interleukin-
10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated
inflammation. Immunity. April 22, 2011;34(4):566–578.

18. McKenna HJ, Stocking KL, Miller RE, Brasel K, De Smedt T, Maraskovsky E, et al. Mice
lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells,
dendritic cells, and natural killer cells. Blood. June 1, 2000;95(11):3489–3497.

19. Waskow C, Liu K, Darrasse-Jèze G, Guermonprez P, Ginhoux F, Merad M, et al. The


receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral
lymphoid tissues. Nat Immunol. June 2008;9(6):676–683.

20. Ginhoux F, Liu K, Helft J, Bogunovic M, Greter M, Hashimoto D, et al. The origin and
development of nonlymphoid tissue CD103+ DCs. J Exp Med. December 21,
2009;206(13):3115–3130.

21. Crocker JF, Digout SC, Lee SH, Rozee KR, Renton K, Field CA, et al. Effects of
antipyretics on mortality due to influenza B virus in a mouse model of Reye’s syndrome.
Clin Invest Med. October 1998;21(4–5):192–202.

16
22. Koufoglou E, Kourlaba G, Michos A. Effect of prophylactic administration of antipyretics on
the immune response to pneumococcal conjugate vaccines in children: a systematic
review. Pneumonia (Nathan). April 25, 2021;13(1):7.

23. Køstner AH, Ellegaard M-BB, Christensen IJ, Bastholt L, Schmidt H. Fever and the use of
paracetamol during IL-2-based immunotherapy in metastatic melanoma. Cancer Immunol
Immunother. March 2015;64(3):349–355.

24. Thiele K, Solano ME, Huber S, Flavell RA, Kessler T, Barikbin R, et al. Prenatal
acetaminophen affects maternal immune and endocrine adaptation to pregnancy, induces
placental damage, and impairs fetal development in mice. Am J Pathol. October
2015;185(10):2805–2818.

25. Wang X, Sun R, Chen Y, Lian Z-X, Wei H, Tian Z. Regulatory T cells ameliorate
acetaminophen-induced immune-mediated liver injury. Int Immunopharmacol. April
2015;25(2):293–301.

f
oo
26. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with
Specialized Functions in Immune Regulation. Immunity. May 17, 2016;44(5):989–1004.

r
27. Kushwah R, Hu J. Role of dendritic cells in the induction of regulatory T cells. Cell Biosci.
May 24, 2011;1(1):20. -p
re
28. Weiss NS. Use of acetaminophen in relation to the occurrence of cancer: a review of
epidemiologic studies. Cancer Causes Control. December 2016;27(12):1411–1418.
lP

29. MANJANI L, DESAI N, KOHLI A, ARYA R, WOODS C, DESALE S. EFFECTS OF


ACETAMINOPHEN ON OUTCOMES IN PATIENTS HOSPITALIZED WITH COVID-19.
Chest. October 2021;160(4):A1072.
na
ur
Jo

17
FIGURES LEGENDS

Figure 1. Acetaminophen (APAP) exposure impairs the efficacy of immune checkpoint

blockers in patients with cancer. (A) Volcano plot representation of the Log Rank p-values of

overall survival (OS) (y axis) and delta median OS (x axis) associated with each plasmatic metabolite in

the CheckMate-025 cohort. Optimal cutoff value for each metabolite marker was used to categorize the

patients as ‘High’ or ‘Low’ status. (B) Kaplan Meier curve of OS according to baseline plasmatic

APAP/APAP glucuronide levels in the CheckMate-025 cohort. (C,D,F,G) Kaplan-Meier curves of the

progression-free survival (C,F) and OS (D,G) according to APAP/APAP glucuronide levels in the BIP

f
(C,D) and PREMIS (F,G) cohorts. (E,H) Proportion of responder (R) and non-responder (NR) patients

oo
in the BIP (E) and PREMIS (H) cohorts according to their baseline plasmatic levels of APAP/APAP

glucuronide classified as ‘Absence’ and ‘Presence’. p-value was calculated by Chi-squared test.

r
-p
re
lP
na
ur
Jo

18
Table 1. Baseline clinical characteristics of patients from the PREMIS Cohort.

Characteristics Patients (N = 297)


Age - yr
Median (range) 63 (26 – 101)
Sex – no. (%)
Female 121 (40.7)
Male 176 (59.3)
ECOG performance-status score* –(%)
0 106 (35.7)
1 142 (47.8)
≥2 48 (16.2)
NA 1 (0.3)
Antibiotic use no. (%)
Yes 6 (2)
No 291 (98)

f
Steroids use no. (%)

oo
Yes 24 (8.1)
No 273 (91.9)
Number of previous treatment regimens –

r
no. (%)
0-1
≥2
-p 94 (31.6)
176 (59.3)
re
NA 27 (9.1)
Number of metastatic sites – no. (%)
0-1 69 (23.2)
lP

≥2 228 (76.8)
Liver metastases no. (%)
Yes 75 (25.2)
na

No 222 (74.8)
Bone metastases no. (%)
Yes 91 (30.6)
ur

No 206 (69.4)
Lactate dehydrogenase levels no(%)
≤ upper limit of normal (ULN) 241 (81.1)
Jo

> upper limit of normal (ULN) 56 (18.9)


Type of immunotherapy – no. (%)
Anti-PD1 164 (55.2)
Anti-PD-L1 102 (34.3)
Combination of immunotherapies 31 (10.4)
Type of cancer
Non-small cell lung cancer 111 (37.4)
Melanoma 24 (8.1)
Soft-tissue sarcoma 22 (7.4)
Renal cell carcinoma 18 (6.1)
Urothelial carcinoma 15 (5)
Others1 107 (36)
RECIST**
PD 186 (62.6)
SD 34 (11.4)
PR 63 (21.2)
CR 12 (4)
NA 2 (0.7)

* Performance-status scores on the Eastern Cooperative Oncology Group (ECOG) scale range from 0
(no disability) to 5 (death).
** Response Evaluation Criteria In Solid Tumors
1Cervix carcinoma, colorectal cancer, gastric cancer, head and neck cancer, renal cancer, soft-tissue
sarcoma, triple negative breast carcinoma

f
r oo
-p
re
lP
na
ur
Jo
Jo
ur
na
lP
re
-p
r oo
f
Table 2. Multivariate analysis of progression-free survival and overall survival in

the PREMIS cohort.

Progression-Free Survival

Independent variables Hazard ratio 95% confidence interval P-value


Age (≥ 63 years, median value) 0.71 0.54–0.94 0.018
APAP exposure (Yes) 1.43 1.07–1.91 0.015
LDH levels (> ULN*) 1.55 1.17–2.06 0.002
ECOG status (≥ 2) 2.05 1.41–2.96 < 0.001
Liver metastasis (Yes) 2.10 1.54–2.87 < 0.001

f
Overall Survival

oo
Independent variables Hazard ratio 95% confidence interval P-value
Sex (Male) 1.45 0.99–2.11 0.056

r
APAP exposure (Yes)
LDH levels (> ULN*)
-p
1.78
1.91
1.18–2.68
1.30–2.81
0.006
0.001
re
Liver metastasis (Yes) 2.60 1.76–3.85 < 0.001
lP

ECOG status (≥ 2) 3.57 2.27–5.60 < 0.001


na

* ULN= upper limit of normal


ur
Jo
Jo
ur
na
lP
re
-p
r oo
f
Jo
ur
na
lP
re
-p
ro
of
Annals of Oncology 29: 1437–1444, 2018
doi:10.1093/annonc/mdy103
Published online 30 March 2018

ORIGINAL ARTICLE

Negative association of antibiotics on clinical activity


of immune checkpoint inhibitors in patients with
advanced renal cell and non-small-cell lung cancer

L. Derosa1,2,3†, M. D. Hellmann4,5,6†, M. Spaziano7, D. Halpenny8, M. Fidelle1,2,3, H. Rizvi9, N. Long8,


A. J. Plodkowski8, K. C. Arbour4, J. E. Chaft4,5, J. A. Rouche10, L. Zitvogel1,2,3,11, G. Zalcman12,
L. Albiges1,3,13,14, B. Escudier1,13,14 & B. Routy1,2,3,15,16*
1
Gustave Roussy Cancer Campus (GRCC), Villejuif; 2Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015; Equipe Labellisée—Ligue Nationale
Contre le Cancer, Villejuif; 3Université Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France; 4Department of Medicine, Thoracic Oncology Service,
Memorial Sloan Kettering Cancer Center, New York; 5Department of Medicine, Weill Cornell Medical College, New York; 6Parker Institute for Cancer
Immunotherapy, New York, USA; 7Cardiology Division, Department of Medicine, McGill University, Montreal, Canada; 8Department of Radiology; 9Druckenmiller
Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, USA; 10Department of Imaging, Gustave Roussy, Villejuif; 11Center of Clinical
Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif; 12Thoracic Oncology Department-CIC1425/CLIP2 Paris-Nord, Hospital Bichat-Claude Bernard, AP-HP,
University Paris-Diderot, Paris; 13Department of Medical Oncology, Gustave Roussy, Villejuif; 14Immunologie Intégrative des Tumeurs et Génétique Oncologique,
GRCC, Villejuif, France; 15Hematology-Oncology Division, Department of Medicine, Centre Hospitalier de l’Université de Montréal (CHUM), Montréal; 16Centre de
Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada

*Correspondence to: Dr Bertrand Routy, CRCHUM, 900 St-Denis Street R10.474, Montreal, QC, Canada H2X 0A9. Tel: þ1-514-890-8000 (Ext. 31335); E-mail: bertrand.routy@umontreal.ca

Both authors contributed equally as senior authors.

Background: The composition of gut microbiota affects antitumor immune responses, preclinical and clinical outcome
following immune checkpoint inhibitors (ICI) in cancer. Antibiotics (ATB) alter gut microbiota diversity and composition leading
to dysbiosis, which may affect effectiveness of ICI.
Patients and methods: We examined patients with advanced renal cell carcinoma (RCC) and non-small-cell lung cancer
(NSCLC) treated with anti-programmed cell death ligand-1 mAb monotherapy or combination at two academic institutions.
Those receiving ATB within 30 days of beginning ICI were compared with those who did not. Objective response, progression-
free survival (PFS) determined by RECIST1.1 and overall survival (OS) were assessed.
Results: Sixteen of 121 (13%) RCC patients and 48 of 239 (20%) NSCLC patients received ATB. The most common ATB were
b-lactam or quinolones for pneumonia or urinary tract infections. In RCC patients, ATB compared with no ATB was associated
with increased risk of primary progressive disease (PD) (75% versus 22%, P < 0.01), shorter PFS [median 1.9 versus 7.4 months,
hazard ratio (HR) 3.1, 95% confidence interval (CI) 1.4–6.9, P < 0.01], and shorter OS (median 17.3 versus 30.6 months, HR 3.5, 95%
CI 1.1–10.8, P ¼ 0.03). In NSCLC patients, ATB was associated with similar rates of primary PD (52% versus 43%, P ¼ 0.26) but
decreased PFS (median 1.9 versus 3.8 months, HR 1.5, 95% CI 1.0–2.2, P ¼ 0.03) and OS (median 7.9 versus 24.6 months, HR 4.4,
95% CI 2.6–7.7, P < 0.01). In multivariate analyses, the impact of ATB remained significant for PFS in RCC and for OS in NSCLC.
Conclusion: ATB were associated with reduced clinical benefit from ICI in RCC and NSCLC. Modulatation of ATB-related
dysbiosis and gut microbiota composition may be a strategy to improve clinical outcomes with ICI.
Key words: antibiotics, immune checkpoint inhibitors, non-small-cell lung cancer, renal cell carcinoma, microbiota

Introduction axis have changed the therapeutic landscape in both renal cell car-
cinoma (RCC) and non-small-cell lung cancer (NSCLC). In RCC,
Immune checkpoint inhibitors (ICIs) targeting the programmed higher objective response rate and longer overall survival (OS)
cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1) were seen with nivolumab (anti-PD-1 mAb) compared with

C The Author(s) 2018. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
V
All rights reserved. For permissions, please email: journals.permissions@oup.com.
Original article Annals of Oncology
everolimus [1] and represents a therapeutic option in relapsed categorical data and t-test for continuous data. Two ATB therapeutic win-
RCC. The robust clinical effect of anti-PD-1 mAb was observed dows were analyzed, either ATB prescribed 30 days or up to 60 days before
regarless of PD-L1 expression and standard prognostic factors [2]. the first injection of ICI, termed ATB-30 and ATB-60, respectively.
In NSCLC, anti-PD-(L)1 mAb improved response rates and sur- Survival curves were estimated by the Kaplan–Meier method and com-
pared with the Log-rank test (univariate analysis). Univariate hazard
vival in a subset of NSCLC patients [3–5] in both first line or se- ratios (HRs) were calculated using log-rank method. Multivariable Cox
cond line but few clinical features reliably predict benefit. regression model was used to determine HRs and 95% confidence inter-
Primary resistance to ICI is common in patients with both vals (CIs) for progression-free survival (PFS) and OS between ATB and
RCC and NSCLC, ranging from 35% to 44% and remains unpre- no ATB, adjusting for other clinicopathologic features. Statistical tests
dictable [1, 3]. Much recent effort has focused on biomarkers of were two-sided, and a P-value <0.05 was considered statistically signifi-
response to immunotherapy (e.g. PD-L1 [6], mutation burden cant. Statistical analyses were carried out using SPSS.
[5], gene expression signatures of inflammation [7, 8]) but the
identification of more reliable predictors associated with resist-
ance (primary or acquired) is also critical to instruct new strat- Results
egies to improve precision and broaden responder groups. One hundred and twenty-one patients with RCC and 239 patients
Building on recent data [9, 10], we hypothesized that the modu- with NSCLC were examined in independent cohorts from two
lation of gut microbiota by antibiotics (ATB) may be associated academic cancer centers. The frequency of ATB use within
with resistance to anti-PD-1 mAb. The intestinal microbiota repre- 30 days of starting PD-(L)1 therapy was relatively similar in
sents a complex ecosystem essential for maintaining gut homeosta- NSCLC (20%) and RCC (13%) patients. b-Lactams 6 inhibitors
sis and prevent systemic inflammation [11, 12]. The interactions were the most commonly administered ATB in both groups
between the host and micro-organisms have been identified as a (82% of RCC and 32% of NSCLC) (supplementary Table S1,
complex, inter-connected network where certain microbes tailor available at Annals of Oncology online). Quinolones and sulfona-
local and systemic immune system [13]. ATB are known to affect mides were also frequently used in NSCLC cohorts.
gut microbiota, including loss of distinct species (poor diversity), Among RCC patients, 106 of 121 (88%) received PD-(L)1 mono-
favoring expansion of others, consequently shifting the metabolic therapy, 10 (8%) PD-(L)1 plus CTLA-4 mAbs and 5 (4%) PD-(L)1
capacity [14]. ATB-induced dysbiosis has been associated with a mAbs plus bevacizumab. All RCC patients were treated in clinical
variety of chronic inflammatory disorders [15–17]. In oncology, trials. Clinicopathologic characteristics were typical of patients with
converging findings demonstrate the negative impact of ATB- advanced RCC and were generally well balanced between those who
induced dysbiosis in mice. We built upon this experience to ex- received ATB or not, with the exception of tumor burden (larger
pand the series of patients examined using patients with advanced tumors in the ATB group) and lines of prior therapy (2þ prior lines
RCC and NSCLC treated with anti-PD-(L)1 therapy at two differ- more common in the ATB group) (Table 1A). No patients with
ent centers to evaluate the impact of ATB on resistance. RCC were hospitalized within 30 days of starting PD-(L)1 therapy.
Among NSCLC patients, 48 (20%) patients received ATB with-
in 30 days of PD-(L)1 therapy. Two hundred and five (86%)
Methods received PD-(L)1 monotherapy and 34 (14%) received PD-(L)1
plus CTLA-4 mAbs. Fifty-four (23%) were enrolled in clinical tri-
Patients als. Clinicopathologic features were also typical of patients with
advanced NSCLC, well balanced between the ATB versus no ATB
Patients with advanced RCC (n ¼ 121) at Gustave Roussy and NSCLC
(n ¼ 239) at Memorial Sloan Kettering Cancer Center treated with ICI groups with the exception of recent hospitalization (more com-
were identified. NSCLC patients and half of RCC patients have been pre- mon in the ATB group) and more patients included in the clinical
viously reported [10] but have been fully reanalyzed for multivariate ana- trials in the no ATB group (Table 1B).
lysis. All patients with RCC received anti-PD-1 or anti-PD-L1 (PD-(L)1) In patients with RCC, recent ATB was associated with
mAb alone or in combination with anti-CTLA-4 mAb or bevacizumab increased rate of primary progressive disease (PD) (75% versus
on clinical trials. Patients with NSCLC received anti-PD-(L)1 mAb alone 22%, P < 0.01) (Figure 1A). PFS and OS were also shorter in
or in combination with anti-CTLA-4 mAb.
those patients with ATB than in those with no ATB (median PFS,
Patient records were reviewed to determine any oral or intravenous
ATB use within the 30 or 60 days before the start of anti-PD-(L)1 therapy. 1.9 months versus 7.4 months, HR 3.1, 95% CI 1.4–6.9, P < 0.01;
The class of antibiotic, indication, route of administration and duration median OS, 17.3 months versus 30.6 months, HR 3.5, 95% CI
were collected. Clinicopathologic characteristics were collected for all 1.1–10.8, P ¼ 0.03) (Figure 1B and C).
patients. In RCC patients, additional features included cumulative size of In patients with NSCLC, recent ATB was not associated with
tumor burden (<10 cm versus  10 cm) [18] and site of metastases. In an increased rate of primary PD (52% versus 43%, P ¼ 0.26)
NSCLC patients, smoking status and PD-L1 expression (high defined as (Figure 1D). Similar to RCC, however, PFS and OS were signifi-
50% expression or low/no expression) were also collected if available.
cantly shorter in NSCLC patients with ATB than in those with no
For the evaluation of tumor response, CT scans were reviewed by local
ATB (median PFS, 1.9 months versus 3.8 months, HR 1.5, 95%
specialized radiologists (JAR at GR; DH, NL, AJP at MSKCC) and re-
sponse was determined by Response Evaluation Criteria in Solid Tumors CI 1.0–2.2, P ¼ 0.03; median OS, 7.9 months versus 24.6 months,
(RECIST) version 1.1 [19]. All patients were followed-up until death or HR 4.4, 95% CI 2.6–7.7, P < 0.01) (Figure 1E and F).
data lock (September 2017 for RCC and March 2017 for NSCLC). To assess the robustness of the observation that recent ATB was
associated with decreased benefit with ICI, we also examined the
Statistical analysis effect of ATB within 60 days of starting therapy [ATB-60 days,
Patient characteristics were described according to the status of ATB (ATB n ¼ 22 for RCC (18%); n ¼ 68 for NSCLC (28%)] (supplementary
versus no ATB) and compared using Fisher or chi-squared test for Tables S2 and S3, available at Annals of Oncology online). Taking

1438 | Derosa et al. Volume 29 | Issue 6 | 2018


Annals of Oncology Original article
Table 1A. Baseline characteristics of renal cell carcinoma (RCC) cohort

Characteristics Total ATB 30-0 No ATB P-value


(n 5 121) (n 5 16) (n 5 105)

Age, years Median 61 61 61


Range 28–83 29–83 30–82
Age, years, n (%) <65 79 (65) 8 (50) 69 (66) 0.22
65 42 (35) 8 (50) 36 (34)
Gender, n (%) Male 80 (66) 9 (56) 71 (68) 0.37
Female 41 (34) 7 (44) 34 (32)
Nephrectomy, n (%) Yes 103 (85) 13 (82) 90 (86) 0.64
No 18 (15) 3 (18) 15 (14)
Histology, n (%) Clear cell 115 (95) 14 (88) 101 (96) 0.13
Nonclear cell 6 (5) 2 (12) 4 (4)
IMDC risk group, n (%) Good 25 (21) 3 (19) 22 (21) 0.96
Intermediate 72 (59) 10 (62) 62 (59)
Poor 24 (20) 3 (19) 21 (20)
Tumor burden (mm), n (%) <100 94 (78) 9 (56) 85 (81) 0.03
100 27 (22) 7 (44) 20 (19)
Site of metastasis, n (%) Lung 84 (69) 8 (50) 76 (72) 0.09
Liver 34 (28) 8 (50) 26 (23)
Bone 33 (27) 5 (31) 28 (27)
Brain 14 (12) 0 (0) 14 (13)
Number of prior lines, n (%) 0–1 69 (57) 4 (25) 65 (62) <0.01
2 52 (43) 12 (75) 40 (38)
Treatment, n (%) a-PD-(L)1 mAb 106 (88) 14 (88) 92 (88) 0.86
a-PD-(L)1 mAb þ a-CTLA-4 10 (8) 1 (6) 9 (8)
a-PD-(L)1 mAb þ Bev 5 (4) 1 (6) 4 (4)
ATB, n (%)a Prophylaxis 0 0 0
Therapy 16 (13) 16 (100) 0

a
List of ATB and indication for prophylaxis and therapy are available in supplementary Table S1, available at Annals of Oncology online.
ATB, antibiotics; IMDC, International Metastatic Renal Cell Carcinoma Database Consortium (includes: Karnofsky performance status, time from diagnosis
to treatment, hemoglobin, serum calcium concentration, neutrophil and platelet counts); a, anti; PD-(L)1, PD-1 or PD-L1; mAb, monoclonal antibody; Bev,
bevacizumab.
Bold values reflect statistical significant; *P<0.05, **P<0.01.

into account that gut dysbiosis from ATB can take 1–3 months to in the respective ATB groups. In both RCC and NSCLC cohorts,
normalize [20], we hypothesized that a trend for similar results although not statistically significant due to small numbers, ATB
would be seen with the extended timeline. In RCC, ATB-60 days group was generally associated with worse PFS and OS within
remained associated with increased risk of primary progression nearly every subgroup examined (Figure 2).
(P < 0.01) (supplementary Figure S1A, available at Annals of Finally, we carried out a multivariate analysis of the effect of
Oncology online), shorter PFS (median PFS, 3.1 months versus ATB administration, taking into account classical prognostic
7.4 months, HR 2.3, 95% CI 1.2–4.4, P < 0.01) (supplementary factors relevant to RCC and NSCLC, respectively. In RCC, both
Figure S1B, available at Annals of Oncology online) and trended to- ATB and tumor burden were significantly associated with worse
ward worse OS (median OS, 23.4 months versus 30 months, HR PFS and OS as univariate (Tables 2A and 2B), and were ana-
1.9, 95% CI 0.8–4.7, P ¼ 0.15) (supplementary Figure S1C, avail- lyzed in multivariate modeling. ATB and tumor burden
able at Annals of Oncology online). In NSCLC, ATB-60 days com- remained independently associated with worse PFS in multi-
pared with no ATB group was not different in terms of objective variate analysis (HR for ATB 2.0, P < 0.01), while neither vari-
response or PFS (supplementary Figure S1D and E, available at able remained statistically significant for OS (HR for ATB 2.1,
Annals of Oncology online), but remained significantly associated P ¼ 0.11). In NSCLC, ATB and several other clinicopathologic
with shorter OS (median OS, 9.8 months versus 21.9 months, HR variables were associated with PFS or OS as univariate and fur-
2.0, 95% CI 1.3–3.2, P < 0.01) (supplementary Figure S1F, avail- ther evaluated in multivariate model (Tables 3A and 3B). In the
able at Annals of Oncology online). Cox regression analysis, ATB were not significantly associated
We next examined the impact of ATB on PFS and OS within with worse PFS (HR 1.3, 95% CI 0.9–1.8, P ¼ 0.17) but
individual subgroups of patients. As noted, there were a few vari- remained significantly associated with OS (HR 2.5, 95% CI
ables in both RCC and NSCLC cohorts that were more common 1.6–3.7, P < 0.01).

Volume 29 | Issue 6 | 2018 doi:10.1093/annonc/mdy103 | 1439


Original article Annals of Oncology
Table 1B. Baseline characteristics of non-small-cell lung cancer (NSCLC) cohort

Characteristics Total ATB 30-0 No ATB P-value


(n 5 239) (n 5 48) (n 5 191)

Age, years Median 66 63 66


Range (22–92) (31–92) (22–88)
Age, years, n (%) <65 110 (46) 27 (56) 83 (43) 0.12
65 129 (54) 21 (44) 108 (57)
Gender, n (%) Male 118 (49) 24 (50) 94 (49) 0.92
Female 121 (51) 24 (50) 97 (51)
ECOG performance status, n (%) 0 53 (22) 5 (11) 48 (25) 0.05
1 183 (77) 43 (89) 140 (74)
2 3 (1) 0 3 (1)
Histology, n (%) Squamous 34 (14) 6 (13) 28 (15) 0.71
Nonsquamous 205 (86) 42 (88) 163 (85)
Smoking status, n (%) Smoker 193 (81) 37 (77) 156 (82) 0.47
Nonsmoker 46 (19) 11 (23) 35 (18)
Number of prior lines, n (%) <3 178 (74) 33 (69) 145 (76) 0.31
3 61 (26) 15 (31) 46 (24)
Hospitalization <30 days, n (%) Yes 20 (8) 16 (33) 4 (2) <0.01
No 219 (93) 32 (67) 187 (98)
Treatment, n (%) a-PD-(L)1 mAb 205 (86) 45 (94) 160 (84) 0.08
a-PD-(L)1 mAb þ a-CTLA-4 34 (14) 3 (6) 31 (16)
Clinical trial, n (%) Yes 54 (23) 5 (11) 49 (25) 0.02
No 185 (77) 43 (89) 142 (74)
PD-L1 expression, n (%) High (50%) 21 (9) 6 (12) 15 (8) 0.12
Low (<50%) 64 (27) 9 (19) 55 (29)
Unknown 154 (64) 33 (69) 121 (63)
ATB, n (%)a Prophylaxis 15 (6) 15 (31) 0
Therapy 33 (14) 33 (69) 0

a
List of ATB and indication for prophylaxis and therapy are available in supplemental Table S1, available at Annals of Oncology online.
ECOG, Eastern Cooperative Oncology Group.
Bold values reflect statistical significant; *P<0.05, **P<0.01.

Discussion taxonomic richness [21]. Another level of complexity exists in the


perturbation of microbiome according to the class, the duration,
This study reports the substantial rate of ATB use (13%–20%)
the route of ATB administration and the presence of resistant
proximal to initiating ICI and the potential clinical impact on commensals which play a key role in microbiota recolonization
benefit from ICI. In independent cohorts of RCC and NSCLC fol- and recomposition. 16S ribosomal RNA sequencing of gene
lowed-up in two cancer centers, prior ATB was associated with amplicons of patients’ feces revealed that microbiota returns to
worse outcomes with ICI. its baseline within 1–3 months after ATB discontinuation.
More precisely, patients in ATB group treated with ICI had a However, some bacteria may take years to fully recover [14, 20].
higher rate of PD in RCC and a significant reduction in PFS and To ascertain the difference in ATB timing, we carried out a
OS compared with those with no ATB in both cancer types. ATB sub-group analysis for patients receiving ATB 60 days before
appears to be a determinant of poor prognosis in the context of starting ICI. Interestingly, the impact of ATB 60 days before was
ICI independently of classical prognostic markers and within in- not as potent as within the first 30 days before ICI. Partial micro-
dividual patient subgroups. This paper builds upon our prior biota recovery following ATB might explain the difference in clin-
publication [10] on the negative impact of ATB, which included ical outcomes observed.
249 patients diagnosed with advanced NSCLC (n ¼ 140), RCC Very recent reports highlighted that the composition of gut
(n ¼ 67) and urothelial cancer (n ¼ 42) treated with anti-PD-1/ microbiota may dictate, at least in part, the anticancer activity of
PD-L1 mAb. In that report ATB use was defined as 60 days before ICI. In three different cancer types, patients with higher diversity
and 30 days after the first injection of ICI, and ATB used was asso- or richness of the stool sample at baseline correlated with a better
ciated with decreased PFS from 3.5 to 4.1 months and OS from clinical outcome at 6 months. In metastatic melanoma, patients
11.5 to 20.6 months compared with the no ATB group [10]. with ‘favorable’ microbiota composition were enriched in
In this expanding field, ATB are recognized to be able to shift Ruminococcaceae family members and Bifidobacterium spp. This
the microbiota composition temporally. ATB dysbiosis is associ- favorable microbiota translated into best overall response rate at
ated with a decrease in microbiota diversity and impacts on 6 months [22, 23]. The third study in patients with NSCLC and

1440 | Derosa et al. Volume 29 | Issue 6 | 2018


Annals of Oncology Original article
A P<0.01 B C
100 4% 100 Median PFS 100 Median OS
13% No ATB: 7.4 mo No ATB: 30.6 mo

Progression-free survival (%)


22% ATB: 1.9 mo ATB: 17.3 mo
80 13% 80 80

Overall survival (%)


HR (95%,CI), 3.1 (1.4-6.9) HR (95%,CI), 3.5 (1.1-10.8)
Patients (%) P <0.01 P =0.03
60 60 60
RCC

RCC

RCC
52%
40 75% 40 40

20 20 20
22%
0 0 0
ATB No ATB 0 5 10 15 20 25 30 0 10 20 30 40
n=16 n=105 105 69 35 20 5 4 1 105 81 25 12 6
16 5 3 2 0 0 0 16 10 2 0 0
CR SD
Months Months
PR PD

D E F
100 Median PFS 100 Median OS
100 2% No ATB: 3.8 mo No ATB: 24.6 mo

Progression-free survival (%)


13%
21% ATB: 1.9 mo ATB: 7.9 mo
80 HR (95%,CI), 1.5 (1.0-2.2) 80 HR (95%,CI), 4.4 (2.6-7.6)

Overall survival (%)


80
P =0.03 P<0.01
35%
Patients (%)

NSCLC 60

NSCLC
60
60
NSCLC

34%

40 40
40

52% 20 20
20 43%

0 0
0 0 5 10 15 20 25 30 0 10 20 30 40
ATB No ATB
191 72 33 21 10 5 2
n=48 n=191 191 101 39 20 9
48 17 8 4 2 0 0
Months Months

Figure 1. Best overall response (A), progression-free survival (PFS) (B), and overall survival (OS) (C) in patients with RCC treated with ICI, strati-
fied by use of ATB within 30 days of initiating ICI. Best overall response (D), PFS (E), and OS (F) in patients with NSCLC treated with ICI, strati-
fied by use of ATB within 30 days of initiating ICI. P-values calculated with chi-squared and log-rank tests.

A PFS in RCC B OS in RCC

P-value for P-value for


interaction interaction
All patients (n=121) All patients (n=121)

>65 (n=44) >65 (n=44)


Age 0.11 Age 0.26
<65 (n=77) <65 (n=77)

Male (n=41) Male (n=41)


Sex 0.40 Sex 0.45
Female (n=80) Female (n=80)

Poor (n=24) Poor (n=24)

IMDC Intermediate (n=72) 0.67 IMDC Intermediate (n=72) 0.23

Good (n=25) Good (n=25)

<100 cm (n=94) <100 cm (n=94)


Tumor burden 0.07 Tumor burden 0.53
>100 cm (n=27) >100 cm (n=27)

0-1 (n=69) Number of 0-1 (n=69)


Number of 0.58 0.61
prior lines prior lines
≥2 (n=52) ≥2 (n=52)

0.5 1 2 3 4 5 10 15 0.2 0.5 1 2 3 4 5 10 20

Antibiotics better No antibiotics better Antibiotics better No antibiotics better

C PFS in NSCLC D OS in NSCLC


P-value for P-value for
All patients (n=239) interaction All patients (n=239) interaction

>65 (n=124) >65 (n=124)


0.20 Age 0.10
Age <65 (n=115) <65 (n=115)

Male (n=118) Male (n=118)


Sex 0.15 Sex 0.13
Female (n=121) Female (n=121)

Squamous (n=34) Squamous (n=34)


Histology 0.49 Histology 0.12
Non-squamous (n=205) Non-squamous (n=205)

Number of 3 or more (n=61) Number of 3 or more (n=61)


0.23 0.32
prior lines prior lines 2 or less (n=178)
2 or less (n=178)

1 or more (n=186) 1 or more (n=186)


ECOG 0.55 ECOG 0.48
0 (n=53) 0 (n=53)

Yes (n=54) Yes (n=54)


Clinical trial 0.71 Clinical trial 0.85
No (n=185) No (n=185)

Yes (n=34) Yes (n=34)


Hospitalization 0.16 Hospitalization 0.006
No (n=205) No (n=205)

Treatment Treatment
α-PD-(L)1 mAb (n=205) α-PD-(L)1 mAb (n=205)
0.76 0.42
α-PD-(L)1 mAb + α-CTLA-4 (n=34) α-PD-(L)1 mAb + α-CTLA-4 (n=34)

0.5 1 2 3 4 5 0.5 1 2 3 4 5 10

Antibiotics better No antibiotics better Antibiotics better No antibiotics better

Figure 2. Subgroup analyses of independent prognostic factors for PFS (A and B) and OS (C and D) stratification in RCC and NSCLC, respect-
ively. P-value for interaction calculated with Cox proportional hazards model.

Volume 29 | Issue 6 | 2018 doi:10.1093/annonc/mdy103 | 1441


Original article Annals of Oncology
Table 2A. Uni- and multivariate analyses for progression-free survival (PFS) Table 3A. Uni- and multivariate analyses for PFS in non-small-cell lung can-
in renal cell carcinoma cohort cer cohort

Univariate analysis Multivariate analysis Univariate analysis Multivariate analysis

Prognostic factor PFS P-value PFS P-value Prognostic factor PFS P-value PFS P-value
HR (95% CI) HR (95% CI) HR (95% CI) HR (95% CI)

ATB 30-0/No ATB 2.3 (1.3–4.0) <0.01 2.2 (1.3–3.3) <0.01 ATB 30-0/No ATB 1.4 (1.0– 2.0) 0.04 1.3 (0.9–1.8) 0.17
Age 1.2 (0.8–1.8) 0.43 – – Age 1.2 (0.9–1.6) 0.2 – –
65 years/<65 years  65 years/<65 years
IMDC risk group 1.0 (0.6–1.7) 0.92 – – Histology 1.1 (0.8–1.5) 0.92 – –
Intermediate/good Squamous/nonsquamous
Poor/good 1.7 (0.9–3.2) 0.11 Smoking status 0.7 (0.5–1.0) 0.04 0.7 (0.5–1.0) 0.03
Tumor burden 2.1 (1.3–3.3 ) <0.01 2.0 (1.2–3.2) <0.01 Smoker/nonsmoker
Number of prior regimens 1.4 (1.0–1.9) 0.05 1.2 (0.8–1.6) 0.40
HR, hazard ratio; CI, confidence interval. 3/<3
Bold values reflect statistical significant; *P<0.05, **P<0.01. ECOG performance status 1.7 (1.2–2.4) <0.01 1.5 (1.0–2.2) 0.03
0 versus 1
Clinical trial 0.7 (0.5–1.0) 0.02 0.8 (0.6–1.1) 0.19
Yes versus no
Table 2B. Uni- and multivariate analyses for overall survival (OS) in renal Hospitalization 1.2 (0.8–1.7) 0.45 – –
cell carcinoma cohort Yes versus no

Univariate analysis Multivariate analysis ECOG, Eastern Cooperative Oncology Group.


Bold values reflect statistical significant; *P<0.05, **P<0.01.
Prognostic factor OS P-value OS P-value
HR (95% CI) HR (95% CI)

ATB 30-0/No ATB 2.4 (1.1–5.7) 0.04 2.1 (0.9–5.0) 0.11


Age 0.8 (0.4–1.5) 0.43 – – Table 3B. Uni- and multivariate analyses for OS in non-small-cell lung
 65 years/< 65 years cancer cohort
IMDC risk group 0.7 (0.3–1.7) 0.45 2.1 (0.8–5.3) 0.12
Univariate analysis Multivariate analysis
Intermediate/good 2.4 (1.0–6.0) 0.06
Poor/good Prognostic factor OS P-value OS P-value
Tumor burden 2.4 (1.2–4.6) <0.01 1.8 (0.9–3.6) 0.09 HR (95% CI) HR (95% CI)

Bold values reflect statistical significant; *P<0.05, **P<0.01. ATB 30-0/No ATB 2.9 (1.9–4.4) <0.01 2.5 (1.6–3.7) <0.01
Age 1.3 (0.9–1.9) 0.23 – –
65 years/<65 years
RCC amenable to anti-PD-1 mAb showed that Akkermansia Histology 1.4 (0.8–1.6) 0.57 – –
Squamous/nonsquamous
muciniphila (accompanied with Ruminococcacae) was signifi-
Smoking status 1.2 (0.7–1.9) 0.55 – –
cantly associated to more favorable outcomes. In this paper, the
Smoker/nonsmoker
changes in the microbiota composition due to ATB uptake were
Number of prior regimens 1.9 (1.3–2.9) <0.01 1.6 (1.1–2.4) 0.02
not addressed. Authors have also characterized other microbiota
3/<3
sites including saliva in cancer patients; however, in MM [23] ECOG performance status 3.6 (1.9–6.5) <0.01 2.6 (1.4–4.9) <0.01
and head and neck squamous-cell carcinoma [24], there was no 0 versus 1
correlation with ICI activity. Clinical trial 0.4 (0.2–0.7) <0.01 0.6 (0.3–1.0) 0.06
In addition, the microbiota composition could account for Yes versus no
uncoupling efficacy from toxicity in metastatic melanoma Hospitalization 1.1 (0.6–1.9) 0.76 – –
patients treated with anti-CTLA-4 [25, 26]. Baseline fecal samples Yes versus no
with high bacterial diversity enriched with Bacteroidetes phylum
and depleted with Firmicutes correlated with the absence of im- ECOG, Eastern Cooperative Oncology Group.
mune colitis. Of note, in the present study, the frequency of im- Bold values reflect statistical significant; *P<0.05, **P<0.01.
mune-related adverse effects was too small (data not shown) to
evaluate the association between ATB and adverse events.
Our study has several limitations. The first one is the retro- patient characteristics, including a higher rate of recent hospital-
spective component of the data entry from two institutions, al- ization in the NSCLC patients treated with ATB. Recent hospital-
though patients’ clinical responses were standardized and ization was not associated with worse PFS or OS as a univariate.
reviewed objectively using the RECIST 1.1 criteria in both cen- In RCC even in the current uncertainty of PD-1 efficacy in naive
ters, the timing of scan acquisition was not prespecified. versus previously treated patients [1, 27–29], we reported a differ-
Furthermore, we acknowledge some notable differences in ence in line of therapy in both groups. To acknowledge the

1442 | Derosa et al. Volume 29 | Issue 6 | 2018


Annals of Oncology Original article
encountered differences in patient characteristics, we examined nivolumab versus everolimus in advanced renal cell carcinoma. Eur Urol
the impact of ATB within individual subgroups and found overall 2017 Mar 3 [Epub ahead of print], doi: 10.1016/j.eururo.2017.02.010.
3. Borghaei H, Paz-Ares L, Horn L et al. Nivolumab versus docetaxel in
consistency of the effect of ATB. We also conducted multivariable
advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;
analysis to adjust for significant prognostic factors identified in 373(17): 1627–1639.
the univariate analysis, which still showed a negative impact of 4. Reck M, Rodrı́guez-Abreu D, Robinson AG et al. Pembrolizumab versus
ATB. Secondly, the analysis did not take into consideration add- chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J
itional factors with a potential impact on the microbiota compos- Med 2016; 375(19): 1823–1833.
ition such as diet, country of origin, or other medications. 5. Carbone DP, Reck M, Paz-Ares L et al. First-line nivolumab in stage IV or re-
Thirdly, we did not characterize the mechanism by which ATB current non-small-cell lung cancer. N Engl J Med 2017; 376(25): 2415–2426.
6. Patel SP, Kurzrock R. PD-L1 Expression as a predictive biomarker in
exert a detrimental effect on clinical outcomes. We speculate that
cancer immunotherapy. Mol Cancer Ther 2015; 14(4): 847–856.
ATB-related dysbiosis decreases the diversity, shifts the micro- 7. Gao J, Shi LZ, Zhao H et al. Loss of IFN-c pathway genes in tumor cells
biota and probably eradicates the most immunogenic bacteria as a mechanism of resistance to anti-CTLA-4 therapy. Cell 2016; 167(2):
required to engaged the immune system unleashed by PD-(L)1 397–404.e9.
blockade. Whether antibiotherapy reflects a general prognostic 8. Ayers M, Lunceford J, Nebozhyn M et al. IFN-c-related mRNA profile predicts
association or is causalitively linked with resistance to ICI clinical response to PD-1 blockade. J Clin Invest 2017; 127(8): 2930–2940.
remains a matter of debate. It is noteworthy that ATB-based con- 9. Vétizou M, Pitt JM, Daillère R et al. Anticancer immunotherapy by
CTLA-4 blockade relies on the gut microbiota. Science 2015; 350(6264):
ditionning of tumor bearing mice blunts the efficacy of PD-1 or 1079–1084.
PD-1 þ CTLA-4 blockade in otherwise ‘normal’ animals, sug- 10. Routy B, Le Chatelier E, Derosa L et al. Gut microbiome influences efficacy
gesting a causal link between ATB and primary resistance to ICI of PD-1-based immunotherapy against epithelial tumors. Science 2017 Nov
and immunogenic chemotherapy [9, 10, 30, 31]. Finally, the lack 2 [Epub ahead of print], doi: 10.1126/science.aan3706.
of PD-L1 status characterization in most of the patients (outside 11. Lozupone CA, Stombaugh JI, Gordon JI et al. Diversity, stability and re-
of some NSCLC) leaves open the possibility that tumor-intrinsic silience of the human gut microbiota. Nature 2012; 489(7415): 220–230.
12. Zitvogel L, Ayyoub M, Routy B, Kroemer G. Microbiome and anticancer
factors could influence these results.
immunosurveillance. Cell 2016; 165(2): 276–287.
Altogether, these results confirm that ATB-associated dysbiosis 13. Ubeda C, Pamer EG. Antibiotics, microbiota, and immune defense.
might be deleterious in patients treated with ICI, suggesting that Trends Immunol 2012; 33(9): 459–466.
an intact gut microbiota is needed to mobilize the immune sys- 14. Lange K, Buerger M, Stallmach A, Bruns T. Effects of antibiotics on gut
tem regardless of the tumor site. Efforts to improve antibiotic microbiota. Dig Dis 2016; 34(3): 260–268.
stewardship are already ongoing to prevent the emergence of 15. Korpela K, de Vos WM. Antibiotic use in childhood alters the gut micro-
multidrug resistant organisms [32], which can be particularly biota and predisposes to overweight. Microb Cell 2016; 3(7): 296–298.
16. Leclercq S, Mian FM, Stanisz AM et al. Low-dose penicillin in early life
dangerous for cancer patients [33, 34]. The data in this report
induces long-term changes in murine gut microbiota, brain cytokines
may add additional incentive to avoid unnecessary ATB. More and behavior. Nat Commun 2017; 8: 15062.
studies are warranted to confirm the deleterious effect of ATB in 17. Arrieta M-C, Stiemsma LT, Dimitriu PA et al. Early infancy microbial
large prospective trials and to develop novel diagnostic tools and metabolic alterations affect risk of childhood asthma. Sci Transl Med
based on gut microbiota in cancer patients, to predict response/ 2015; 7: 307ra152.
resistance to ICI and identified the key microbiota signatures for 18. Iacovelli R, Lanoy E, Albiges L, Escudier B. Tumour burden is an inde-
each tumor site. The discovery of bacteria capable of shifting an pendent prognostic factor in metastatic renal cell carcinoma. BJU Int
2012; 110(11): 1747–1753.
unfavorable ATB-associated dysbiosis to a favorable microbiota 19. Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation cri-
will help build a future therapeutic concept, whereby modulation teria in solid tumours: revised RECIST guideline (version 1.1). Eur J
of gut microbiota by bacteria could increase ICI clinical activity. Cancer Oxf Engl 1990 2009; 45: 228–247.
20. Jakobsson HE, Jernberg C, Andersson AF et al. Short-term antibiotic
treatment has differing long-term impacts on the human throat and gut
microbiome. PLoS One 2010; 5(3): e9836.
Funding 21. Jernberg C, Löfmark S, Edlund C, Jansson JK. Long-term ecological
impacts of antibiotic administration on the human intestinal microbiota.
Gustave Roussy Fondation Philanthropia (no grant number ISME J 2007; 1: 56–66.
applies) to LD and BR; RK Smiley Canadian Hematology Society 22. Matson V, Fessler J, Bao R et al. The commensal microbiome is associ-
2017 (National Canadian grant) (no grant number applies). ated with anti–PD-1 efficacy in metastatic melanoma patients. Science
2018; 359(6371): 104–108.
23. Gopalakrishnan V, Spencer CN, Nezi L et al. Gut microbiome modulates
response to anti-PD-1 immunotherapy in melanoma patients. Science
Disclosure 2017 Nov 2 [Epub ahead of print], doi: 10.1126/science.aan4236.
24. Ferris RL, Blumenschein G, Harrington K et al. Abstract CT022: evalu-
The authors have declared no conflicts of interest. ation of oral microbiome profiling as a response biomarker in squamous
cell carcinoma of the head and neck: analyses from CheckMate 141.
Cancer Res. 2017; 77(13 Suppl): CT022–CT022.
References 25. Dubin K, Callahan MK, Ren B et al. Intestinal microbiome analyses iden-
1. Motzer RJ, Escudier B, McDermott DF et al. Nivolumab versus everoli- tify melanoma patients at risk for checkpoint-blockade-induced colitis.
mus in advanced renal-cell carcinoma. N Engl J Med 2015; 373: Nat Commun 2016; 7: 10391.
1803–1813. 26. Chaput N, Lepage P, Coutzac C et al. Baseline gut microbiota predicts clinic-
2. Escudier B, Sharma P, McDermott DF et al. CheckMate 025 randomized al response and colitis in metastatic melanoma patients treated with ipilimu-
phase 3 study: outcomes by key baseline factors and prior therapy for mab. Ann Oncol Off J Eur Soc Med Oncol 2017; 28(suppl_5): 1368–1379.

Volume 29 | Issue 6 | 2018 doi:10.1093/annonc/mdy103 | 1443


Original article Annals of Oncology
27. Hammers HJ, Plimack ER, Infante JR et al. Safety and efficacy of 31. Viaud S, Saccheri F, Mignot G et al. The intestinal microbiota modulates
nivolumab in combination with ipilimumab in metastatic renal cell the anticancer immune effects of cyclophosphamide. Science 2013;
carcinoma: the CheckMate 016 Study. J Clin Oncol 2017; 35(34): 342(6161): 971–976.
3851–3858. 32. Rosa RG, Goldani LZ, dos Santos RP. Association between adherence to
28. Motzer RJ, Rini BI, McDermott DF et al. Nivolumab for metastatic renal an antimicrobial stewardship program and mortality among hospitalised
cell carcinoma: results of a randomized phase II trial. J Clin Oncol 2015; cancer patients with febrile neutropaenia: a prospective cohort study.
33(13): 1430–1437. BMC Infect Dis 2014; 14(1): 286.
29. Meeting Library j Safety and efficacy of nivolumab in metastatic renal 33. Cometta A, Calandra T, Bille J, Glauser MP. Escherichia coli resistant to
cell carcinoma (mRCC): Results from the NIVOREN GETUG-AFU 26 fluoroquinolones in patients with cancer and neutropenia. N Engl J Med
study. https://meetinglibrary.asco.org/record/157690/abstract (3 March 1994; 330(17): 1240–1241.
2018, date last accessed). 34. Montassier E, Batard E, Gastinne T et al. Recent changes in bacteremia
30. Iida N, Dzutsev A, Stewart CA et al. Commensal bacteria control cancer in patients with cancer: a systematic review of epidemiology and anti-
response to therapy by modulating the tumor microenvironment. biotic resistance. Eur J Clin Microbiol Infect Dis. 2013; 32(7):
Science 2013; 342(6161): 967–970. 841–850.

1444 | Derosa et al. Volume 29 | Issue 6 | 2018


ORIGINAL ARTICLE

Do We Really Need to Thank the Beatles for the


Financing of the Development of the Computed
Tomography Scanner?
Zeev V. Maizlin, MD* and Patrick M. Vos, MDÞ

CT scanner, endangering the company’s financial assets (by


Abstract: It is commonly believed that the revenues from the selling of stepping into a new unknown foreign, mainly the United States,
the Beatles’ records by Electric and Musical Industries (EMI) allowed the market, that was controlled by other major well-established
company to develop the computed tomography (CT) scanner. Some went players), would have been a very risky business strategy that is
to define this as the Beatles’ gift to medicine. However, significant hardly ever successful. Even EMI’s history demonstrated that its
controversies and discrepancies arise from analysis of this statement, control over the company’s own American subsidiary was lim-
making its correctness doubtful. The details of financing required for the ited (at the time when EMI’s Parlophone was making records of
CT development and the part of EMI in financial input have never been the Beatles in Britain, EMI’s American subdivisionVCapitol
publicly announced. This work analyzes the financial contributions to the RecordsVdeclined to issue either of the first popular Beatles’
CT development and investigates if the revenues received from the sales singles (‘‘Please Please Me,’’ ‘‘From Me to You’’), and they were
of the Beatles’ records were used for the creation of the CT scanner. released through independent US labels.
Timeline of the development of the EMI CT scanner and the financial Third, before launching the development of CT, EMI had
inputs of EMI and British Department of Health and Social Security not consulted radiologists, neurologists, or neurosurgeons to get
(DHSS) were assessed. Without salary expenses to Godfrey Hounsfield their opinion and support of the new technology and took no
and his team, the development of the CT scanner cost EMI approximately steps to avoid a substantial risk that the new product would not
U100,000. The British DHSS’s expenses were U606,000. Hence, the fi- be accepted by the doctors and hospital administration; EMI
nancial contribution of DHSS into the development of the CT scanner had no previous significant experience in medical electronics
was significantly bigger than that of EMI. Accordingly, British tax payers production and sales. It had also sold off its computer divi-
and officials of British DHSS are to be thanked for the CT scanner. sion6 in 1962 (not a natural move for a company that would
The Beatles’ input into the world’s culture is valuable and does not initiate research and production involving a computer-based
require decoration by nonexistent connection to the development of CT. technology).
A positive aspect to this misconception is that it keeps in public memory These considerations question the accuracy of the claim
the name of the company that developed the CT scanner. that EMI did not know what to do with the money it got from
Key Words: CT scanner, Hounsfield, history selling the Beatles’ records and hence spent it developing the CT
scanner.
(J Comput Assist Tomogr 2012;36: 161Y164)
This work assesses the financial contributions to the CT
development and investigates if the revenues received from the
sales of the Beatles records were used for the development of the

I t is commonly believed that the revenues from the selling of


the Beatles records by Electric and Musical Industries (EMI)
allowed the company to finance its Central Research Laboratory
CT scanner.

(CRL) to develop a new medical diagnostic imaging device for BACKGROUND


the head scanVthe computed tomography (CT) scanner.1Y4 Electric and Musical Industries emerged from World War II
Some went to define this as the Beatles’ gift to medicine.5 with an experience in electronics, which was then largely related
However, there are significant controversies and dis- to defense-associated products. The transition to peacetime was
crepancies that arise from analysis of this statement, making its difficult. However, EMI did exciting pioneering work and tried
correctness doubtful. to become a leading computer company in Britain. With music
First, the financial costs and sources involved in the CT business booming, in 1955 EMI acquired Capitol Records in the
development were not clearly stated or publicly announced. To United States. This purchase and the subsequent success of the
some extent, it could be due to extensive litigation activity that recording groups, including the Beatles (Parlophone Records, an
followed the introduction of CT scanners in the 1970s. EMI subsidiary, signed the recording contract with the Fab Four
Second, the claim that flush with cash from selling the in 1962, paying them a minimal royalty rate of 1 farthing per
Beatles’ records, EMI went on to finance the development of the double-sided disc7), put the company in a very strong financial
position. As a result, in 1970, the company earned U21 million.
In 1967, Godfrey Hounsfield, a research scientist in EMI’s
*From the Centre for Medical and Radiology History and Department of CRL, conceived the idea of the reversed radar.8 In an attempt to
Radiology, McMaster University Medical Centre, Hamilton, Ontario; and find the practical application to this device, radiology came to
†Department of Radiology, St Paul’s Hospital, University of British Colum-
bia, Vancouver, British Columbia, Canada. his attention. This implied that the company would branch out
Received for publication November 20, 2011; accepted December 6, 2011. into the highly competitive field of medical electronics, where it
Reprints: Zeev V. Maizlin, MD, Centre for Medical and Radiology History essentially lacked experience offering only 2 small medical
and Department of Radiology, McMaster University Medical Centre, products (a patient-monitoring device and an infrared ther-
1200 Main St W, Hamilton, Ontario, Canada L8N 3Z5 (e-mail: zeev25@
yahoo.com). mography device), which together represented less than 0.5% of
The authors report no conflicts of interest. the company’s sales.9 It would be difficult to build a medical
Copyright * 2012 by Lippincott Williams & Wilkins imaging operation from scratch without knowledge of the North

J Comput Assist Tomogr & Volume 36, Number 2, March/April 2012 www.jcat.org 161

Copyright © 2012 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Maizlin and Vos J Comput Assist Tomogr & Volume 36, Number 2, March/April 2012

TABLE 1. Summary of the Funding Allocated to Development of the CT Scanner

Year Research and Engineering Period Financing Financing Source


1967 Hounsfield conceived the idea of the scanner Regular salary from CRL EMI
The salary paid to G. Hounsfield
for his work in CRL
(exact total sum unknown)
1968 ‘‘Lathe bed model’’ created from an old Team worked on a very low EMI
lathe that Hounsfield had been using in a budget of U25,000
previous project working on computer stores.
A lathe bed provided the means for moving
and rotating the gamma-ray source,
Americium 95, around the bottles or Perspex
jars with a photoncounter as the detector
placed on the other side. It took 9 days to
collect sufficient information of 28,000
measurements (and 2.5 h to reconstruct
the image on an ICL 1905 mainframe
computer). This work was done by a very
small team composed of G. Hounsfield,
Stephen Bates (programming), Peter Langstone
(electronics), and Mel King (mechanics)
1968 An x-ray tube was purchased and used, allowing U6000  2 = U12,000 EMI and the DHSS:
more controlled source, as well as having a for the acquisition of an U6000 each
much higher output, thus reducing data x-ray tube and a generator
acquisition time to 9 h
1968Y1969 Completion of the research and construction (1) Funding of the remaining DHSS: U600,000
of the prototype research costs (U69,000)
(2) Order for a prototype scanner EMI: the remaining
and 3 clinical machines research costs:
U150,000 for each: approximately U69,000
U150,000  4 = U600,000
1972 Manufacturing of the first scanners by EMI The DHHS purchased the first 3 DHSSVsee above
EMI scanners and placed them
in Manchester, Glasgow,
and London

American market, where most of the demand for scanners was to diagnosis.9 Electric and Musical Industries expected that the
be expected. Moreover, EMI’s experience in electronics work scanner cost would be around $400,000, and it was clear that
had been mainly in producing small numbers of highly spe- only the largest and financially strongest hospitals would be able
cialized defense products on government contracts. If a scanner to afford the scanner.
was going to be produced, most of the components would be To save on the development costs and to secure the initial
purchased from subcontractors and had to be integrated into a sale contract, EMI management decided to apply for the assis-
functioning system. A diversification move with unclear effect tance of the British government.
on the company’s prosperity was going to be dangerous. There Therefore, in 1968, EMI and G. Hounsfield approached the
was considerable disagreement among top management at EMI British Department of Health and Social Security (DHSS). They
regarding the CT scanner. Even Dr Len Broadway, head of the asked the Medical Research Council for development funds
CRL and one of the CT scanner’s earliest supporters, was among in exchange for a share of the profits. The government thought
the strongest opponents of EMI’s self-development of the new that it would be commendable foresight and agreed to support
business, being convinced that EMI’s potential competitors in EMI’s development of a head scanner. This development got a
the field had considerably greater technical capabilities and significant and vital support from the DHSS officials: E. Lennon
resources.9 and G. Higson.
Considerations of the CT scanner supporters were that in Dr Evan Lennon was a principal medical officer in radi-
the 1960s the radiology departments in many hospitals were ology at the DHSS. He connected between G. Hounsfield and
recognized as important money-making operators. Radiologists neuroradiologist Dr James Ambrose, after a few failed attempts
were able to make manufacturers build specially designed and when some reputable radiologists had refused to work with EMI
expensive. and Hounsfield on the project (one can only imagine what they
felt later when the CT scanner produced its clinical images!).
X-ray Systems and Applications With admirable support of Mr Gordon Higson, who worked
The size of the x-ray market was growing by almost 10% at the scientific and technical branch of the DHSS, the backing
annually. Initially, EMI estimated that they would be able to sell of EMI’s project was ensured. The DHSS agreed to fund half the
5 scanners in the first 12 months. This opinion then changed to remaining research costs in exchange for a small royalty on
12 and finally to 50 scanners. The company was encouraged by sales. At the time, it was calculated that it would cost U69,000
the opinion that the time might come when neurologists would to build a complete working system, and the DHSS committed to
feel ethically compelled to order a CT scan before making a pay U150,000 for each of the 4 systems, placing the order for a

162 www.jcat.org * 2012 Lippincott Williams & Wilkins

Copyright © 2012 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
J Comput Assist Tomogr & Volume 36, Number 2, March/April 2012 The Beatles and the Development of the CT Scanner

TABLE 2. Financial Summary for EMI (in U’000)*

1969 1970 1971 1972 1973 1974 1975 1976


EMI sales in medical electronics V V V V 321 5076 20,406 42,104
EMI profit (loss) from sales in medical electronics V V V V (67) 1242 9230 12,502
Profit attributable to ordinary stockholders 7259 8736 4562 7297 10,864 13,327 13,124 24,399
*Based on data from Bartlett.9

prototype and 3 clinical machines. It was intended to generate of the CT scanner. Since 1968 (starting from the White Album),
sufficient income to fund the fifth machine for Hounsfield and the group’s records were also released by a new Apple Corpo-
his team to keep and work on.10,11 ration founded by the Beatles. Moreover, in 1969, the band got
Dr Ambrose and Dr Louis Kreel supplied the human ca- from EMI (temporarily concealing the fact that John Lennon had
daver samples and provided the radiologic consult for the work. already effectively quit the group) the highest royalty rate ever
Hounsfield and his small team were stationed in the Radiology paid to a recording group up to that time.14
Department at the Atkinson Morley’s Hospital in Wimbledon. On the other hand, the development process and 4 CT
The location was chosen to avoid widespread publicity in the scanners (including the first) cost the DHSS at least U606,000
development phase.1 Thereafter, the development took place (U6000 + U600,000). It is hard to overestimate the value of this
largely in secret. government contract for EMI, which effectively allowed the
The first clinical trial was very successful. A prototype EMI company to complete the development and commence the pro-
head scanner (Mark I) in October 1971 provided the first clinical duction of the CT scanner. What is more, the whole development
image of a female patient of Atkinson Morley’s Hospital, re- was greatly and vitally supported by the British DHSS officials.
vealing the presence and location of a brain lesion.
CONCLUSIONS
Many factors, significantly more complex and diverse than
THE TIMELINE AND FINANCING OF THE the cash inflow from one of the groups, even as successful as the
DEVELOPMENT OF THE CT SCANNER Beatles, ensured the development of the CT scanner by EMI. It
Chronological stages and financial contribution of the CT would be inaccurate to claim that EMI did not have what to do
development are summarized in Table 1. with the money it got from selling the Beatles’ records. As usual,
Based on the available sources, it is difficult to establish if these profits were distributed to shareholders and used for the
U69,000, necessary for the funding of the remaining research company’s expansion in the familiar market of the music busi-
costs in 1968Y1969, were shared between EMI and the DHSS or ness. Moreover, there is no evidence to presume that the de-
paid entirely by EMI. To avoid the underestimation of EMI’s velopment of the CT scanner was the ‘‘gift’’ from the Beatles
expenses, for calculation purposes it was presumed that U69,000 because there is no confirmation that the group was ever in-
was paid entirely by EMI. Without salary expenses to G. volved in any way in the decision-making process of the com-
Hounsfield and his team, the development of the CT scanner pany’s management. The linkage between 2 successful business
cost EMI approximately U100,000 (U25,000 + U6000 + ventures of one of the eminent British corporations was very
U69,000). beneficial for EMI, helping it to be perceived as a justified (and
The salary component of EMI expenses cannot be precisely there is no doubting this) developer of the first practical CT
calculated because of the private character of this information. scanner.
However, hiring advertisements in that period12,13 mentioned the Comparing the financial contributions into the develop-
annual salary of engineers, physicists, and technicians to be in ment of the CT scanner from EMI and DHSS, the latter’s part
the range of U1500 to U2500. Assuming that G. Hounsfield was significantly bigger. Accordingly, British tax payers and
could be paid 4 times more and 3 members of his team could be officials of British DHSS are to be thanked for the CT scanner.
paid 2 times more than the upper range of salaries at that time, an It is clear for all Beatles’ fans (including the authors) that
approximate estimation of EMI salary expenses for the team is the group’s input into the world’s cultural heritage is valuable
U25,000 per annum or U100,000 for the years of 1968 to 1971. enough and does not require to be decorated by nonexistent
Accordingly, the development of the CT scanner cost EMI ap- connection to the development of the new revolutionary imaging
proximately U100,000 plus U100,000 of salaries to G. Houns- modality. Ironically, there was, however, a positive aspect to this
field and his team over the years 1967 to 1971. It constituted misconception. It helped to keep in the public’s (and young
total costs for EMI of U200,000Vless than 1% of the company’s radiology residents’) memory the name of the company that
profits (U20,557,000) in this period (Table 2) and could not be developed the CT scanner.
attributed to the revenues from the record sales of one of the
groups. The development of the CT scanner in 1968 to 1970 did ACKNOWLEDGMENT
not affect the funds available for distribution to shareholders in The authors thank Ms Monika Ferrier, BA, whose efforts
1969 to 1976.9 The drop in 1971 was attributed to extraordinary and professional expertise helped to enrich this article with
losses unrelated to CRL financing and was recovered already in important details. More details about development of the CT
1972. Later, EMI sales in the United States from the first scanner scanner are available in the book ‘‘Wonders of Radiology.’’
in midY1973 until 1975, when 160 machines were installed at an REFERENCES
average cost of U160,000 ($400,000), provided EMI with gain of
U25,600,000 ($64,000,000). 1. Isherwood I. Sir Godfrey Hounsfield. Radiology. 2005;234:975Y976.
Two important factors negatively affected the financial 2. Rogers LF. ‘‘My word, what is that?’’: Hounsfield and the triumph of
input from the Beatles to EMI in the period of the development clinical research. Am J Roentgenol. 2003;180:1501.

* 2012 Lippincott Williams & Wilkins www.jcat.org 163

Copyright © 2012 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Maizlin and Vos J Comput Assist Tomogr & Volume 36, Number 2, March/April 2012

3. Goodman LR. The Beatles, the Nobel Prize, and CT scanning of 8. Strong AB, Hurst RA. EMI patents on computed tomography: history
the chest. Radiol Clin N Am. 2010;48:1Y7. of legal actions. Br J Radiol. 1994;67:315Y316.
4. Berland LL. Commentary on ‘‘Computed tomography of the body: 9. Bartlett CA. EMI and the CT Scanner. Boston, MA, Harvard Business
initial clinical trial with the EMI prototype’’ and ‘‘Computed School Press. 1983. Available at: http://www.blackwellpublishing.com/
tomography of the body: early trends in application grant/docs/10EMI.pdf. Accessed December 10, 2010.
and accuracy of the method’’. Am J Roentgenol. 2008;
10. Beckmann EC. CT scanning the early days. Br J Radiol. 2006;79:5Y8.
191:16Y18.
11. Süsskind C. The invention of computed tomography. Hist Technol.
5. The Beatles gift to medicine. Available at: http://
1981;6:39Y80.
doctorrw.blogspot.com/2007/03/beatles-gift-to-medicine.html.
Accessed December 10, 2010. 12. Positions available. New Scientist. April, 30, 1964:323.
6. The Beatles. 1960Y1963 Business. Available at: http:// 13. Positions available. New Scientist. December, 31, 1970:620.
www.independent.co.uk/news/obituaries/sir-godfrey-hounsfield- 14. Biography for The Beatles. Available at: http://webcache.
550312.html. Accessed December 10, 2010. googleusercontent.com/search?hl=en&start=40&q=cache:
7. Available at: http://www.beatlemoney.com/beatles6063business.htm. Nqf FU9cI8MAJ; http://www.imdb.com/name/nm1397313/bio+
Accessed September 23, 2011 beatles+sales+peak+records+emi&ct=clnk. Accessed December 4, 2011.

164 www.jcat.org * 2012 Lippincott Williams & Wilkins

Copyright © 2012 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Cell, Vol. 100, 57–70, January 7, 2000, Copyright 2000 by Cell Press

The Hallmarks of Cancer Review

Douglas Hanahan* and Robert A. Weinberg† evolve progressively from normalcy via a series of pre-
* Department of Biochemistry and Biophysics and malignant states into invasive cancers (Foulds, 1954).
Hormone Research Institute These observations have been rendered more con-
University of California at San Francisco crete by a large body of work indicating that the ge-
San Francisco, California 94143 nomes of tumor cells are invariably altered at multiple
† Whitehead Institute for Biomedical Research and sites, having suffered disruption through lesions as sub-
Department of Biology tle as point mutations and as obvious as changes in
Massachusetts Institute of Technology chromosome complement (e.g., Kinzler and Vogelstein,
Cambridge, Massachusetts 02142 1996). Transformation of cultured cells is itself a
multistep process: rodent cells require at least two intro-
duced genetic changes before they acquire tumorigenic
After a quarter century of rapid advances, cancer re- competence, while their human counterparts are more
search has generated a rich and complex body of knowl- difficult to transform (Hahn et al., 1999). Transgenic
edge, revealing cancer to be a disease involving dy- models of tumorigenesis have repeatedly supported the
namic changes in the genome. The foundation has been conclusion that tumorigenesis in mice involves multiple
set in the discovery of mutations that produce onco- rate-limiting steps (Bergers et al., 1998; see Oncogene,
genes with dominant gain of function and tumor sup- 1999, R. DePinho and T. E. Jacks, volume 18[38], pp.
pressor genes with recessive loss of function; both 5248–5362). Taken together, observations of human
classes of cancer genes have been identified through cancers and animal models argue that tumor develop-
their alteration in human and animal cancer cells and ment proceeds via a process formally analogous to Dar-
by their elicitation of cancer phenotypes in experimental winian evolution, in which a succession of genetic
models (Bishop and Weinberg, 1996). changes, each conferring one or another type of growth
Some would argue that the search for the origin and advantage, leads to the progressive conversion of nor-
treatment of this disease will continue over the next mal human cells into cancer cells (Foulds, 1954; Nowell,
quarter century in much the same manner as it has in 1976).
the recent past, by adding further layers of complexity
to a scientific literature that is already complex almost An Enumeration of the Traits
beyond measure. But we anticipate otherwise: those The barriers to development of cancer are embodied
researching the cancer problem will be practicing a dra- in a teleology: cancer cells have defects in regulatory
matically different type of science than we have experi- circuits that govern normal cell proliferation and homeo-
enced over the past 25 years. Surely much of this change stasis. There are more than 100 distinct types of cancer,
will be apparent at the technical level. But ultimately, and subtypes of tumors can be found within specific
the more fundamental change will be conceptual. organs. This complexity provokes a number of ques-
We foresee cancer research developing into a logical tions. How many distinct regulatory circuits within each
science, where the complexities of the disease, de- type of target cell must be disrupted in order for such
scribed in the laboratory and clinic, will become under- a cell to become cancerous? Does the same set of
standable in terms of a small number of underlying prin- cellular regulatory circuits suffer disruption in the cells
ciples. Some of these principles are even now in the of the disparate neoplasms arising in the human body?
midst of being codified. We discuss one set of them in Which of these circuits operate on a cell-autonomous
the present essay: rules that govern the transformation basis, and which are coupled to the signals that cells
of normal human cells into malignant cancers. We sug- receive from their surrounding microenvironment within
gest that research over the past decades has revealed a tissue? Can the large and diverse collection of cancer-
a small number of molecular, biochemical, and cellular associated genes be tied to the operations of a small
traits—acquired capabilities—shared by most and per- group of regulatory circuits?
haps all types of human cancer. Our faith in such simplifi- We suggest that the vast catalog of cancer cell geno-
cation derives directly from the teachings of cell biology types is a manifestation of six essential alterations in cell
that virtually all mammalian cells carry a similar molecu- physiology that collectively dictate malignant growth
lar machinery regulating their proliferation, differentia- (Figure 1): self-sufficiency in growth signals, insensitivity
tion, and death. to growth-inhibitory (antigrowth) signals, evasion of pro-
Several lines of evidence indicate that tumorigenesis grammed cell death (apoptosis), limitless replicative
in humans is a multistep process and that these steps potential, sustained angiogenesis, and tissue invasion
reflect genetic alterations that drive the progressive and metastasis. Each of these physiologic changes—
transformation of normal human cells into highly malig- novel capabilities acquired during tumor development—
nant derivatives. Many types of cancers are diagnosed represents the successful breaching of an anticancer
in the human population with an age-dependent inci- defense mechanism hardwired into cells and tissues.
dence implicating four to seven rate-limiting, stochastic We propose that these six capabilities are shared in
events (Renan, 1993). Pathological analyses of a number common by most and perhaps all types of human tu-
of organ sites reveal lesions that appear to represent mors. This multiplicity of defenses may explain why can-
the intermediate steps in a process through which cells cer is relatively rare during an average human lifetime.
Cell
58

Acquired GS autonomy was the first of the six capabili-


ties to be clearly defined by cancer researchers, in large
part because of the prevalence of dominant oncogenes
that have been found to modulate it. Three common
molecular strategies for achieving autonomy are evi-
dent, involving alteration of extracellular growth signals,
of transcellular transducers of those signals, or of intra-
cellular circuits that translate those signals into action.
While most soluble mitogenic growth factors (GFs) are
made by one cell type in order to stimulate proliferation
of another—the process of heterotypic signaling—many
cancer cells acquire the ability to synthesize GFs to
which they are responsive, creating a positive feedback
signaling loop often termed autocrine stimulation (Fedi
et al., 1997). Clearly, the manufacture of a GF by a cancer
cell obviates dependence on GFs from other cells within
the tissue. The production of PDGF (platelet-derived
growth factor) and TGF␣ (tumor growth factor ␣) by
glioblastomas and sarcomas, respectively, are two illus-
trative examples (Fedi et al., 1997).
The cell surface receptors that transduce growth-
stimulatory signals into the cell interior are themselves
targets of deregulation during tumor pathogenesis. GF
receptors, often carrying tyrosine kinase activities in
their cytoplasmic domains, are overexpressed in many
cancers. Receptor overexpression may enable the can-
Figure 1. Acquired Capabilities of Cancer cer cell to become hyperresponsive to ambient levels
We suggest that most if not all cancers have acquired the same set of GF that normally would not trigger proliferation (Fedi
of functional capabilities during their development, albeit through et al., 1997). For example, the epidermal GF receptor
various mechanistic strategies. (EGF-R/erbB) is upregulated in stomach, brain, and
breast tumors, while the HER2/neu receptor is overex-
pressed in stomach and mammary carcinomas (Slamon
We describe each capability in turn below, illustrate with et al., 1987; Yarden and Ullrich, 1988). Additionally, gross
a few examples its functional importance, and indicate overexpression of GF receptors can elicit ligand-inde-
strategies by which it is acquired in human cancers. pendent signaling (DiFiore et al., 1987). Ligand-indepen-
dent signaling can also be achieved through structural
alteration of receptors; for example, truncated versions
Acquired Capability: Self-Sufficiency
of the EGF receptor lacking much of its cytoplasmic
in Growth Signals
domain fire constitutively (Fedi et al., 1997).
Normal cells require mitogenic growth signals (GS) be-
Cancer cells can also switch the types of extracellular
fore they can move from a quiescent state into an active
matrix receptors (integrins) they express, favoring ones
proliferative state. These signals are transmitted into the
that transmit progrowth signals (Lukashev and Werb,
cell by transmembrane receptors that bind distinctive 1998; Giancotti and Ruoslahti, 1999). These bifunctional,
classes of signaling molecules: diffusible growth fac- heterodimeric cell surface receptors physically link cells
tors, extracellular matrix components, and cell-to-cell to extracellular superstructures known as the extracellu-
adhesion/interaction molecules. To our knowledge, no lar matrix (ECM). Successful binding to specific moieties
type of normal cell can proliferate in the absence of of the ECM enables the integrin receptors to transduce
such stimulatory signals. Many of the oncogenes in the signals into the cytoplasm that influence cell behavior,
cancer catalog act by mimicking normal growth signal- ranging from quiescence in normal tissue to motility,
ing in one way or another. resistance to apoptosis, and entrance into the active
Dependence on growth signaling is apparent when cell cycle. Conversely, the failure of integrins to forge
propagating normal cells in culture, which typically pro- these extracellular links can impair cell motility, induce
liferate only when supplied with appropriate diffusible apoptosis, or cause cell cycle arrest (Giancotti and Ru-
mitogenic factors and a proper substratum for their inte- oslahti, 1999). Both ligand-activated GF receptors and
grins. Such behavior contrasts strongly with that of tu- progrowth integrins engaged to extracellular matrix
mor cells, which invariably show a greatly reduced components can activate the SOS-Ras-Raf-MAP kinase
dependence on exogenous growth stimulation. The con- pathway (Aplin et al., 1998; Giancotti and Ruoslahti,
clusion is that tumor cells generate many of their own 1999).
growth signals, thereby reducing their dependence on The most complex mechanisms of acquired GS auton-
stimulation from their normal tissue microenvironment. omy derive from alterations in components of the down-
This liberation from dependence on exogenously de- stream cytoplasmic circuitry that receives and pro-
rived signals disrupts a critically important homeostatic cesses the signals emitted by ligand-activated GF
mechanism that normally operates to ensure a proper receptors and integrins. The SOS-Ras-Raf-MAPK cas-
behavior of the various cell types within a tissue. cade plays a central role here. In about 25% of human
Review
59

Figure 2. The Emergent Integrated Circuit of the Cell


Progress in dissecting signaling pathways has begun to lay out a circuitry that will likely mimic electronic integrated circuits in complexity
and finesse, where transistors are replaced by proteins (e.g., kinases and phosphatases) and the electrons by phosphates and lipids, among
others. In addition to the prototypical growth signaling circuit centered around Ras and coupled to a spectrum of extracellular cues, other
component circuits transmit antigrowth and differentiation signals or mediate commands to live or die by apoptosis. As for the genetic
reprogramming of this integrated circuit in cancer cells, some of the genes known to be functionally altered are highlighted in red.

tumors, Ras proteins are present in structurally altered multiple cell biological effects. For example, the direct
forms that enable them to release a flux of mitogenic interaction of the Ras protein with the survival-promot-
signals into cells, without ongoing stimulation by their ing PI3 kinase enables growth signals to concurrently
normal upstream regulators (Medema and Bos, 1993). evoke survival signals within the cell (Downward, 1998).
We suspect that growth signaling pathways suffer While acquisition of growth signaling autonomy by
deregulation in all human tumors. Although this point cancer cells is conceptually satisfying, it is also too
is hard to prove rigorously at present, the clues are simplistic. We have traditionally explored tumor growth
abundant (Hunter, 1997). For example, in the best stud- by focusing our experimental attentions on the geneti-
ied of tumors—human colon carcinomas—about half cally deranged cancer cells (Figure 3, left panel). It is,
of the tumors bear mutant ras oncogenes (Kinzler and however, increasingly apparent that the growth deregu-
Vogelstein, 1996). We suggest that the remaining colonic lation within a tumor can only be explained once we
tumors carry defects in other components of the growth understand the contributions of the ancillary cells pres-
signaling pathways that phenocopy ras oncogene acti- ent in a tumor—the apparently normal bystanders such
vation. The nature of these alternative, growth-stimulat- as fibroblasts and endothelial cells—which must play
ing mechanisms remains elusive. key roles in driving tumor cell proliferation (Figure 3,
Under intensive study for two decades, the wiring right panel). Within normal tissue, cells are largely in-
diagram of the growth signaling circuitry of the mamma- structed to grow by their neighbors (paracrine signals)
lian cell is coming into focus (Figure 2). New downstream or via systemic (endocrine) signals. Cell-to-cell growth
effector pathways that radiate from the central SOS- signaling is likely to operate in the vast majority of human
Ras-Raf-MAP kinase mitogenic cascade are being dis- tumors as well; virtually all are composed of several
covered with some regularity (Hunter, 1997; Rommel distinct cell types that appear to communicate via het-
and Hafen, 1998). This cascade is also linked via a variety erotypic signaling.
of cross-talking connections with other pathways; these Heterotypic signaling between the diverse cell types
cross connections enable extracellular signals to elicit within a tumor may ultimately prove to be as important
Cell
60

Figure 3. Tumors as Complex Tissues


The field of cancer research has largely been
guided by a reductionist focus on cancer cells
and the genes within them (left panel)—a fo-
cus that has produced an extraordinary body
of knowledge. Looking forward in time, we
believe that important new inroads will come
from regarding tumors as complex tissues in
which mutant cancer cells have conscripted
and subverted normal cell types to serve as
active collaborators in their neoplastic agenda
(right panel). The interactions between the
genetically altered malignant cells and these
supporting coconspirators will prove critical
to understanding cancer pathogenesis and to
the development of novel, effective therapies.

in explaining tumor cell proliferation as the cancer cell- the components governing the transit of the cell through
autonomous mechanisms enumerated above. For ex- the G1 phase of its growth cycle. Cells monitor their
ample, we suspect that many of the growth signals driv- external environment during this period and, on the ba-
ing the proliferation of carcinoma cells originate from sis of sensed signals, decide whether to proliferate, to
the stromal cell components of the tumor mass. While be quiescent, or to enter into a postmitotic state. At the
difficult to validate at present, such thinking recasts the molecular level, many and perhaps all antiproliferative
logic of acquired GS autonomy: successful tumor cells signals are funneled through the retinoblastoma protein
are those that have acquired the ability to co-opt their (pRb) and its two relatives, p107 and p130. When in a
normal neighbors by inducing them to release abundant hypophosphorylated state, pRb blocks proliferation by
fluxes of growth-stimulating signals (Skobe and Fu- sequestering and altering the function of E2F transcrip-
senig, 1998). Indeed, in some tumors, these cooperating tion factors that control the expression of banks of genes
cells may eventually depart from normalcy, coevolving essential for progression from G1 into S phase (Wein-
with their malignant neighbors in order to sustain the berg, 1995).
growth of the latter (Kinzler and Vogelstein, 1998; Olumi Disruption of the pRb pathway liberates E2Fs and
et al., 1999). Further, inflammatory cells attracted to sites thus allows cell proliferation, rendering cells insensitive
of neoplasia may promote (rather than eliminate) cancer to antigrowth factors that normally operate along this
cells (Cordon-Cardo and Prives, 1999; Coussens et al., pathway to block advance through the G1 phase of the
1999; Hudson et al., 1999), another example of normal cell cycle. The effects of the soluble signaling molecule
cells conscripted to enhance tumor growth potential, TGF␤ are the best documented, but we envision other
another means to acquire necessary capabilities. antigrowth factors will be found to signal through this
pathway as well. TGF␤ acts in a number of ways, most
Acquired Capability: Insensitivity still elusive, to prevent the phosphorylation that inacti-
to Antigrowth Signals vates pRb; in this fashion, TGF␤ blocks advance through
Within a normal tissue, multiple antiproliferative signals G1. In some cell types, TGF␤ suppresses expression
operate to maintain cellular quiescence and tissue ho- of the c-myc gene, which regulates the G1 cell cycle
meostasis; these signals include both soluble growth machinery in still unknown ways (Moses et al., 1990).
inhibitors and immobilized inhibitors embedded in the More directly, TGF␤ causes synthesis of the p15INK4B and
extracellular matrix and on the surfaces of nearby cells. p21 proteins, which block the cyclin:CDK complexes
These growth-inhibitory signals, like their positively act- responsible for pRb phosphorylation (Hannon and
ing counterparts, are received by transmembrane cell Beach, 1994; Datto et al., 1997).
surface receptors coupled to intracellular signaling cir- The pRb signaling circuit, as governed by TGF␤ and
cuits. other extrinsic factors, can be disrupted in a variety of
Antigrowth signals can block proliferation by two dis- ways in different types of human tumors (Fynan and
tinct mechanisms. Cells may be forced out of the active Reiss, 1993). Some lose TGF␤ responsiveness through
proliferative cycle into the quiescent (G0) state from downregulation of their TGF␤ receptors, while others
which they may reemerge on some future occasion display mutant, dysfunctional receptors (Fynan and
when extracellular signals permit. Alternatively, cells Reiss, 1993; Markowitz et al., 1995). The cytoplasmic
may be induced to permanently relinquish their prolifera- Smad4 protein, which transduces signals from ligand-
tive potential by being induced to enter into postmitotic activated TGF␤ receptors to downstream targets, may
states, usually associated with acquisition of specific be eliminated through mutation of its encoding gene
differentiation-associated traits. (Schutte et al., 1996). The locus encoding p15INK4B may be
Incipient cancer cells must evade these antiprolifera- deleted (Chin et al., 1998). Alternatively, the immediate
tive signals if they are to prosper. Much of the circuitry downstream target of its actions, CDK4, may become
that enables normal cells to respond to antigrowth sig- unresponsive to the inhibitory actions of p15INK4B be-
nals is associated with the cell cycle clock, specifically cause of mutations that create amino acid substitutions
Review
61

in its INK4A/B-interacting domain; the resulting cyclin in virtually all cell types throughout the body. Once trig-
D:CDK4 complexes are then given a free hand to inacti- gered by a variety of physiologic signals, this program
vate pRb by hyperphosphorylation (Zuo et al., 1996). unfolds in a precisely choreographed series of steps.
Finally, functional pRb, the end target of this pathway, Cellular membranes are disrupted, the cytoplasmic and
may be lost through mutation of its gene. Alternatively, nuclear skeletons are broken down, the cytosol is ex-
in certain DNA virus-induced tumors, notably cervical truded, the chromosomes are degraded, and the nu-
carcinomas, pRb function is eliminated through seques- cleus is fragmented, all in a span of 30–120 min. In the
tration by viral oncoproteins, such as the E7 oncoprotein end, the shriveled cell corpse is engulfed by nearby cells
of human papillomavirus (Dyson et al., 1989). In addition, in a tissue and disappears, typically within 24 hr (Wyllie
cancer cells can also turn off expression of integrins and et al., 1980).
other cell adhesion molecules that send antigrowth sig- The apoptotic machinery can be broadly divided into
nals, favoring instead those that convey progrowth sig- two classes of components—sensors and effectors. The
nals; these adherence-based antigrowth signals likely sensors are responsible for monitoring the extracellular
impinge on the pRb circuit as well. The bottom line is and intracellular environment for conditions of normality
that the antigrowth circuit converging onto Rb and the or abnormality that influence whether a cell should live
cell division cycle is, one way or another, disrupted in or die. These signals regulate the second class of com-
a majority of human cancers, defining the concept and ponents, which function as effectors of apoptotic death.
a purpose of tumor suppressor loss in cancer. The sentinels include cell surface receptors that bind
Cell proliferation depends on more than an avoidance survival or death factors. Examples of these ligand/
of cytostatic antigrowth signals. Our tissues also con- receptor pairs include survival signals conveyed by IGF-
strain cell multiplication by instructing cells to enter irre- 1/IGF-2 through their receptor, IGF-1R, and by IL-3 and
versibly into postmitotic, differentiated states, using di- its cognate receptor, IL-3R (Lotem and Sachs, 1996;
verse mechanisms that are incompletely understood; it Butt et al., 1999). Death signals are conveyed by the
is apparent that tumor cells use various strategies to FAS ligand binding the FAS receptor and by TNF␣ bind-
avoid this terminal differentiation. One strategy for ing TNF-R1 (Ashkenazi and Dixit, 1999). Intracellular
avoiding differentiation directly involves the c-myc on- sensors monitor the cell’s well-being and activate the
cogene, which encodes a transcription factor. During death pathway in response to detecting abnormalities,
normal development, the growth-stimulating action of including DNA damage, signaling imbalance provoked
Myc, in association with another factor, Max, can be by oncogene action, survival factor insufficiency, or hyp-
supplanted by alternative complexes of Max with a oxia (Evan and Littlewood, 1998). Further, the life of most
group of Mad transcription factors; the Mad–Max com- cells is in part maintained by cell–matrix and cell–cell
plexes elicit differentiation-inducing signals (Foley and adherence-based survival signals whose abrogation
Eisenman, 1999). However, overexpression of the c-Myc elicits apoptosis (Ishizaki et al., 1995; Giancotti and Ru-
oncoprotein, as is seen in many tumors, can reverse this oslahti, 1999). Both soluble and immobilized apoptotic
process, shifting the balance back to favor Myc–Max regulatory signals likely reflect the needs of tissues to
complexes, thereby impairing differentiation and pro- maintain their constituent cells in appropriate architec-
moting growth. During human colon carcinogenesis, in- tural configurations.
activation of the APC/␤-catenin pathway serves to block Many of the signals that elicit apoptosis converge
the egress of enterocytes in the colonic crypts into a on the mitochondria, which respond to proapoptotic
differentiated, postmitotic state (Kinzler and Vogelstein, signals by releasing cytochrome C, a potent catalyst of
1996). Analogously, during the generation of avian eryth- apoptosis (Green and Reed, 1998). Members of the Bcl-2
roblastosis, the erbA oncogene acts to prevent irrevers- family of proteins, whose members have either pro-
ible erythrocyte differentiation (Kahn et al., 1986). apoptotic (Bax, Bak, Bid, Bim) or antiapoptotic (Bcl-2,
While the components and interconnections between Bcl-XL, Bcl-W) function, act in part by governing mito-
the various antigrowth and differentiation-inducing sig- chondrial death signaling through cytochrome C re-
nals and the core cell cycle machinery are still being lease. The p53 tumor suppressor protein can elicit apo-
delineated, the existence of an antigrowth signaling cir- ptosis by upregulating expression of proapoptotic Bax
cuitry is clear (Figure 2), as is the necessity for its circum- in response to sensing DNA damage; Bax in turn stimu-
vention by developing cancers. lates mitochondria to release cytochrome C.
The ultimate effectors of apoptosis include an array
Acquired Capability: Evading Apoptosis of intracellular proteases termed caspases (Thornberry
The ability of tumor cell populations to expand in number and Lazebnik, 1998). Two “gatekeeper” caspases, ⫺8
is determined not only by the rate of cell proliferation and ⫺9, are activated by death receptors such as FAS
but also by the rate of cell attrition. Programmed cell or by the cytochrome C released from mitochondria,
death—apoptosis—represents a major source of this respectively. These proximal caspases trigger the acti-
attrition. The evidence is mounting, principally from vation of a dozen or more effector caspases that execute
studies in mouse models and cultured cells, as well as the death program, through selective destruction of sub-
from descriptive analyses of biopsied stages in human cellular structures and organelles, and of the genome.
carcinogenesis, that acquired resistance toward apo- The possibility that apoptosis serves as a barrier to
ptosis is a hallmark of most and perhaps all types of cancer was first raised in 1972, when Kerr, Wyllie, and
cancer. Currie described massive apoptosis in the cells populat-
Observations accumulated over the past decade indi- ing rapidly growing, hormone-dependent tumors follow-
cate that the apoptotic program is present in latent form ing hormone withdrawal (Kerr et al., 1972). The discovery
Cell
62

of the bcl-2 oncogene by its upregulation via chromo- abnormalities, including hypoxia and oncogene hyper-
somal translocation in follicular lymphoma (reviewed in expression, are also funneled in part via p53 to the apo-
Korsmeyer, 1992) and its recognition as having anti- ptotic machinery; these too are impaired at eliciting
apoptotic activity (Vaux et al., 1988) opened up the in- apoptosis when p53 function is lost (Levine, 1997). Addi-
vestigation of apoptosis in cancer at the molecular level. tionally, the PI3 kinase–AKT/PKB pathway, which trans-
When coexpressed with a myc oncogene in transgenic mits antiapoptotic survival signals, is likely involved in
mice, the bcl-2 gene was able to promote formation of mitigating apoptosis in a substantial fraction of human
B cell lymphomas by enhancing lymphocyte survival, not tumors. This survival signaling circuit can be activated
by further stimulating their myc-induced proliferation by extracellular factors such as IGF-1/2 or IL-3 (Evan
(Strasser et al., 1990); further, 50% of the infrequent and Littlewood, 1998), by intracellular signals emanating
lymphomas arising in bcl-2 single transgenic transgenic from Ras (Downward, 1998), or by loss of the pTEN
mice had somatic translocations activating c-myc, con- tumor suppressor, a phospholipid phosphatase that
firming a selective pressure during lymphomagenesis normally attenuates the AKT survival signal (Cantley and
to upregulate both Bcl-2 and c-Myc (McDonnell and Neel, 1999). Recently, a mechanism for abrogating the
Korsmeyer, 1991). FAS death signal has been revealed in a high fraction
Further insight into the myc-bcl-2 interaction emerged of lung and colon carcinoma cell lines: a nonsignaling
later from studying the effects of a myc oncogene on decoy receptor for FAS ligand is upregulated, titrating
fibroblasts cultured in low serum. Widespread apoptosis the death-inducing signal away from the FAS death re-
was induced in myc-expressing cells lacking serum; the ceptor (Pitti et al., 1998). We expect that virtually all
consequent apoptosis could be abrogated by exoge- cancer cells harbor alterations that enable evasion of
nous survival factors (e.g., IGF-1), by forced overexpres- apoptosis.
sion of Bcl-2 or the related Bcl-XL protein, or by disrup- It is now possible to lay out a provisional apoptotic
tion of the FAS death signaling circuit (Hueber et al., signaling circuitry (Figure 2); while incomplete, it is evi-
dent that most regulatory and effector components are
1997). Collectively, the data indicate that a cell’s apo-
present in redundant form. This redundancy holds im-
ptotic program can be triggered by an overexpressed
portant implications for the development of novel types
oncogene. Indeed, elimination of cells bearing activated
of antitumor therapy, since tumor cells that have lost
oncogenes by apoptosis may represent the primary
proapoptotic components are likely to retain other simi-
means by which such mutant cells are continually culled
lar ones. We anticipate that new technologies will be
from the body’s tissues.
able to display the apoptotic pathways still operative in
Other examples strengthen the consensus that apo-
specific types of cancer cells and that new drugs will
ptosis is a major barrier to cancer that must be circum-
enable cross-talk between the still intact components
vented. Thus, in transgenic mice where the pRb tumor
of parallel apoptotic signaling pathways in tumor cells,
suppressor was functionally inactivated in the choroid
resulting in restoration of the apoptotic defense mecha-
plexus, slowly growing microscopic tumors arose, ex- nism, with substantial therapeutic benefit.
hibiting high apoptotic rates; the additional inactivation
of the p53 tumor suppressor protein, a component of
Acquired Capability: Limitless Replicative Potential
the apoptotic signaling circuitry, led to rapidly growing
Three acquired capabilities—growth signal autonomy,
tumors containing low numbers of apoptotic cells (Sy-
insensitivity to antigrowth signals, and resistance to
monds et al., 1994). The role of extracellular survival
apoptosis—all lead to an uncoupling of a cell’s growth
factors is illustrated by disease progression in trans-
program from signals in its environment. In principle,
genic mice prone to pancreatic islet tumors. If IGF-2 the resulting deregulated proliferation program should
gene expression, which is activated in this tumorigene- suffice to enable the generation of the vast cell popula-
sis pathway, was abrogated using gene knockout mice, tions that constitute macroscopic tumors. However, re-
tumor growth and progression were impaired, as evi- search performed over the past 30 years indicates that
denced by the appearance of comparatively small, be- this acquired disruption of cell-to-cell signaling, on its
nign tumors showing high rates of apoptosis (Christofori own, does not ensure expansive tumor growth. Many
et al., 1994). In these cells, the absence of IGF-2 did not and perhaps all types of mammalian cells carry an intrin-
affect cell proliferation rates, clearly identifying it as an sic, cell-autonomous program that limits their multiplica-
antiapoptotic survival factor. Collectively, these obser- tion. This program appears to operate independently of
vations argue that altering components of the apoptotic the cell-to-cell signaling pathways described above. It
machinery can dramatically affect the dynamics of tu- too must be disrupted in order for a clone of cells to
mor progression, providing a rationale for the inactiva- expand to a size that constitutes a macroscopic, life-
tion of this machinery during tumor development. threatening tumor.
Resistance to apoptosis can be acquired by cancer The early work of Hayflick demonstrated that cells in
cells through a variety of strategies. Surely, the most culture have a finite replicative potential (reviewed in
commonly occurring loss of a proapoptotic regulator Hayflick, 1997). Once such cell populations have pro-
through mutation involves the p53 tumor suppressor gressed through a certain number of doublings, they
gene. The resulting functional inactivation of its product, stop growing—a process termed senescence. The se-
the p53 protein, is seen in greater than 50% of human nescence of cultured human fibroblasts can be circum-
cancers and results in the removal of a key component vented by disabling their pRb and p53 tumor suppressor
of the DNA damage sensor that can induce the apoptotic proteins, enabling these cells to continue multiplying for
effector cascade (Harris, 1996). Signals evoked by other additional generations until they enter into a second
Review
63

state termed crisis. The crisis state is characterized by threshold, and this in turn permits unlimited multiplica-
massive cell death, karyotypic disarray associated with tion of descendant cells. Both mechanisms seem to be
end-to-end fusion of chromosomes, and the occasional strongly suppressed in most normal human cells in order
emergence of a variant (1 in 107) cell that has acquired to deny them unlimited replicative potential.
the ability to multiply without limit, the trait termed im- The role of telomerase in immortalizing cells can be
mortalization (Wright et al., 1989). demonstrated directly by ectopically expressing the en-
Provocatively, most types of tumor cells that are prop- zyme in cells, where it can convey unlimited replicative
agated in culture appear to be immortalized, suggesting potential onto a variety of normal early passage, prese-
that limitless replicative potential is a phenotype that nescent cells in vitro (Bodnar et al., 1998; Vaziri and
was acquired in vivo during tumor progression and was Benchimol, 1998). Further, late passage cells poised to
essential for the development of their malignant growth enter crisis continue to proliferate without giving any
state (Hayflick, 1997). This result suggests that at some evidence of crisis when supplied with this enzyme
point during the course of multistep tumor progression, (Counter et al., 1998; Halvorsen et al., 1999; Zhu et al.,
evolving premalignant cell populations exhaust their en- 1999). Additional clues into the importance of telomere
dowment of allowed doublings and can only complete maintenance for cancer comes from analysis of mice
their tumorigenic agenda by breaching the mortality bar- lacking telomerase function. For example, mice carrying
rier and acquiring unlimited replicative potential. a homozygous knockout of the cell cycle inhibitor
Observations of cultured cells indicate that various p16INK4A are tumor prone, particularly when exposed to
normal human cell types have the capacity for 60–70 carcinogens; the tumors that arise show comparatively
doublings. Taken at face value, these numbers make elevated telomerase activity. When carcinogens were
little sense when attempting to invoke cell mortality as applied to p16INK4A-null mice that also lacked telomerase,
an impediment to cancer formation: 60–70 doublings tumor incidence was reduced, concomitant with sub-
should enable clones of tumor cells to expand to num- stantial telomere shortening and karyotypic disarray in
bers that vastly exceed the number of cells in the human those tumors that did appear (Greenberg et al., 1999).
body. If clues from evaluation of proliferation and apo- While telomere maintenance is clearly a key compo-
ptotic rates in certain human tumors (Wyllie et al., 1980) nent of the capability for unlimited replication, we remain
and transgenic mouse models (Symonds et al., 1994; uncertain about another one, the circumvention of cellu-
Shibata et al., 1996; Bergers et al., 1998) prove generaliz- lar senescence. The phenomenon of senescence was
able, the paradox can be resolved: evolving premalig- originally observed as a delayed response of primary
nant and malignant cell populations evidence chronic, cells to extended propagation in vitro and has thus been
widespread apoptosis and consequently suffer consid- associated with mechanisms of divisional counting
erable cell attrition concomitant with cell accumulation. (Hayflick, 1997). More recently, the senescent state has
Thus, the number of cells in a tumor greatly underrepre- been observed to be inducible in certain cultured cells
sents the cell generations required to produce it, raising in response to high level expression of genes such as
the generational limit of normal somatic cells as a barrier the activated ras oncogene (Serrano et al., 1997).
to cancer. The above-cited observations might argue that senes-
The counting device for cell generations has been cence, much like apoptosis, reflects a protective mecha-
discovered over the past decade: the ends of chromo- nism that can be activated by shortened telomeres or
somes, telomeres, which are composed of several thou- conflicting growth signals that forces aberrant cells irre-
sand repeats of a short 6 bp sequence element. Replica- versibly into a G0-like state, thereby rendering them inca-
tive generations are counted by the 50–100 bp loss of
pable of further proliferation. If so, circumvention of se-
telomeric DNA from the ends of every chromosome dur-
nescence in vivo may indeed represent an essential step
ing each cell cycle. This progressive shortening has
in tumor progression that is required for the subsequent
been attributed to the inability of DNA polymerases to
approach to and breaching of the crisis barrier. But we
completely replicate the 3⬘ ends of chromosomal DNA
consider an alternative model equally plausible: senes-
during each S phase. The progressive erosion of telo-
cence could be an artifact of cell culture that does not
meres through successive cycles of replication eventu-
reflect a phenotype of cells within living tissues and
ally causes them to lose their ability to protect the ends
does not represent an impediment to tumor progression
of chromosomal DNA. The unprotected chromosomal
in vivo. Resolution of this quandary will be critical to
ends participate in end-to-end chromosomal fusions,
yielding the karyotypic disarray associated with crisis completely understand the acquisition of limitless repli-
and resulting, almost inevitably, in the death of the af- cative potential.
fected cell (Counter et al., 1992).
Telomere maintenance is evident in virtually all types of Acquired Capability: Sustained Angiogenesis
malignant cells (Shay and Bacchetti, 1997); 85%–90% The oxygen and nutrients supplied by the vasculature
of them succeed in doing so by upregulating expression are crucial for cell function and survival, obligating virtu-
of the telomerase enzyme, which adds hexanucleotide ally all cells in a tissue to reside within 100 ␮m of a
repeats onto the ends of telomeric DNA (Bryan and capillary blood vessel. During organogenesis, this close-
Cech, 1999), while the remainder have invented a way ness is ensured by coordinated growth of vessels and
of activating a mechanism, termed ALT, which appears parenchyma. Once a tissue is formed, the growth of
to maintain telomeres through recombination-based in- new blood vessels—the process of angiogenesis—is
terchromosomal exchanges of sequence information transitory and carefully regulated. Because of this de-
(Bryan et al., 1995). By one or the other mechanism, pendence on nearby capillaries, it would seem plausible
telomeres are maintained at a length above a critical that proliferating cells within a tissue would have an
Cell
64

intrinsic ability to encourage blood vessel growth. But case angiogenesis was found to be activated in mid-
the evidence is otherwise. The cells within aberrant pro- stage lesions, prior to the appearance of full-blown tu-
liferative lesions initially lack angiogenic ability, curtail- mors. Similarly, angiogenesis can be discerned in pre-
ing their capability for expansion. In order to progress malignant lesions of the human cervix, breast, and skin
to a larger size, incipient neoplasias must develop angio- (melanocytes) (Hanahan and Folkman, 1996); we expect
genic ability (Bouck et al., 1996; Hanahan and Folkman, that induction of angiogenesis will prove to be an early
1996; Folkman, 1997). to midstage event in many human cancers. These obser-
Counterbalancing positive and negative signals en- vations, taken together with the effects of angiogenesis
courage or block angiogenesis. One class of these sig- inhibitors, indicate that neovascularization is a prerequi-
nals is conveyed by soluble factors and their receptors, site to the rapid clonal expansion associated with the
the latter displayed on the surface of endothelial cells; formation of macroscopic tumors.
integrins and adhesion molecules mediating cell–matrix Tumors appear to activate the angiogenic switch by
and cell–cell association also play critical roles. The changing the balance of angiogenesis inducers and
angiogenesis-initiating signals are exemplified by vas- countervailing inhibitors (Hanahan and Folkman, 1996).
cular endothelial growth factor (VEGF) and acidic and One common strategy for shifting the balance involves
basic fibroblast growth factors (FGF1/2). Each binds to altered gene transcription. Many tumors evidence in-
transmembrane tyrosine kinase receptors displayed by creased expression of VEGF and/or FGFs compared to
endothelial cells (Fedi et al., 1997; Veikkola and Alitalo, their normal tissue counterparts. In others, expression
1999). A prototypical angiogenesis inhibitor is throm- of endogenous inhibitors such as thrombospondin-1 or
bospondin-1, which binds to CD36, a transmembrane ␤-interferon is downregulated. Moreover, both transi-
receptor on endothelial cells coupled to intracellular Src- tions may occur, and indeed be linked, in some tumors
like tyrosine kinases (Bull et al., 1994). There are cur- (Singh et al., 1995; Volpert et al., 1997).
rently more than two dozen angiogenic inducer factors The mechanisms underlying shifts in the balances be-
known and a similar number of endogenous inhibitor tween angiogenic regulators remain incompletely un-
proteins. derstood. In one well-documented example, the inhibi-
Integrin signaling also contributes to this regulatory tor thrombospondin-1 has been found to positively
balance. Quiescent vessels express one class of inte- regulated by the p53 tumor suppressor protein in some
grins, whereas sprouting capillaries express another. cell types. Consequently, loss of p53 function, which
Interference with signaling from the latter class of inte- occurs in most human tumors, can cause thrombospon-
grins can inhibit angiogenesis (Varner and Cheresh, din-1 levels to fall, liberating endothelial cells from its
1996; Giancotti and Ruoslahti, 1999), underscoring the inhibitory effects (Dameron et al., 1994). The VEGF gene
important contribution of cell adhesion to the angiogenic is also under complex transcriptional control. For exam-
program (Hynes and Wagner, 1996). Extracellular prote- ple, activation of the ras oncogene or loss of the VHL
ases are physically and functionally connected with pro- tumor suppressor gene in certain cell types causes
angiogenic integrins, and both help dictate the invasive upregulation of VEGF expression (Rak et al., 1995; Max-
capability of angiogenic endothelial cells (Stetler-Ste- well et al., 1999).
venson, 1999). Another dimension of regulation is emerging in the
Experimental evidence for the importance of inducing form of proteases, which can control the bioavailability
and sustaining angiogenesis in tumors is both extensive of angiogenic activators and inhibitors. Thus, a variety
and compelling (Bouck et al., 1996; Hanahan and Folk- of proteases can release bFGF stored in the ECM
man, 1996; Folkman, 1997). The story begins almost 30 (Whitelock et al., 1996), whereas plasmin, a proangio-
years ago with Folkman and colleagues, who used in genic component of the clotting system, can cleave itself
vivo bioassays to demonstrate the necessity of angio- into an angiogenesis inhibitor form called angiostatin
genesis for explosive growth of tumor explants (re- (Gately et al., 1997). The coordinated expression of pro-
viewed in Folkman, 1997). Molecular proof of principle and antiangiogenic signaling molecules, and their mod-
came, for example, when anti-VEGF antibodies proved ulation by proteolysis, appear to reflect the complex
able to impair neovascularization and growth of subcu- homeostatic regulation of normal tissue angiogenesis
taneous tumors in mice (Kim et al., 1993), as did a domi- and of vascular integrity.
nant-interfering version of the VEGF receptor 2 (flk-1) As is already apparent, tumor angiogenesis offers a
(Millauer et al., 1994); both results have motivated the uniquely attractive therapeutic target, indeed one that
development of specific VEGF/VEGF-R inhibitors now is shared in common by most and perhaps all types of
in late stage clinical trials. human tumors. The next decade will produce a catalog
The essential role of angiogenesis is further supported of the angiogenic regulatory molecules expressed by
by the ability of an increasing catalog of antiangiogenic different types of tumors, and in many cases, by their
substances to impair the growth of tumor cells inocu- progenitor stages. Use of increasingly sophisticated
lated subcutaneously in mice (Folkman, 1997). Tumors mouse models will make it possible to assign specific
arising in cancer-prone transgenic mice are similarly roles to each of these regulators and to discern the
susceptible to angiogenic inhibitors (Bergers et al., molecular mechanisms that govern their production and
1999). activity. Already available evidence indicates that differ-
The ability to induce and sustain angiogenesis seems ent types of tumor cells use distinct molecular strategies
to be acquired in a discrete step (or steps) during tumor to activate the angiogenic switch. This raises the ques-
development, via an “angiogenic switch” from vascular tion of whether a single antiangiogenic therapeutic will
quiescence. When three transgenic mouse models were suffice to treat all tumor types, or whether an ensemble
analyzed throughout multistep tumorigenesis, in each of such therapeutics will need to be developed, each
Review
65

responding to a distinct program of angiogenesis that Changes in expression of CAMs in the immunoglobu-
has been developed by a specific class of human lin superfamily also appear to play critical roles in the
tumors. processes of invasion and metastasis (Johnson, 1991).
The clearest case involves N-CAM, which undergoes a
Acquired Capability: Tissue Invasion and Metastasis switch in expression from a highly adhesive isoform to
Sooner or later during the development of most types poorly adhesive (or even repulsive) forms in Wilms’ tu-
of human cancer, primary tumor masses spawn pioneer mor, neuroblastoma, and small cell lung cancer (John-
cells that move out, invade adjacent tissues, and thence son, 1991; Kaiser et al., 1996) and reduction in overall
travel to distant sites where they may succeed in found- expression level in invasive pancreatic and colorectal
ing new colonies. These distant settlements of tumor cancers (Fogar et al., 1997). Experiments in transgenic
cells—metastases—are the cause of 90% of human can- mice support a functional role for the normal adhesive
cer deaths (Sporn, 1996). The capability for invasion and form of N-CAM in suppressing metastasis (Perl et al.,
metastasis enables cancer cells to escape the primary 1999).
tumor mass and colonize new terrain in the body where, Changes in integrin expression are also evident in
at least initially, nutrients and space are not limiting. The invasive and metastatic cells. Invading and metastasiz-
newly formed metastases arise as amalgams of cancer ing cancer cells experience changing tissue microenvi-
cells and normal supporting cells conscripted from the ronments during their journeys, which can present novel
host tissue. Like the formation of the primary tumor matrix components. Accordingly, successful coloniza-
mass, successful invasion and metastasis depend upon tion of these new sites (both local and distant) demands
all of the other five acquired hallmark capabilities. But adaptation, which is achieved through shifts in the spec-
what additional cellular changes enable the acquisition trum of integrin ␣ or ␤ subunits displayed by the migrat-
of these final capabilities during tumorigenesis? ing cells. These novel permutations result in different
Invasion and metastasis are exceedingly complex integrin subtypes (of which there are greater than 22)
processes, and their genetic and biochemical determi- having distinct substrate preferences. Thus, carcinoma
nants remain incompletely understood. At the mecha- cells facilitate invasion by shifting their expression of
nistic level, they are closely allied processes, which justi- integrins from those that favor the ECM present in nor-
fies their association with one another as one general mal epithelium to other integrins (e.g., ␣3␤1 and ␣V␤3)
capability of cancer cells. Both utilize similar operational that preferentially bind the degraded stromal compo-
nents produced by extracellular proteases (Varner and
strategies, involving changes in the physical coupling
Cheresh, 1996; Lukashev and Werb, 1998). Forced ex-
of cells to their microenvironment and activation of ex-
pression of integrin subunits in cultured cells can induce
tracellular proteases.
or inhibit invasive and metastatic behavior, consistent
Several classes of proteins involved in the tethering
with a role of these receptors in acting as central deter-
of cells to their surroundings in a tissue are altered in
minants of these processes (Varner and Cheresh, 1996).
cells possessing invasive or metastatic capabilities. The
Attempts at explaining the cell biological effects of
affected proteins include cell–cell adhesion molecules
integrins in terms of a small number of mechanistic rules
(CAMs)—notably members of the immunoglobulin and
have been confounded by the large number of distinct
calcium-dependent cadherin families, both of which me-
integrin genes, by the even larger number of heterodi-
diate cell-to-cell interactions—and integrins, which link
meric receptors resulting from combinatorial expression
cells to extracellular matrix substrates. Notably, all of
of various ␣ and ␤ receptor subunits, and by the increas-
these “adherence” interactions convey regulatory sig-
ing evidence of complex signals emitted by the cyto-
nals to the cell (Aplin et al., 1998). The most widely
plasmic domains of these receptors (Aplin et al., 1998;
observed alteration in cell-to-environment interactions Giancotti and Ruoslahti, 1999). Still, there is little doubt
in cancer involves E-cadherin, a homotypic cell-to-cell that these receptors play central roles in the capability
interaction molecule ubiquitously expressed on epithe- for tissue invasion and metastasis.
lial cells. Coupling between adjacent cells by E-cadherin The second general parameter of the invasive and
bridges results in the transmission of antigrowth and metastatic capability involves extracellular proteases
other signals via cytoplasmic contacts with ␤-catenin (Coussens and Werb, 1996; Chambers and Matrisian,
to intracellular signaling circuits that include the Lef/ 1997). Protease genes are upregulated, protease inhibi-
Tcf transcription factor (Christofori and Semb, 1999). tor genes are downregulated, and inactive zymogen
E-cadherin function is apparently lost in a majority of forms of proteases are converted into active enzymes.
epithelial cancers, by mechanisms that include muta- Matrix-degrading proteases are characteristically asso-
tional inactivation of the E-cadherin or ␤-catenin genes, ciated with the cell surface, by synthesis with a trans-
transcriptional repression, or proteolysis of the extracel- membrane domain, binding to specific protease re-
lular cadherin domain (Christofori and Semb, 1999). ceptors, or association with integrins (Werb, 1997;
Forced expression of E-cadherin in cultured cancer cells Stetler-Stevenson, 1999). One imagines that docking of
and in a transgenic mouse model of carcinogenesis im- active proteases on the cell surface can facilitate inva-
pairs invasive and metastatic phenotypes, whereas in- sion by cancer cells into nearby stroma, across blood
terference with E-cadherin function enhances both vessel walls, and through normal epithelial cell layers.
capabilities (Christofori and Semb, 1999). Thus, E-cad- That notion notwithstanding, it is difficult to unambigu-
herin serves as a widely acting suppressor of invasion ously ascribe the functions of particular proteases solely
and metastasis by epithelial cancers, and its functional to this capability, given their evident roles in other
elimination represents a key step in the acquisition of hallmark capabilities, including angiogenesis (Stetler-
this capability. Stevenson, 1999) and growth signaling (Werb, 1997;
Cell
66

Figure 4. Parallel Pathways of Tumorigen-


esis
While we believe that virtually all cancers
must acquire the same six hallmark capabili-
ties (A), their means of doing so will vary sig-
nificantly, both mechanistically (see text) and
chronologically (B). Thus, the order in which
these capabilities are acquired seems likely
be quite variable across the spectrum of can-
cer types and subtypes. Moreover, in some
tumors, a particular genetic lesion may confer
several capabilities simultaneously, decreas-
ing the number of distinct mutational steps
required to complete tumorigenesis. Thus,
loss of function of the p53 tumor suppressor
can facilitate both angiogenesis and resis-
tance to apoptosis (e.g., in the five-step path-
way shown), as well as enabling the charac-
teristic of genomic instability. In other tumors,
a capability may only be acquired through the
collaboration of two or more distinct genetic
changes, thereby increasing the total number
necessary for completion of tumor progres-
sion. Thus, in the eight-step pathway shown,
invasion/metastasis and resistance to apo-
ptosis are each acquired in two steps.

Bergers and Coussens, 2000), which in turn contribute The available evidence suggests that most are acquired,
directly or indirectly to the invasive/metastatic capa- directly or indirectly, through changes in the genomes
bility. of cancer cells. But mutation of specific genes is an
A further dimension of complexity derives from the inefficient process, reflecting the unceasing, fastidious
multiple cell types involved in protease expression and maintenance of genomic integrity by a complex array
display. In many types of carcinomas, matrix-degrading of DNA monitoring and repair enzymes. These genome
proteases are produced not by the epithelial cancer cells maintenance teams strive to ensure that DNA sequence
but rather by conscripted stromal and inflammatory cells information remains pristine. Karyotypic order is guaran-
(Werb, 1997); once released by these cells, they may be teed by yet other watchmen, manning so-called check-
wielded by the carcinoma cells. For example, certain points, that operate at critical times in the cell’s life,
cancer cells induce urokinase (uPA) expression in cocul- notably mitosis. Together, these systems ensure that
tured stromal cells, which then binds to the urokinase mutations are rare events, indeed so rare that the multi-
receptor (uPAR) expressed on the cancer cells (Johnsen ple mutations known to be present in tumor cell ge-
et al., 1998). nomes are highly unlikely to occur within a human life
The activation of extracellular proteases and the al- span.
tered binding specificities of cadherins, CAMs, and inte- Yet cancers do appear at substantial frequency in
grins are clearly central to the acquisition of invasive- the human population, causing some to argue that the
ness and metastatic ability. But the regulatory circuits genomes of tumor cells must acquire increased mutabil-
and molecular mechanisms that govern these shifts re- ity in order for the process of tumor progression to reach
main elusive and, at present, seem to differ from one completion in several decades time (Loeb, 1991). Mal-
tissue environment to another. The acquired capability function of specific components of these genomic
for invasion and metastasis represents the last great “caretaker” systems has been invoked to explain this
frontier for exploratory cancer research. We envision increased mutability (Lengauer et al., 1998). The most
that evolving analytic techniques will soon make it possi- prominent member of these systems is the p53 tumor
ble to construct comprehensive profiles of the expres- suppressor protein, which, in response to DNA damage,
sion and functional activities of proteases, integrins, and elicits either cell cycle arrest to allow DNA repair to take
CAMs in a wide variety of cancer types, both before and place or apoptosis if the damage is excessive. Indeed, it
after they acquire invasive and metastatic abilities. The is now clear that the functioning of the p53 DNA damage
challenge will then be to apply the new molecular in- signaling pathway is lost in most, if not all, human can-
sights about tissue invasiveness and metastasis to the cers (Levine, 1997). Moreover, a growing number of
development of effective therapeutic strategies. other genes involved in sensing and repairing DNA dam-
age, or in assuring correct chromosomal segregation
An Enabling Characteristic: Genome Instability during mitosis, is found to be lost in different cancers,
The acquisition of the enumerated six capabilities during labeling these caretakers as tumor suppressors (Len-
the course of tumor progression creates a dilemma. gauer et al., 1998). Their loss of function is envisioned
Review
67

to allow genome instability and variability and the gener- the signals exchanged between the various cell types
ation of consequently mutant cells with selective advan- existing symbiotically within a tumor mass and knowing
tages. Interestingly, recent evidence suggests that apo- their effects on the integrated circuits of each of those
ptosis may also be a vehicle of genomic instability, in cell types.
that DNA within apoptotic cell bodies can be incorpo- Our ability to analyze individual human cancers at
rated into neighboring cells following phagoctytosis the genetic and biochemical levels will also undergo a
(Holmgren et al., 1999), in principle genetically diversify- dramatic change. At present, description of a recently
ing any of the constituent cell types of a tumor. We place diagnosed tumor in terms of its underlying genetic le-
this acquired characteristic of genomic instability apart sions remains a distant prospect. Nonetheless, we look
from the six acquired capabilities associated with tumor ahead 10 or 20 years to the time when the diagnosis of
cell phenotype and tumor physiology: it represents the all the somatically acquired lesions present in a tumor
means that enables evolving populations of premalig- cell genome will become a routine procedure. By then,
nant cells to reach these six biological endpoints. genome-wide gene expression profiles of tumor cells
will also be routine. With all this information in hand, it
Alternative Pathways to Cancer will become possible to test definitively our proposition
The paths that cells take on their way to becoming malig- that the development of all types of human tumor cells
nant are highly variable. Within a given cancer type, is governed by a common set of rules such as those
mutation of particular target genes such as ras or p53 implied by the six acquired capabilities enumerated
may be found in only a subset of otherwise histologically here.
identical tumors. Further, mutations in certain onco- We anticipate far deeper insight into the roles played
genes and tumor suppressor genes can occur early in by inherited alleles in cancer susceptibility and patho-
some tumor progression pathways and late in others. As genesis. At present, our understanding of the interplay
a consequence, the acquisition of biological capabilities at the cellular level between inherited cancer modifier
such as resistance to apoptosis, sustained angiogen- genes with oncogenes and tumor suppressor genes that
esis, and unlimited replicative potential can appear at are altered somatically is rudimentary; modifiers can in
different times during these various progressions. Ac- principle act in any of the constituent cell types of a
cordingly, the particular sequence in which capabilities tumor, or elsewhere in the body, whereas the classical
are acquired can vary widely, both among tumors of the cancer genes largely act in the cancer cells themselves.
same type and certainly between tumors of different These gaps will be bridged in part by new informatics
types (Figure 4). Furthermore, in certain tumors, a spe- technologies, enabling us to process and interpret the
cific genetic event may, on its own, contribute only par- inundation of genetic information that will soon flow from
tially to the acquisition of a single capability, while in automated sequencing instruments. New technologies
others, this event may aid in the simultaneous acquisi- will also aid us in rationalizing the complex constella-
tion of several distinct capabilities. Nonetheless, we be- tions of interacting alleles in terms of a systematics of
lieve that independent of how the steps in these genetic cancer formation of the type that we propose here.
pathways are arranged, the biological endpoints that The metaphors used to conceptualize cancer cell
are ultimately reached—the hallmark capabilities of can- function will also shift dramatically. For decades now,
cer—will prove to be shared in common by all types of we have been able to predict with precision the behavior
tumors. of an electronic integrated circuit in terms of its constit-
uent parts—its interconnecting components, each re-
Synthesis sponsible for acquiring, processing, and emitting signals
Cancer cells propagated in culture and dissected into according to a precisely defined set of rules. Two de-
their molecular components have yielded much of the cades from now, having fully charted the wiring dia-
wealth of information that we currently possess about grams of every cellular signaling pathway, it will be pos-
the molecular processes underlying cancer develop- sible to lay out the complete “integrated circuit of the
ment. Yet by simplifying the nature of cancer—por- cell” upon its current outline (Figure 2). We will then be
traying it as a cell-autonomous process intrinsic to the able to apply the tools of mathematical modeling to
cancer cell—these experimental models have turned explain how specific genetic lesions serve to reprogram
their back on a central biological reality of tumor forma- this integrated circuit in each of the constituent cell
tion in vivo: cancer development depends upon changes types so as to manifest cancer.
in the heterotypic interactions between incipient tumor With holistic clarity of mechanism, cancer prognosis
cells and their normal neighbors. Moreover, once formed, and treatment will become a rational science, unrecog-
virtually all types of human tumors, including their meta- nizable by current practitioners. It will be possible to
static outgrowths, continue to harbor complex mixtures understand with precision how and why treatment regi-
of several cell types that collaborate to create malignant mens and specific antitumor drugs succeed or fail. We
growth (Figure 3). This reconceptualization of cancer envision anticancer drugs targeted to each of the hall-
cell biology has begun to drive profound changes in mark capabilities of cancer; some, used in appropriate
how we study this disease experimentally. Continuing combinations and in concert with sophisticated technol-
elucidation of cancer pathogenesis will depend increas- ogies to detect and identify all stages of disease pro-
ingly upon heterotypic organ culture systems in vitro gression, will be able to prevent incipient cancers from
and evermore refined mouse models in vivo. Looking developing, while others will cure preexisting cancers,
ahead into the future, these systems will help us chart elusive goals at present. One day, we imagine that can-
comprehensive maps of growth signaling networks in cer biology and treatment—at present, a patchwork quilt
cancer, an endeavor that will depend on defining all of of cell biology, genetics, histopathology, biochemistry,
Cell
68

immunology, and pharmacology—will become a sci- molecule E-cadherin as a tumour-suppressor gene. Trends Bio-
ence with a conceptual structure and logical coherence chem. Sci. 24, 73–76.
that rivals that of chemistry or physics. Christofori, G., Naik, P., and Hanahan, D. (1994). A second signal
supplied by insulin-like growth factor II in oncogene-induced tumori-
genesis. Nature 369, 414–418.
Acknowledgments Cordon-Cardo, C., and Prives, C. (1999). At the crossroads of inflam-
mation and tumorigenesis. J. Exp. Med. 190, 1367–1370.
We wish to thank Terry Schoop of Biomed Arts Associates, San
Counter, C.M., Avilion, A.A., LeFeuvre, C.E., Stewart, N.G., Greider,
Francisco, for preparation of the figures, Cori Bargmann and Zena
C.W., Harley, C.B., and Bacchetti, S. (1992). Telomere shortening
Werb for insightful comments on the manuscript, and Normita San-
associated with chromosome instability is arrested in immortal cells
tore for editorial assistance. In addition, we are indebted to Joe
which express telomerase activity. EMBO J. 11, 1921–1929.
Harford and Richard Klausner, who allowed us to adapt and expand
Counter, C.M., Hahn, W.C., Wei, W., Dickinson Caddle, S., Beijers-
their depiction of the cell signaling network, and we appreciate
bergen, R.L., Lansdorp, P.M., Sedivy, J.M., and Weinberg, R.A.
suggestions on signaling pathways from Randy Watnick, Brian Elen-
(1998). Dissociation between telomerase activity, telomere mainte-
bas, Bill Lundberg, Dave Morgan, and Henry Bourne. R. A. W. is
nance and cellular immortalization. Proc. Natl. Acad. Sci. USA 95,
a Ludwig Foundation and American Cancer Society Professor of
14723–14728.
Biology. His work has been supported by the Department of the
Army and the National Institutes of Health. D. H. acknowledges Coussens, L.M., and Werb, Z. (1996). Matrix metalloproteinases and
the support and encouragement of the National Cancer Institute. the development of cancer. Chem. Biol. 3, 895–904.
Editorial policy has rendered the citations illustrative but not com- Coussens, L.M., Raymond, W.W., Bergers, G., Laig-Webster, M.,
prehensive. Behrendtsen, O., Werb, Z., Caughey, G.H., and Hanahan, D. (1999).
Inflammatory mast cells up-regulate angiogenesis during squamous
epithelial carcinogenesis. Genes Dev. 13, 1382–1397.
References
Dameron, K.M., Volpert, O.V., Tainsky, M.A., and Bouck, N. (1994).
Control of angiogenesis in fibroblasts by p53 regulation of throm-
Aplin, A.E., Howe, A., Alahari, S.K., and Juliano, R.L. (1998). Signal
bospondin-1. Science 265, 1582–1584.
transduction and signal modulation by cell adhesion receptors: the
role of integrins, cadherins, immunoglobulin-cell adhesion mole- Datto, M.B., Hu, P.P., Kowalik, T.F., Yingling, J., and Wang, X.F.
cules, and selectins. Pharmacol. Rev. 50, 197–263. (1997). The viral oncoprotein E1A blocks transforming growth factor
␤-mediated induction of p21/WAF1/Cip1 and p15/INK4B Mol. Cell.
Ashkenazi, A., and Dixit, V.M. (1999). Apoptosis control by death
Biol. 17, 2030–2037.
and decoy receptors. Curr. Opin. Cell Biol. 11, 255–260.
DiFiore, P.P., Pierce, J.H., Kraus, M.H., Segatto, O., King, C.R., and
Bergers, G., and Coussens, L.M. (2000). Extrinsic regulators of epi- Aaronson, S.A. (1987). erbB-2 is a potent oncogene when overex-
thelial tumor progression: metalloproteinases. Curr. Opin. Genet. pressed in NIH/3T3 cells. Science 237, 178–182.
Dev., in press.
Downward, J. (1998). Mechanisms and consequences of activation
Bergers, G., Hanahan, D., and Coussens, L.M. (1998). Angiogenesis of protein kinase B/Akt. Curr. Opin. Cell Biol. 10, 262–267.
and apoptosis are cellular parameters of neoplastic progression in
Dyson, N., Howley, P.M., Munger, K., and Harlow, E. (1989). The
transgenic mouse models of tumorigenesis. Int. J. Dev. Biol. 42, human papillomavirus-16 E7 oncoprotein is able to bind to the reti-
995–1002. noblastoma gene product. Science 243, 934–937.
Bergers, G., Javaherian, K., Lo, K.-M., Folkman, J., and Hanahan, Evan, G., and Littlewood, T. (1998). A matter of life and cell death.
D. (1999). Effects of angiogenesis inhibitors on multistage carcino- Science 281, 1317–1322.
genesis in mice. Science 284, 808–812.
Fedi, P., Tronick, S.R., and Aaronson, S.A. (1997). Growth factors.
Bishop, J.M., and Weinberg, R.A., eds. (1996). Molecular Oncology In Cancer Medicine, J.F. Holland, R.C. Bast, D.L. Morton, E. Frei,
(New York: Scientific American, Inc.). D.W. Kufe, and R.R. Weichselbaum, eds. (Baltimore, MD: Williams
Bodnar, A.G., Ouellete, M., Frolkis, M., Holt, S.E., Chiu, C., Morin, and Wilkins), pp. 41–64.
G.B., Harley, C.B., Shay, J.W., Lichtsteiner, S., and Wright, W.E. Fogar, P., Basso, D., Pasquali, C., De Paoli, C., Sperti, C., Roveroni,
(1998). Extension of life-span by introduction of telomerase into G., Pedrazzoli, G., and Plebani, M. (1997). Neural cell adhesion mole-
normal human cells. Science 279, 349–352. cule (N-CAM) in gastrointestinal neoplasias. Anticancer Res. 17,
Bouck, N., Stellmach, V., and Hsu, S.C. (1996). How tumors become 1227–1230.
angiogenic. Adv. Cancer Res. 69, 135–174. Foley, K.P., and Eisenman, R.N. (1999). Two MAD tails: what the
Bryan, T.M., and Cech, T.R. (1999). Telomerase and the maintenance recent knockouts of Mad1 and Mx1 tell us about the MYC/MAX/
of chromosome ends. Curr. Opin. Cell Biol. 11, 318–324. MAD network. Biochim. Biophys. Acta 1423, M37–47.
Bryan, T.M., Englezou, A., Gupta, J., Bacchetti, S., and Reddel, Folkman, J. (1997). Tumor angiogenesis. In Cancer Medicine, J.F.
R.R. (1995). Telomere elongation in immortal human cells without Holland, R.C. Bast, D.L. Morton, E. Frei, D.W. Kufe, and R.R. Weich-
selbaum, eds. (Baltimore, MD: Williams and Wilkins), pp. 181–204.
detectable telomerase activity. EMBO J. 14, 4240–4248.
Foulds, L. (1954). The Experimental Study of Tumor Progression.
Bull, H.A., Brickell, P.M., and Dowd, P.M. (1994). Src-related protein
Volumes I–III (London: Academic Press).
tyrosine kinases are physically associated with the surface antigen
CD36 in human dermal microvascular endothelial cells. FEBS Lett. Fynan, T.M., and Reiss, M. (1993). Resistance to inhibition of cell
351, 41–44. growth by transforming growth factor-␤ and its role in oncogenesis.
Crit. Rev. Oncog. 4, 493–540.
Butt, A.J., Firth, S.M., and Baxter, R.C. (1999). The IGF axis and
programmed cell death. Immunol. Cell Biol. 77, 256–262. Gately, S., Twardowski, P., Stack, M.S., Cundiff, D.L., Grella, D.,
Castellino, F.J., Enghild, J., Kwaan, H.C., Lee, F., Kramer, R.A., et
Cantley, L.C., and Neel, B.G. (1999). New insights into tumor sup- al. (1997). The mechanism of cancer-mediated conversion of plas-
pression: PTEN suppresses tumor formation by restraining the minogen to the angiogenesis inhibitor angiostatin. Proc. Natl. Acad.
phosphoinositide 3-kinase/AKT pathway. Proc. Natl. Acad. Sci. USA Sci. USA 94, 10868–10872.
96, 4240–4245.
Giancotti, F.G., and Ruoslahti, E. (1999). Integrin signaling. Science
Chambers, A.F., and Matrisian, L.M. (1997). Changing views of the 285, 1028–1032.
role of matrix metalloproteinases in metastasis. J. Natl. Cancer Inst.
Green, D.R., and Reed, J.C. (1998). Mitochondria and apoptosis.
89, 1260–1270. Science 281, 1309–1312.
Chin, L., Pomerantz, J., and DePinho, R.A. (1998). The INK4a/ARF Greenberg, R.A., Chin, L., Femino, A., Lee, K.H., Gottlieb, G.J.,
tumor suppressor: one gene—two products—two pathways. Trends Singer, R.H., Greider, C.W., and DePinho, R.A. (1999). Short dysfunc-
Biochem. Sci. 23, 291–296. tional telomeres impair tumorigenesis in the INK4a⌬2/3 cancer-prone
Christofori, G., and Semb, H. (1999). The role of the cell-adhesion mouse. Cell 97, 515–525.
Review
69

Hahn, W.C., Counter, C.M., Lundberg, A.S., Beijersbgern, R.L., Markowitz, S., Wang, J., Meyeroff, L., Parsons, R., Sun, L., Lutter-
Brooks, M.W., and Weinberg, R.A. (1999). Creation of human tumor baugh, J., Fan, R., Zborowska, E., Kinzler, K., Vogelstein, B., et al.
cells with defined genetic elements. Nature 400, 464–468. (1995). Inactivation of the type II TGF-␤ receptor in colon cancer
Halvorsen, T.L., Leibowitz, G., and Levine, F. (1999). Telomerase cells with microsatellite instability. Science 268, 1336–1338.
activity is sufficient to allow transformed cells to escape from crisis. Maxwell, P.H., Wiesener, M.S., Chang, G.-W., Clifford, S.C., Vaux,
Mol. Cell. Biol. 19, 1864–1870. E.C., Cockman, M.E., Wykoff, C.C., Pugh, C.W., Maher, E.R., and
Hanahan, D., and Folkman, J. (1996). Patterns and emerging mecha- Ratcliffe, P.J. (1999). The tumour suppressor protein VHL targets
nisms of the angiogenic switch during tumorigenesis. Cell 86, hypoxia-inducible factors for oxygen-dependent proteolysis. Nature
353–364. 399, 271–275.
Hannon, G.J., and Beach, D. (1994). P15INK4B is a potential effector McDonnell, T.J., and Korsmeyer, S.J. (1991). Progression from
of TGF-beta-induced cell cycle arrest. Nature 371, 257–261. lymphoid hyperplasia to high-grade malignant lymphoma in mice
transgenic for the t(14;18). Nature 349, 254–256.
Harris, C.C. (1996). p53 tumor suppressor gene: from the basic
research laboratory to the clinic—an abridged historical perspec- Medema, R.H., and Bos, J.L. (1993). The role of p21-ras in receptor
tive. Carcinogenesis 17, 1187–1198. tyrosine kinase signaling. Crit. Rev. Oncog. 4, 615–661.
Hayflick, L. (1997). Mortality and immortality at the cellular level. A Millauer, B., Shawver, L.K., Plate, K.H., Risau, W., and Ullrich, A.
review. Biochemistry 62, 1180–1190. (1994). Glioblastoma growth inhibited in vivo by a dominant-negative
Flk-1 mutant. Nature 367, 576–579.
Holmgren, L., Szeles, A., Rajnavolgyi, E., Folkman, J., Klein, G.,
Ernberg, I., and Falk, K.I. (1999). Horizontal transfer of DNA by the Moses, H.L., Yang, E.Y., and Pietenpol, J.A. (1990). TGF-␤ stimula-
uptake of apoptotic bodies. Blood 93, 3956–3963. tion and inhibition of cell proliferation: new mechanistic insights.
Cell 63, 245–247.
Hudson, J.D., Shoaibi, M.A., Maestro, R., Carnero, A., Hannon, G.J.,
and Beach, D.H. (1999). A proinflammatory cytokine inhibits p53 Nowell, P.C. (1976). The clonal evolution of tumor cell populations.
tumor suppressor activity. J. Exp. Med. 190, 1375–1382. Science 194, 23–28.
Hueber, A.O., Zornig, M., Lyon, D., Suda, T., Nagata, S., and Evan, Olumi, A.F., Grossfeld, G.D., Hayward, S.W., Carroll, P.R., Tlsty, T.D.,
G.I. (1997). Requirement for the CD95 receptor-ligand pathway in and Cunha, G.R. (1999). Carcinoma-associated fibroblasts direct
c-Myc-induced apoptosis. Science 278, 1305–1309. tumor progression of initiated human prostatic epithelium. Cancer
Res. 59, 5002–5011.
Hunter, T. (1997). Oncoprotein networks. Cell 88, 333–346.
Perl, A.-K., Dahl, U., Wilgenbus, P., Cremer, H., Semb, H., and
Hynes, R.O., and Wagner, D.D. (1996). Genetic manipulation of vas-
Christofori, G. (1999). Reduced expresion of neural cell adhesion
cular adhesion molecules in mice. J. Clin. Invest. 98, 2193–2195.
molecule induces metastatic dissemination of pancreatic ␤ tumor
Ishizaki, Y., Cheng, L., Mudge, A.W., and Raff, M.C. (1995). Pro- cells. Nat. Med. 5, 286–291.
grammed cell death by default in embryonic cells, fibroblasts, and
Pitti, R.M., Marsters, S.A., Lawrence, D.A., Roy, M., Kischkel, F.C.,
cancer cells. Mol. Biol. Cell 6, 1443–1458.
Dowd, P., Huang, A., Donahue, C.J., Sherwood, S.W., Baldwin, D.T.,
Johnsen, M., Lund, L.R., Romer, J., Almholt, K., and Dano K. (1998). et al. (1998). Genomic amplification of a decoy receptor for Fas
Cancer invasion and tissue remodeling: common themes in proteo- ligand in lung and colon cancer. Nature 396, 699–703.
lytic matrix degradation. Curr. Opin. Cell Biol. 10, 667–671.
Rak, J., Filmus, J., Finkenzeller, G., Grugel, S., Marme, D., and Ker-
Johnson, J.P. (1991). Cell adhesion molecules of the immunoglobu- bel, R.S. (1995). Oncogenes as inducers of tumor angiogenesis.
lin supergene family and their role in malignant transformation and Cancer Metastasis Rev. 14, 263–277.
progression to metastatic disease. Cancer Metastasis Rev. 10,
Renan, M.J. (1993). How many mutations are required for tumorigen-
11–22.
esis? Implications from human cancer data. Mol. Carcinogenesis 7,
Kahn, P., Frykberg, L., Brady, C., Stanley, I., Beug, H., Vennström, 139–146.
B., and Graf, T. (1986). v-erbA cooperates with sarcoma oncogenes
Rommel, C., and Hafen, E. (1998). Ras—a versatile cellular switch.
in leukemic cell transformation. Cell 45, 349–356.
Curr. Opin. Genet. Dev. 8, 412–418.
Kaiser, U., Auerbach, B., and Oldenburg, M. (1996). The neural cell
Schutte, M., Hruban, R., Hedrick, L., Cho, K., Nadasdy, G., Weinstein,
adhesion molecule NCAM in multiple myeloma. Leuk. Lymphoma
C., Bova, G., Isaacs, W., Cairns, P., Nawroz, H., et al. (1996). DPC4
20, 389–395.
gene in various tumor types. Cancer Res. 56, 2527–2530.
Kerr, J.F., Wyllie, A.H., and Currie, A.R. (1972). Apoptosis: a basic
Serrano, M., Lin, A.W., McCurrach, M.E., Beach, D., and Lowe, S.W.
biological phenomenon with wide-ranging implications in tissue ki-
(1997). Oncogeneic ras provokes premature cell senescence associ-
netics. Br. J. Cancer 26, 239–257.
ated with accumulation of p53 and p16INK4A. Cell 88, 593–602.
Kim, K.J., Li, B., Winer, J., Armanini, M., Gillett, N., Philipps, H.S., and
Shibata, M.A., Maroulakou, I.G., Jorcyk, C.L., Gold, L.G., Ward, J.M.,
Ferrara, N. (1993). Inhibition of vascular endothelial growth factor-
and Green, J.E. (1996). p53-independent apoptosis during mammary
induced angiogenesis suppresses tumour growth in vivo. Nature
tumor progression in C3(1)/SV40 large T antigen transgenic mice:
362, 841–844.
suppression of apoptosis during the transition from preneoplasia
Kinzler, K.W., and Vogelstein, B. (1996). Lessons from hereditary to carcinoma. Cancer Res. 56, 2998–3003.
colorectal cancer. Cell 87, 159–170.
Shay, J.W., and Bacchetti, S. (1997). A survey of telomerase activity
Kinzler, K.W., and Vogelstein, B. (1998). Landscaping the cancer in human cancer. Eur. J. Cancer 33, 787–791.
terrain. Science 280, 1036–1037.
Singh, R.K., Gutman, M., Bucana, C.D., Sanchez, R., Llansa, N.,
Korsmeyer, S.J. (1992). Chromosomal translocations in lymphoid and Fidler, I.J. (1995). Interferons alpha and beta down-regulate the
malignancies reveal novel proto-oncogenes. Annu. Rev. Immunol. expression of basic fibroblast growth factor in human carcinomas.
10, 785–807. Proc. Natl. Acad. Sci. USA 92, 4562–4566.
Lengauer, C., Kinzler, K.W., and Vogelstein, B. (1998). Genetic insta- Skobe, M., and Fusenig, N.E. (1998). Tumorigenic conversion of
bilities in human cancers. Nature 396, 643–649. immortal human keratinocytes through stromal cell activation. Proc.
Levine, A.J. (1997). p53, the cellular gatekeeper for growth and divi- Natl. Acad. Sci. USA 95, 1050–1055.
sion. Cell 88, 323–331. Slamon, D.J., Clark, G.M., Wong, S.G., Levin, W.J., Ullrich, A., and
Loeb, L.A. (1991). Mutator phenotype may be required for multistep McGuire, W.L. (1987). Human breast cancer: correlation of relapse
carcinogenesis. Cancer Res. 51, 3073–3079. and survival with amplification of the HER-2/neu oncogene. Science
Lotem, J., and Sachs, L. (1996). Control of apoptosis in hematopoie- 235, 177–182.
sis and leukemia by cytokines, tumor suppressor and oncogenes. Sporn, M.B. (1996). The war on cancer. Lancet 347, 1377–1381.
Leukemia 10, 925–931. Stetler-Stevenson, W.G. (1999). Matrix metalloproteinases in angio-
Lukashev, M.E., and Werb, Z. (1998). ECM signaling: orchestrating genesis: a moving target for therapeutic intervention. J. Clin. Invest.
cell behaviour and misbehaviour. Trends Cell Biol. 8, 437–441. 103, 1237–1241.
Cell
70

Strasser, A., Harris, A.W., Bath, M.L., and Cory, S. (1990). Novel
primitive lymphoid tumours induced in transgenic mice by coopera-
tion between myc and bcl-2. Nature 348, 331–333.
Symonds, H., Krall, L., Remington, L., Saenz-Robles, M., Lowe, S.,
Jacks, T., and Van Dyke, T. (1994). p53-dependent apoptosis sup-
presses tumor growth and progression in vivo. Cell 78, 703–711.
Thornberry, N.A., and Lazebnik, Y. (1998). Caspases: enemies within.
Science 281, 1312–1316.
Varner, J.A., and Cheresh, D.A. (1996). Integrins and cancer. Curr.
Opin. Cell Biol. 8, 724–730.
Vaux, D.L., Cory, S., and Adams, T.M. (1988). Bcl-2 promotes the
survival of hematopoietic cells and cooperates with c-myc to immor-
talize pre-B cells. Nature 335, 440–442.
Vaziri, H., and Benchimol, S. (1998). Reconstitution of telomerase
activity in normal human cells leads to elongation of telomeres and
extended replicative life span. Curr. Biol. 8, 279–282.
Veikkola, T., and Alitalo, K. (1999). VEGFs, receptors and angiogen-
esis. Semin. Cancer Biol. 9, 211–220.
Volpert, O.V., Dameron, K.M., and Bouck, N. (1997). Sequential de-
velopment of an angiogenic phenotype by human fibroblasts pro-
gressing to tumorigenicity. Oncogene 14, 1495–1502.
Weinberg, R.A. (1995). The retinoblastoma protein and cell cycle
control. Cell 81, 323–330.
Werb, Z. (1997). ECM and cell surface proteolysis: regulating cellular
ecology. Cell 91, 439–442.
Whitelock, J.M., Murdoch, A.D., Iozzo, R.V., and Underwood, P.A.
(1996). The degradation of human endothelial cell-derived perlecan
and release of bound basic fibroblast growth factor by stromelysin,
collagenase, plasmin, and heparanases. J. Biol. Chem. 271, 10079–
10086.
Wright, W.E., Pereira-Smith, O.M., and Shay, J.W. (1989). Reversible
cellular senescence: implications for immortalization of normal hu-
man diploid fibroblasts. Mol. Cell. Biol. 9, 3088–3092.
Wyllie, A.H., Kerr, J.F., and Currie, A.R. (1980). Cell death: the signifi-
cance of apoptosis. Int. Rev. Cytol. 68, 251–306.
Yarden, Y., and Ullrich, A. (1988). EGF and erbB2 receptor overex-
pression in human tumors. Growth factor recepor tyrosine kinases.
Annu. Rev. Biochem. 57, 443–478.
Zhu, J., Wang, H., Bishop, J.M., and Blackburn, E.H. (1999). Telo-
merase extends the lifespan of virus-transformed human cells with-
out net telomere lengthening. Proc. Natl. Acad. Sci. USA 96, 3723–
3728.
Zuo, L., Weger, J., Yang, Q., Goldstein, A.M., Tucker, M.A., Walker,
G.J., Hayward, N., and Dracopoli, N.C. (1996). Germline mutations
in the p16INK4A binding domain of CDK4 in familial melanoma. Nat.
Genet. 12, 97–99.
Leading Edge
Review

Hallmarks of Cancer: The Next Generation


Douglas Hanahan1,2,* and Robert A. Weinberg3,*
1The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne CH-1015, Switzerland
2The Department of Biochemistry & Biophysics, UCSF, San Francisco, CA 94158, USA
3Whitehead Institute for Biomedical Research, Ludwig/MIT Center for Molecular Oncology, and MIT Department of Biology, Cambridge,

MA 02142, USA
*Correspondence: dh@epfl.ch (D.H.), weinberg@wi.mit.edu (R.A.W.)
DOI 10.1016/j.cell.2011.02.013

The hallmarks of cancer comprise six biological capabilities acquired during the multistep develop-
ment of human tumors. The hallmarks constitute an organizing principle for rationalizing the
complexities of neoplastic disease. They include sustaining proliferative signaling, evading growth
suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and acti-
vating invasion and metastasis. Underlying these hallmarks are genome instability, which generates
the genetic diversity that expedites their acquisition, and inflammation, which fosters multiple hall-
mark functions. Conceptual progress in the last decade has added two emerging hallmarks of
potential generality to this list—reprogramming of energy metabolism and evading immune
destruction. In addition to cancer cells, tumors exhibit another dimension of complexity: they
contain a repertoire of recruited, ostensibly normal cells that contribute to the acquisition of hall-
mark traits by creating the ‘‘tumor microenvironment.’’ Recognition of the widespread applicability
of these concepts will increasingly affect the development of new means to treat human cancer.

INTRODUCTION vated by these developments, we now revisit the original hall-


marks, consider new ones that might be included in this roster,
We have proposed that six hallmarks of cancer together consti- and expand upon the functional roles and contributions made
tute an organizing principle that provides a logical framework for by recruited stromal cells to tumor biology.
understanding the remarkable diversity of neoplastic diseases
(Hanahan and Weinberg, 2000). Implicit in our discussion was HALLMARK CAPABILITIES—CONCEPTUAL PROGRESS
the notion that as normal cells evolve progressively to
a neoplastic state, they acquire a succession of these hallmark The six hallmarks of cancer—distinctive and complementary
capabilities, and that the multistep process of human tumor capabilities that enable tumor growth and metastatic dissemina-
pathogenesis could be rationalized by the need of incipient tion—continue to provide a solid foundation for understanding
cancer cells to acquire the traits that enable them to become the biology of cancer (Figure 1; see the Supplemental Informa-
tumorigenic and ultimately malignant. tion for downloadable versions of the figures for presentations).
We noted as an ancillary proposition that tumors are more than In the first section of this Review, we summarize the essence
insular masses of proliferating cancer cells. Instead, they are of each hallmark as described in the original presentation in
complex tissues composed of multiple distinct cell types that 2000, followed by selected illustrations (demarcated by sub-
participate in heterotypic interactions with one another. We de- headings in italics) of the conceptual progress made over the
picted the recruited normal cells, which form tumor-associated past decade in understanding their mechanistic underpinnings.
stroma, as active participants in tumorigenesis rather than In subsequent sections we address new developments that
passive bystanders; as such, these stromal cells contribute to broaden the scope of the conceptualization, describing in turn
the development and expression of certain hallmark capabilities. two enabling characteristics crucial to the acquisition of the six
During the ensuing decade this notion has been solidified and hallmark capabilities, two new emerging hallmark capabilities,
extended, revealing that the biology of tumors can no longer the constitution and signaling interactions of the tumor microen-
be understood simply by enumerating the traits of the cancer vironment crucial to cancer phenotypes, and we finally discuss
cells but instead must encompass the contributions of the the new frontier of therapeutic application of these concepts.
‘‘tumor microenvironment’’ to tumorigenesis.
In the course of remarkable progress in cancer research Sustaining Proliferative Signaling
subsequent to this publication, new observations have served Arguably the most fundamental trait of cancer cells involves their
both to clarify and to modify the original formulation of the hall- ability to sustain chronic proliferation. Normal tissues carefully
mark capabilities. In addition, yet other observations have raised control the production and release of growth-promoting signals
questions and highlighted mechanistic concepts that were not that instruct entry into and progression through the cell growth-
integral to our original elaboration of the hallmark traits. Moti- and-division cycle, thereby ensuring a homeostasis of cell

646 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


Figure 1. The Hallmarks of Cancer
This illustration encompasses the six hallmark
capabilities originally proposed in our 2000 per-
spective. The past decade has witnessed
remarkable progress toward understanding the
mechanistic underpinnings of each hallmark.

surface, rendering such cells hyperre-


sponsive to otherwise-limiting amounts
of growth factor ligand; the same
outcome can result from structural alter-
ations in the receptor molecules that
facilitate ligand-independent firing.
Growth factor independence may also
derive from the constitutive activation of
components of signaling pathways oper-
ating downstream of these receptors,
obviating the need to stimulate these
pathways by ligand-mediated receptor
number and thus maintenance of normal tissue architecture and activation. Given that a number of distinct downstream signaling
function. Cancer cells, by deregulating these signals, become pathways radiate from a ligand-stimulated receptor, the activa-
masters of their own destinies. The enabling signals are tion of one or another of these downstream pathways, for
conveyed in large part by growth factors that bind cell-surface example, the one responding to the Ras signal transducer,
receptors, typically containing intracellular tyrosine kinase may only recapitulate a subset of the regulatory instructions
domains. The latter proceed to emit signals via branched intra- transmitted by an activated receptor.
cellular signaling pathways that regulate progression through Somatic Mutations Activate Additional Downstream
the cell cycle as well as cell growth (that is, increases in cell Pathways
size); often these signals influence yet other cell-biological prop- High-throughput DNA sequencing analyses of cancer cell
erties, such as cell survival and energy metabolism. genomes have revealed somatic mutations in certain human
Remarkably, the precise identities and sources of the prolifer- tumors that predict constitutive activation of signaling circuits
ative signals operating within normal tissues were poorly under- usually triggered by activated growth factor receptors. Thus,
stood a decade ago and in general remain so. Moreover, we still we now know that 40% of human melanomas contain
know relatively little about the mechanisms controlling the activating mutations affecting the structure of the B-Raf protein,
release of these mitogenic signals. In part, the understanding resulting in constitutive signaling through the Raf to mitogen-
of these mechanisms is complicated by the fact that the growth activated protein (MAP)-kinase pathway (Davies and Samuels
factor signals controlling cell number and position within tissues 2010). Similarly, mutations in the catalytic subunit of phosphoi-
are thought to be transmitted in a temporally and spatially regu- nositide 3-kinase (PI3-kinase) isoforms are being detected in
lated fashion from one cell to its neighbors; such paracrine an array of tumor types, which serve to hyperactivate the PI3-
signaling is difficult to access experimentally. In addition, the kinase signaling circuitry, including its key Akt/PKB signal
bioavailability of growth factors is regulated by sequestration in transducer (Jiang and Liu, 2009; Yuan and Cantley, 2008). The
the pericellular space and extracellular matrix, and by the actions advantages to tumor cells of activating upstream (receptor)
of a complex network of proteases, sulfatases, and possibly versus downstream (transducer) signaling remain obscure, as
other enzymes that liberate and activate them, apparently in does the functional impact of crosstalk between the multiple
a highly specific and localized fashion. pathways radiating from growth factor receptors.
The mitogenic signaling in cancer cells is, in contrast, better Disruptions of Negative-Feedback Mechanisms that
understood (Lemmon and Schlessinger, 2010; Witsch et al., Attenuate Proliferative Signaling
2010; Hynes and MacDonald, 2009; Perona, 2006). Cancer cells Recent results have highlighted the importance of negative-
can acquire the capability to sustain proliferative signaling in feedback loops that normally operate to dampen various types
a number of alternative ways: They may produce growth factor of signaling and thereby ensure homeostatic regulation of the
ligands themselves, to which they can respond via the expres- flux of signals coursing through the intracellular circuitry (Wertz
sion of cognate receptors, resulting in autocrine proliferative and Dixit, 2010; Cabrita and Christofori, 2008; Amit et al.,
stimulation. Alternatively, cancer cells may send signals to stim- 2007; Mosesson et al., 2008). Defects in these feedback mech-
ulate normal cells within the supporting tumor-associated anisms are capable of enhancing proliferative signaling. The
stroma, which reciprocate by supplying the cancer cells with prototype of this type of regulation involves the Ras oncoprotein:
various growth factors (Cheng et al., 2008; Bhowmick et al., the oncogenic effects of Ras do not result from a hyperactivation
2004). Receptor signaling can also be deregulated by elevating of its signaling powers; instead, the oncogenic mutations
the levels of receptor proteins displayed at the cancer cell affecting ras genes compromise Ras GTPase activity, which

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 647


operates as an intrinsic negative-feedback mechanism that nor- Evading Growth Suppressors
mally ensures that active signal transmission is transitory. In addition to the hallmark capability of inducing and sustaining
Analogous negative-feedback mechanisms operate at positively acting growth-stimulatory signals, cancer cells must
multiple nodes within the proliferative signaling circuitry. A prom- also circumvent powerful programs that negatively regulate
inent example involves the PTEN phosphatase, which counter- cell proliferation; many of these programs depend on the actions
acts PI3-kinase by degrading its product, phosphatidylinositol of tumor suppressor genes. Dozens of tumor suppressors that
(3,4,5) trisphosphate (PIP3). Loss-of-function mutations in PTEN operate in various ways to limit cell growth and proliferation
amplify PI3K signaling and promote tumorigenesis in a variety have been discovered through their characteristic inactivation
of experimental models of cancer; in human tumors, PTEN in one or another form of animal or human cancer; many of these
expression is often lost by promoter methylation (Jiang and genes have been validated as bona fide tumor suppressors
Liu, 2009; Yuan and Cantley, 2008). through gain- or loss-of-function experiments in mice. The two
Yet another example involves the mTOR kinase, a coordinator prototypical tumor suppressors encode the RB (retinoblas-
of cell growth and metabolism that lies both upstream and down- toma-associated) and TP53 proteins; they operate as central
stream of the PI3K pathway. In the circuitry of some cancer cells, control nodes within two key complementary cellular regulatory
mTOR activation results, via negative feedback, in the inhibition circuits that govern the decisions of cells to proliferate or, alter-
of PI3K signaling. Thus, when mTOR is pharmacologically natively, activate senescence and apoptotic programs.
inhibited in such cancer cells (such as by the drug rapamycin), The RB protein integrates signals from diverse extracellular
the associated loss of negative feedback results in increased and intracellular sources and, in response, decides whether or
activity of PI3K and its effector Akt/PKB, thereby blunting the not a cell should proceed through its growth-and-division cycle
antiproliferative effects of mTOR inhibition (Sudarsanam and (Burkhart and Sage, 2008; Deshpande et al., 2005; Sherr and
Johnson, 2010; O’Reilly et al., 2006). It is likely that compromised McCormick, 2002). Cancer cells with defects in RB pathway
negative-feedback loops in this and other signaling pathways function are thus missing the services of a critical gatekeeper
will prove to be widespread among human cancer cells and of cell-cycle progression whose absence permits persistent
serve as an important means by which these cells can achieve cell proliferation. Whereas RB transduces growth-inhibitory
proliferative independence. Moreover, disruption of such self- signals that originate largely outside of the cell, TP53 receives
attenuating signaling may contribute to the development of inputs from stress and abnormality sensors that function within
adaptive resistance toward drugs targeting mitogenic signaling. the cell’s intracellular operating systems: if the degree of
Excessive Proliferative Signaling Can Trigger Cell damage to the genome is excessive, or if the levels of nucleotide
Senescence pools, growth-promoting signals, glucose, or oxygenation are
Early studies of oncogene action encouraged the notion that suboptimal, TP53 can call a halt to further cell-cycle progression
ever-increasing expression of such genes and the signals mani- until these conditions have been normalized. Alternatively, in the
fested in their protein products would result in correspondingly face of alarm signals indicating overwhelming or irreparable
increased cancer cell proliferation and thus tumor growth. More damage to such cellular subsystems, TP53 can trigger
recent research has undermined this notion, in that excessively apoptosis. Notably, the various effects of activated TP53 are
elevated signaling by oncoproteins such as RAS, MYC, and complex and highly context dependent, varying by cell type as
RAF can provoke counteracting responses from cells, specifi- well as by the severity and persistence of conditions of cell stress
cally induction of cell senescence and/or apoptosis (Collado and genomic damage.
and Serrano, 2010; Evan and d’Adda di Fagagna, 2009; Lowe Although the two canonical suppressors of proliferation—
et al., 2004). For example, cultured cells expressing high levels TP53 and RB—have preeminent importance in regulating cell
of the Ras oncoprotein may enter into the nonproliferative but proliferation, various lines of evidence indicate that each oper-
viable state called senescence; in contrast, cells expressing ates as part of a larger network that is wired for functional redun-
lower levels of this protein may avoid senescence and proliferate. dancy. For example, chimeric mice populated throughout their
Cells with morphological features of senescence, including bodies with individual cells lacking a functional Rb gene are
enlarged cytoplasm, the absence of proliferation markers, and surprisingly free of proliferative abnormalities, despite the expec-
expression of the senescence-induced b-galactosidase tation that loss of RB function would allow continuous firing of the
enzyme, are abundant in the tissues of mice engineered to over- cell division cycle in these cells and their lineal descendants;
express certain oncogenes (Collado and Serrano, 2010; Evan some of the resulting clusters of Rb null cells should, by all rights,
and d’Adda di Fagagna, 2009) and are prevalent in some cases progress to neoplasia. Instead, the Rb null cells in such chimeric
of human melanoma (Mooi and Peeper, 2006). These ostensibly mice have been found to participate in relatively normal tissue
paradoxical responses seem to reflect intrinsic cellular defense morphogenesis throughout the body; the only neoplasia
mechanisms designed to eliminate cells experiencing excessive observed was in the development of pituitary tumors late in life
levels of certain types of signaling. Accordingly, the relative (Lipinski and Jacks, 1999). Similarly, TP53 null mice develop nor-
intensity of oncogenic signaling in cancer cells may represent mally, show largely proper cell and tissue homeostasis, and
compromises between maximal mitogenic stimulation and again develop abnormalities later in life, in the form of leukemias
avoidance of these antiproliferative defenses. Alternatively, and sarcomas (Ghebranious and Donehower, 1998). Both exam-
some cancer cells may adapt to high levels of oncogenic ples must reflect the operations of redundantly acting mecha-
signaling by disabling their senescence- or apoptosis-inducing nisms that serve to constrain inappropriate replication of cells
circuitry. lacking these key proliferation suppressors.

648 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


Mechanisms of Contact Inhibition and Its Evasion Littlewood, 1998). Elucidation of the signaling circuitry governing
Four decades of research have demonstrated that the cell-to- the apoptotic program has revealed how apoptosis is triggered
cell contacts formed by dense populations of normal cells prop- in response to various physiologic stresses that cancer cells
agated in two-dimensional culture operate to suppress further experience during the course of tumorigenesis or as a result of
cell proliferation, yielding confluent cell monolayers. Importantly, anticancer therapy. Notable among the apoptosis-inducing
such ‘‘contact inhibition’’ is abolished in various types of cancer stresses are signaling imbalances resulting from elevated levels
cells in culture, suggesting that contact inhibition is an in vitro of oncogene signaling, as mentioned earlier, and DNA damage
surrogate of a mechanism that operates in vivo to ensure normal associated with hyperproliferation. Yet other research has re-
tissue homeostasis, one that is abrogated during the course of vealed how apoptosis is attenuated in those tumors that
tumorigenesis. Until recently, the mechanistic basis for this succeed in progressing to states of high-grade malignancy and
mode of growth control remained obscure. Now, however, resistance to therapy (Adams and Cory, 2007; Lowe et al., 2004).
mechanisms of contact inhibition are beginning to emerge. The apoptotic machinery is composed of both upstream regu-
One mechanism involves the product of the NF2 gene, long lators and downstream effector components (Adams and Cory,
implicated as a tumor suppressor because its loss triggers 2007). The regulators, in turn, are divided into two major circuits,
a form of human neurofibromatosis. Merlin, the cytoplasmic one receiving and processing extracellular death-inducing
NF2 gene product, orchestrates contact inhibition via coupling signals (the extrinsic apoptotic program, involving for example
cell-surface adhesion molecules (e.g., E-cadherin) to transmem- the Fas ligand/Fas receptor), and the other sensing and inte-
brane receptor tyrosine kinases (e.g., the EGF receptor). In so grating a variety of signals of intracellular origin (the intrinsic
doing, Merlin strengthens the adhesivity of cadherin-mediated program). Each culminates in activation of a normally latent
cell-to-cell attachments. Additionally, by sequestering growth protease (caspases 8 and 9, respectively), which proceeds to
factor receptors, Merlin limits their ability to efficiently emit mito- initiate a cascade of proteolysis involving effector caspases
genic signals (Curto et al., 2007; Okada et al., 2005). responsible for the execution phase of apoptosis, in which the
A second mechanism of contact inhibition involves the LKB1 cell is progressively disassembled and then consumed, both
epithelial polarity protein, which organizes epithelial structure by its neighbors and by professional phagocytic cells. Currently,
and helps maintain tissue integrity. LKB1 can, for example, the intrinsic apoptotic program is more widely implicated as
overrule the mitogenic effects of the powerful Myc oncogene a barrier to cancer pathogenesis.
when the latter is upregulated in organized, quiescent epithelial The ‘‘apoptotic trigger’’ that conveys signals between the regu-
structures; in contrast, when LKB1 expression is suppressed, lators and effectors is controlled by counterbalancing pro- and
epithelial integrity is destabilized, and epithelial cells become antiapoptotic members of the Bcl-2 family of regulatory proteins
susceptible to Myc-induced transformation (Partanen et al., (Adams and Cory, 2007). The archetype, Bcl-2, along with its
2009; Hezel and Bardeesy, 2008). LKB1 has also been identified closest relatives (Bcl-xL, Bcl-w, Mcl-1, A1) are inhibitors of
as a tumor suppressor gene that is lost in certain human malig- apoptosis, acting in large part by binding to and thereby suppress-
nancies (Shaw, 2009), possibly reflecting its normal function as ing two proapoptotic triggering proteins (Bax and Bak); the latter
a suppressor of inappropriate proliferation. It remains to be are embedded in the mitochondrial outer membrane. When
seen how frequently these two mechanisms of contact-medi- relieved of inhibition by their antiapoptotic relatives, Bax and
ated growth suppression are compromised in human cancers; Bak disrupt the integrity of the outer mitochondrial membrane,
no doubt yet other contact-induced proliferative barriers are causing the release of proapoptotic signaling proteins, the most
yet to be discovered. Clearly mechanisms like these that enable important of which is cytochrome c. The released cytochrome c
cells to construct and maintain architecturally complex tissues activates, in turn, a cascade of caspases that act via their proteo-
represent important means of suppressing and counterbalanc- lytic activities to induce the multiple cellular changes associated
ing inappropriate proliferative signals. with the apoptotic program. Bax and Bak share protein-protein
Corruption of the TGF-b Pathway Promotes Malignancy interaction domains, termed BH3 motifs, with the antiapoptotic
TGF-b is best known for its antiproliferative effects, and evasion Bcl-2-like proteins that mediate their various physical interac-
by cancer cells of these effects is now appreciated to be far more tions. The activities of a subfamily of related proteins, each of
elaborate than simple shutdown of its signaling circuitry (Ikush- which contains a single such BH3 motif, are coupled to a variety
ima and Miyazono, 2010; Massagué, 2008; Bierie and Moses, of sensors of cellular abnormality; these ‘‘BH3-only’’ proteins
2006). In many late-stage tumors, TGF-b signaling is redirected act either by interfering with antiapoptotic Bcl-2 proteins or by
away from suppressing cell proliferation and is found instead directly stimulating the proapoptotic members of this family
to activate a cellular program, termed the epithelial-to-mesen- (Adams and Cory, 2007; Willis and Adams, 2005).
chymal transition (EMT), that confers on cancer cells traits asso- Although the cellular conditions that trigger apoptosis remain
ciated with high-grade malignancy, as discussed in further detail to be fully enumerated, several abnormality sensors that play
below. key roles in tumor development have been identified (Adams
and Cory, 2007; Lowe et al., 2004). Most notable is a DNA-
Resisting Cell Death damage sensor that functions via the TP53 tumor suppressor
The concept that programmed cell death by apoptosis serves as (Junttila and Evan, 2009); TP53 induces apoptosis by upregulat-
a natural barrier to cancer development has been established by ing expression of the Noxa and Puma BH3-only proteins, doing
compelling functional studies conducted over the last two so in response to substantial levels of DNA breaks and other
decades (Adams and Cory, 2007; Lowe et al., 2004: Evan and chromosomal abnormalities. Alternatively, insufficient survival

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 649


factor signaling (for instance inadequate levels of interleukin-3 in proteins, and its BH3 domain allows it to bind the Bcl-2/Bcl-xL
lymphocytes or of insulin-like growth factor 1/2 [Igf1/2] in epithe- proteins. Stress-sensor-coupled BH3 proteins can displace Be-
lial cells) can elicit apoptosis through a BH3-only protein called clin-1 from its association with Bcl-2/Bcl-xL, enabling the liber-
Bim. Yet another condition leading to cell death involves hyper- ated Beclin-1 to trigger autophagy, much as they can release
active signaling by certain oncoproteins, such as Myc, which proapoptotic Bax and Bak to trigger apoptosis. Hence, stress-
triggers apoptosis (in part via Bim and other BH3-only proteins) transducing BH3 proteins (e.g., Bid, Bad, Puma, et al.) can
unless counterbalanced by antiapoptotic factors (Junttila and induce apoptosis and/or autophagy depending on the physio-
Evan, 2009; Lowe et al., 2004). logic state of the cell.
Tumor cells evolve a variety of strategies to limit or circumvent Mice bearing inactivated alleles of the Beclin-1 gene or of
apoptosis. Most common is the loss of TP53 tumor suppressor certain other components of the autophagy machinery exhibit
function, which eliminates this critical damage sensor from the increased susceptibility to cancer (White and DiPaola, 2009:
apoptosis-inducing circuitry. Alternatively, tumors may achieve Levine and Kroemer, 2008). These results suggest that induction
similar ends by increasing expression of antiapoptotic regulators of autophagy can serve as a barrier to tumorigenesis that may
(Bcl-2, Bcl-xL) or of survival signals (Igf1/2), by downregulating operate independently of or in concert with apoptosis. Accord-
proapoptotic factors (Bax, Bim, Puma), or by short-circuiting ingly, autophagy appears to represent yet another barrier that
the extrinsic ligand-induced death pathway. The multiplicity of needs to be circumvented during tumor development (White
apoptosis-avoiding mechanisms presumably reflects the diver- and DiPaola, 2009).
sity of apoptosis-inducing signals that cancer cell populations Perhaps paradoxically, nutrient starvation, radiotherapy, and
encounter during their evolution to the malignant state. certain cytotoxic drugs can induce elevated levels of autophagy
The structure of the apoptotic machinery and program, and that are apparently cytoprotective for cancer cells, impairing
the strategies used by cancer cells to evade its actions, were rather than accentuating the killing actions of these stress-
widely appreciated by the beginning of the last decade. The inducing situations (White and DiPaola, 2009; Apel et al., 2009;
most notable conceptual advances since then have involved Amaravadi and Thompson, 2007; Mathew et al., 2007). More-
other forms of cell death that broaden the scope of ‘‘pro- over, severely stressed cancer cells have been shown to shrink
grammed cell death’’ as a barrier to cancer. via autophagy to a state of reversible dormancy (White and
Autophagy Mediates Both Tumor Cell Survival and Death DiPaola, 2009; Lu et al., 2008). This survival response may
Autophagy represents an important cell-physiologic response enable the persistence and eventual regrowth of some late-
that, like apoptosis, normally operates at low, basal levels in cells stage tumors following treatment with potent anticancer agents.
but can be strongly induced in certain states of cellular stress, Thus, in analogy to TGF-b signaling, which can be tumor sup-
the most obvious of which is nutrient deficiency (Levine and pressing at early stages of tumorigenesis and tumor promoting
Kroemer, 2008; Mizushima, 2007). The autophagic program later on, autophagy seems to have conflicting effects on tumor
enables cells to break down cellular organelles, such as ribo- cells and thus tumor progression (Apel et al., 2009; White and
somes and mitochondria, allowing the resulting catabolites to DiPaola, 2009). An important agenda for future research will
be recycled and thus used for biosynthesis and energy metabo- involve clarifying the genetic and cell-physiologic conditions
lism. As part of this program, intracellular vesicles termed auto- that dictate when and how autophagy enables cancer cells to
phagosomes envelope intracellular organelles and then fuse with survive or causes them to die.
lysosomes wherein degradation occurs. In this fashion, low- Necrosis Has Proinflammatory and Tumor-Promoting
molecular-weight metabolites are generated that support Potential
survival in the stressed, nutrient-limited environments experi- In contrast to apoptosis, in which a dying cell contracts into an
enced by many cancer cells. almost-invisible corpse that is soon consumed by neighbors,
Like apoptosis, the autophagy machinery has both regulatory necrotic cells become bloated and explode, releasing their
and effector components (Levine and Kroemer, 2008; Mizush- contents into the local tissue microenvironment. Although
ima, 2007). Among the latter are proteins that mediate autopha- necrosis has historically been viewed much like organismic
gosome formation and delivery to lysosomes. Of note, recent death, as a form of system-wide exhaustion and breakdown,
research has revealed intersections between the regulatory the conceptual landscape is changing: cell death by necrosis
circuits governing autophagy, apoptosis, and cellular homeo- is clearly under genetic control in some circumstances, rather
stasis. For example, the signaling pathway involving the PI3- than being a random and undirected process (Galluzzi and
kinase, AKT, and mTOR kinases, which is stimulated by survival Kroemer, 2008; Zong and Thompson, 2006).
signals to block apoptosis, similarly inhibits autophagy; when Perhaps more important, necrotic cell death releases proin-
survival signals are insufficient, the PI3K signaling pathway is flammatory signals into the surrounding tissue microenviron-
downregulated, with the result that autophagy and/or apoptosis ment, in contrast to apoptosis and autophagy, which do not.
may be induced (Levine and Kroemer, 2008; Sinha and Levine, As a consequence, necrotic cells can recruit inflammatory cells
2008; Mathew et al., 2007). of the immune system (Grivennikov et al., 2010; White et al.,
Another interconnection between these two programs resides 2010; Galluzzi and Kroemer, 2008), whose dedicated function
in the Beclin-1 protein, which has been shown by genetic studies is to survey the extent of tissue damage and remove associated
to be necessary for induction of autophagy (Levine and Kroemer, necrotic debris. In the context of neoplasia, however, multiple
2008; Sinha and Levine, 2008; Mizushima, 2007). Beclin-1 is lines of evidence indicate that immune inflammatory cells can
a member of the BH3-only subfamily of apoptotic regulatory be actively tumor promoting, given that such cells are capable

650 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


of fostering angiogenesis, cancer cell proliferation, and invasive- sion of telomerase activity leads to telomere shortening and to
ness (see below). Additionally, necrotic cells can release bio- activation of one or the other of these proliferative barriers.
active regulatory factors, such as IL-1a, which can directly stim- The two barriers to proliferation—senescence and crisis/
ulate neighboring viable cells to proliferate, with the potential, apoptosis—have been rationalized as crucial anticancer
once again, to facilitate neoplastic progression (Grivennikov defenses that are hard-wired into our cells, being deployed to
et al., 2010). Consequently, necrotic cell death, while seemingly impede the outgrowth of clones of preneoplastic and frankly
beneficial in counterbalancing cancer-associated hyperprolifer- neoplastic cells. According to this thinking, most incipient
ation, may ultimately do more damage than good. Accordingly, neoplasias exhaust their endowment of replicative doublings
incipient neoplasias and potentially invasive and metastatic and are stopped in their tracks by one or the other of these
tumors may gain an advantage by tolerating some degree of barriers. The eventual immortalization of rare variant cells that
necrotic cell death, doing so in order to recruit tumor-promoting proceed to form tumors has been attributed to their ability to
inflammatory cells that bring growth-stimulating factors to the maintain telomeric DNA at lengths sufficient to avoid triggering
surviving cells within these growths. senescence or apoptosis, achieved most commonly by upre-
gulating expression of telomerase or, less frequently, via an
Enabling Replicative Immortality alternative recombination-based telomere maintenance mech-
By 2000, it was widely accepted that cancer cells require unlim- anism. Hence, telomere shortening has come to be viewed as
ited replicative potential in order to generate macroscopic a clocking device that determines the limited replicative poten-
tumors. This capability stands in marked contrast to the behavior tial of normal cells and thus one that must be overcome by
of the cells in most normal cell lineages in the body, which are cancer cells.
able to pass through only a limited number of successive cell Reassessing Replicative Senescence
growth-and-division cycles. This limitation has been associated Whereas telomere maintenance has been increasingly substan-
with two distinct barriers to proliferation: senescence, a typically tiated as a condition critical to the neoplastic state, the concept
irreversible entrance into a nonproliferative but viable state, and of replication-induced senescence as a general barrier requires
crisis, which involves cell death. Accordingly, when cells are refinement and reformulation. (Differences in telomere structure
propagated in culture, repeated cycles of cell division lead first and function in mouse versus human cells have also complicated
to induction of senescence and then, for those cells that succeed investigation of the roles of telomeres and telomerase in replica-
in circumventing this barrier, to a crisis phase, in which the great tive senescence.) Recent experiments have revealed that the
majority of cells in the population die. On rare occasion, cells induction of senescence in certain cultured cells can be delayed
emerge from a population in crisis and exhibit unlimited replica- and possibly eliminated by the use of improved cell culture
tive potential. This transition has been termed immortalization, conditions, suggesting that recently explanted primary cells
a trait that most established cell lines possess by virtue of their may be able to proliferate unimpeded in culture up the point of
ability to proliferate in culture without evidence of either senes- crisis and the associated induction of apoptosis triggered by crit-
cence or crisis. ically shortened telomeres (Ince et al., 2007; Passos et al., 2007;
Multiple lines of evidence indicate that telomeres protecting Zhang et al., 2004; Sherr and DePinho, 2000). In contrast, exper-
the ends of chromosomes are centrally involved in the capability iments in mice engineered to lack telomerase indicate that the
for unlimited proliferation (Blasco, 2005; Shay and Wright, 2000). consequently shortened telomeres can shunt premalignant cells
The telomeres, composed of multiple tandem hexanucleotide into a senescent state that contributes (along with apoptosis) to
repeats, shorten progressively in nonimmortalized cells propa- attenuated tumorigenesis in mice genetically destined to
gated in culture, eventually losing the ability to protect the develop particular forms of cancer (Artandi and DePinho,
ends of chromosomal DNAs from end-to-end fusions; such 2010). Such telomerase null mice with highly eroded telomeres
fusions generate unstable dicentric chromosomes whose reso- exhibit multiorgan dysfunction and abnormalities that include
lution results in a scrambling of karyotype that threatens cell evidence for both senescence and apoptosis, perhaps analo-
viability. Accordingly, the length of telomeric DNA in a cell gous to the senescence and apoptosis observed in cell culture
dictates how many successive cell generations its progeny can (Artandi and DePinho, 2010; Feldser and Greider, 2007).
pass through before telomeres are largely eroded and have Of note, and as discussed earlier, a morphologically similar
consequently lost their protective functions, triggering entrance form of cell senescence induced by excessive or unbalanced
into crisis. oncogene signaling is now well documented as a protective
Telomerase, the specialized DNA polymerase that adds telo- mechanism against neoplasia; the possible interconnections of
mere repeat segments to the ends of telomeric DNA, is almost this form of senescence with telomerase and telomeres remain
absent in nonimmortalized cells but expressed at functionally to be ascertained. Thus, cell senescence is emerging conceptu-
significant levels in the vast majority (90%) of spontaneously ally as a protective barrier to neoplastic expansion that can be
immortalized cells, including human cancer cells. By extending triggered by various proliferation-associated abnormalities,
telomeric DNA, telomerase is able to counter the progressive including high levels of oncogenic signaling and, apparently,
telomere erosion that would otherwise occur in its absence. subcritical shortening of telomeres.
The presence of telomerase activity, either in spontaneously Delayed Activation of Telomerase May Both Limit
immortalized cells or in the context of cells engineered to and Foster Neoplastic Progression
express the enzyme, is correlated with a resistance to induction There is now evidence that clones of incipient cancer cells often
of both senescence and crisis/apoptosis; conversely, suppres- experience telomere loss-induced crisis relatively early during

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 651


the course of multistep tumor progression due to their inability to the Wnt pathway, by serving as a cofactor of the b-catenin/LEF
express significant levels of telomerase. Thus, extensively transcription factor complex (Park et al., 2009). Other ascribed
eroded telomeres have been documented in premalignant telomere-independent effects include demonstrable enhance-
growths through the use of fluorescence in situ hybridization ment of cell proliferation and/or resistance to apoptosis (Kang
(FISH), which has also revealed the end-to-end chromosomal et al., 2004), involvement in DNA-damage repair (Masutomi
fusions that signal telomere failure and crisis (Kawai et al., et al., 2005), and RNA-dependent RNA polymerase function
2007; Hansel et al., 2006). These results also suggest that such (Maida et al., 2009). Consistent with these broader roles, TERT
cells have passed through a substantial number of successive can be found associated with chromatin at multiple sites along
telomere-shortening cell divisions during their evolution from the chromosomes, not just at the telomeres (Park et al., 2009;
fully normal cells-of-origin. Accordingly, the development of Masutomi et al., 2005). Hence, telomere maintenance is proving
some human neoplasias may be aborted by telomere-induced to be the most prominent of a diverse series of functions to which
crisis long before they succeed in becoming macroscopic, TERT contributes. The contributions of these additional func-
frankly neoplastic growths. tions of telomerase to tumorigenesis remain to be fully eluci-
In contrast, the absence of TP53-mediated surveillance of dated.
genomic integrity may permit other incipient neoplasias to
survive initial telomere erosion and attendant chromosomal Inducing Angiogenesis
breakage-fusion-bridge (BFB) cycles. The genomic alterations Like normal tissues, tumors require sustenance in the form of
resulting from these BFB cycles, including deletions and ampli- nutrients and oxygen as well as an ability to evacuate metabolic
fications of chromosomal segments, evidently serve to increase wastes and carbon dioxide. The tumor-associated neovascula-
the mutability of the genome, thereby accelerating the acquisi- ture, generated by the process of angiogenesis, addresses these
tion of mutant oncogenes and tumor suppressor genes. The real- needs. During embryogenesis, the development of the vascula-
ization that impaired telomere function can actually foster tumor ture involves the birth of new endothelial cells and their assembly
progression has come from the study of mutant mice that lack into tubes (vasculogenesis) in addition to the sprouting (angio-
both p53 and telomerase function (Artandi and DePinho, 2010, genesis) of new vessels from existing ones. Following this
2000). The proposition that these two defects can cooperatively morphogenesis, the normal vasculature becomes largely quies-
enhance human tumorigenesis has not yet been directly docu- cent. In the adult, as part of physiologic processes such as
mented. wound healing and female reproductive cycling, angiogenesis
Circumstantial support for the importance of transient telo- is turned on, but only transiently. In contrast, during tumor
mere deficiency in facilitating malignant progression has come, progression, an ‘‘angiogenic switch’’ is almost always activated
in addition, from comparative analyses of premalignant and and remains on, causing normally quiescent vasculature to
malignant lesions in the human breast (Raynaud et al., 2010; continually sprout new vessels that help sustain expanding
Chin et al., 2004). The premalignant lesions did not express neoplastic growths (Hanahan and Folkman, 1996).
significant levels of telomerase and were marked by telomere A compelling body of evidence indicates that the angiogenic
shortening and nonclonal chromosomal aberrations. In contrast, switch is governed by countervailing factors that either induce
overt carcinomas exhibited telomerase expression concordantly or oppose angiogenesis (Baeriswyl and Christofori, 2009; Berg-
with the reconstruction of longer telomeres and the fixation (via ers and Benjamin, 2003). Some of these angiogenic regulators
clonal outgrowth) of the aberrant karyotypes that would seem are signaling proteins that bind to stimulatory or inhibitory cell-
to have been acquired after telomere failure but before the acqui- surface receptors displayed by vascular endothelial cells. The
sition of telomerase activity. When portrayed in this way, the well-known prototypes of angiogenesis inducers and inhibitors
delayed acquisition of telomerase function serves to generate are vascular endothelial growth factor-A (VEGF-A) and thrombo-
tumor-promoting mutations, whereas its subsequent activation spondin-1 (TSP-1), respectively.
stabilizes the mutant genome and confers the unlimited replica- The VEGF-A gene encodes ligands that are involved in orches-
tive capacity that cancer cells require in order to generate clini- trating new blood vessel growth during embryonic and postnatal
cally apparent tumors. development, and then in homeostatic survival of endothelial
New Functions of Telomerase cells, as well as in physiological and pathological situations in
Telomerase was discovered because of its ability to elongate the adult. VEGF signaling via three receptor tyrosine kinases
and maintain telomeric DNA, and almost all telomerase research (VEGFR-1–3) is regulated at multiple levels, reflecting this
has been posited on the notion that its functions are confined to complexity of purpose. Thus, VEGF gene expression can by
this crucial function. However, in recent years it has become upregulated both by hypoxia and by oncogene signaling (Fer-
apparent that telomerase exerts functions that are relevant to rara, 2009; Mac Gabhann and Popel, 2008; Carmeliet, 2005).
cell proliferation but unrelated to telomere maintenance. The Additionally, VEGF ligands can be sequestered in the extracel-
noncanonical roles of telomerase, and in particular its protein lular matrix in latent forms that are subject to release and activa-
subunit TERT, have been revealed by functional studies in tion by extracellular matrix-degrading proteases (e.g., MMP-9;
mice and cultured cells; in some cases novel functions have Kessenbrock et al., 2010). In addition, other proangiogenic
been demonstrated in conditions where the telomerase enzy- signals, such as members of the fibroblast growth factor (FGF)
matic activity has been eliminated (Cong and Shay, 2008). family, have been implicated in sustaining tumor angiogenesis
Among the growing list of telomere-independent functions of when their expression is chronically upregulated (Baeriswyl
TERT/telomerase is the ability of TERT to amplify signaling by and Christofori, 2009). TSP-1, a key counterbalance in the

652 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


angiogenic switch, also binds transmembrane receptors dis- Kazerounian, et al., 2008; Folkman, 2006, 2002; Nyberg et al.,
played by endothelial cells and thereby evokes suppressive 2005). The last decade has seen reports of another dozen
signals that can counteract proangiogenic stimuli (Kazerounian such agents (Ribatti, 2009; Folkman, 2006; Nyberg et al.,
et al., 2008). 2005). Most are proteins, and many are derived by proteolytic
The blood vessels produced within tumors by chronically acti- cleavage of structural proteins that are not themselves angio-
vated angiogenesis and an unbalanced mix of proangiogenic genic regulators. A number of these endogenous inhibitors of
signals are typically aberrant: tumor neovasculature is marked angiogenesis can be detected in the circulation of normal
by precocious capillary sprouting, convoluted and excessive mice and humans. The genes encoding several endogenous
vessel branching, distorted and enlarged vessels, erratic blood angiogenesis inhibitors have been deleted from the mouse
flow, microhemorrhaging, leakiness, and abnormal levels of germline without untoward physiological effects; the growth of
endothelial cell proliferation and apoptosis (Nagy et al., 2010; autochthonous and implanted tumors, however, is enhanced
Baluk et al., 2005). as a consequence (Ribatti, 2009; Nyberg et al., 2005). By
Angiogenesis is induced surprisingly early during the multi- contrast, if the circulating levels of an endogenous inhibitor
stage development of invasive cancers both in animal models are genetically increased (e.g., via overexpression in transgenic
and in humans. Histological analyses of premalignant, noninva- mice or in xenotransplanted tumors), tumor growth is impaired
sive lesions, including dysplasias and in situ carcinomas arising (Ribatti, 2009; Nyberg et al., 2005); interestingly, wound healing
in a variety of organs, have revealed the early tripping of the and fat deposition are impaired or accelerated by elevated or
angiogenic switch (Raica et al., 2009; Hanahan and Folkman, ablated expression of such genes (Cao, 2010; Seppinen et al.,
1996). Historically, angiogenesis was envisioned to be important 2008). The data suggest that such endogenous angiogenesis
only when rapidly growing macroscopic tumors had formed, but inhibitors serve under normal circumstances as physiologic
more recent data indicate that angiogenesis also contributes to regulators that modulate transitory angiogenesis during tissue
the microscopic premalignant phase of neoplastic progression, remodeling and wound healing; they may also act as intrinsic
further cementing its status as an integral hallmark of cancer. barriers to induction and/or persistence of angiogenesis by
The past decade has witnessed an astonishing outpouring of incipient neoplasias.
research on angiogenesis. Amid this wealth of new knowledge, Pericytes Are Important Components
we highlight several advances of particular relevance to tumor of the Tumor Neovasculature
physiology. Pericytes have long been known as supporting cells that are
Gradations of the Angiogenic Switch closely apposed to the outer surfaces of the endothelial tubes
Once angiogenesis has been activated, tumors exhibit diverse in normal tissue vasculature, where they provide important
patterns of neovascularization. Some tumors, including such mechanical and physiologic support to the endothelial cells.
highly aggressive types as pancreatic ductal adenocarcinomas, Tumor-associated vasculature, in contrast, was portrayed as
are hypovascularized and replete with stromal ‘‘deserts’’ that are lacking appreciable coverage by these auxiliary cells. However,
largely avascular and indeed may even be actively antiangio- careful microscopic studies conducted in recent years have re-
genic (Olive et al., 2009). Many other tumors, including human vealed that pericytes are associated, albeit loosely, with the neo-
renal and pancreatic neuroendocrine carcinomas, are highly vasculature of most if not all tumors (Raza et al., 2010; Bergers
angiogenic and consequently densely vascularized (Zee et al., and Song, 2005). More importantly, mechanistic studies dis-
2010; Turner et al., 2003). cussed below have revealed that pericyte coverage is important
Collectively, such observations suggest an initial tripping of for the maintenance of a functional tumor neovasculature.
the angiogenic switch during tumor development that is followed A Variety of Bone Marrow-Derived Cells Contribute
by a variable intensity of ongoing neovascularization, the latter to Tumor Angiogenesis
being controlled by a complex biological rheostat that involves It is now clear that a repertoire of cell types originating in the bone
both the cancer cells and the associated stromal microenviron- marrow play crucial roles in pathological angiogenesis (Qian and
ment (Baeriswyl and Christofori, 2009; Bergers and Benjamin, Pollard, 2010; Zumsteg and Christofori, 2009; Murdoch et al.,
2003). Of note, the switching mechanism can vary in its form, 2008; De Palma et al., 2007). These include cells of the innate
even though the net result is a common inductive signal (e.g., immune system—notably macrophages, neutrophils, mast cells,
VEGF). In some tumors, dominant oncogenes operating within and myeloid progenitors—that infiltrate premalignant lesions
tumor cells, such as Ras and Myc, can upregulate expression and progressed tumors and assemble at the margins of such
of angiogenic factors, whereas in others, such inductive signals lesions; the peri-tumoral inflammatory cells help to trip the angio-
are produced indirectly by immune inflammatory cells, as dis- genic switch in previously quiescent tissue and to sustain
cussed below. The direct induction of angiogenesis by onco- ongoing angiogenesis associated with tumor growth, in addition
genes that also drive proliferative signaling illustrates the impor- to facilitating local invasion, as noted below. In addition, they can
tant principle that distinct hallmark capabilities can be help protect the vasculature from the effects of drugs targeting
coregulated by the same transforming agents. endothelial cell signaling (Ferrara, 2010). Additionally, several
Endogenous Angiogenesis Inhibitors Present Natural types of bone marrow-derived ‘‘vascular progenitor cells’’ have
Barriers to Tumor Angiogenesis been observed in certain cases to have migrated into neoplastic
Research in the 1990s revealed that TSP-1 as well as fragments lesions and become intercalated into the neovasculature as peri-
of plasmin (angiostatin) and type 18 collagen (endostatin) can cytes or endothelial cells (Patenaude et al., 2010; Kovacic and
act as endogenous inhibitors of angiogenesis (Ribatti, 2009; Boehm, 2009; Lamagna and Bergers, 2006).

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 653


Activating Invasion and Metastasis can acquire the abilities to invade, to resist apoptosis, and to
In 2000, the mechanisms underlying invasion and metastasis disseminate (Klymkowsky and Savagner, 2009; Polyak and
were largely an enigma. It was clear that as carcinomas arising Weinberg, 2009; Thiery et al., 2009; Yilmaz and Christofori,
from epithelial tissues progressed to higher pathological grades 2009; Barrallo-Gimeno and Nieto, 2005). By co-opting a process
of malignancy, reflected in local invasion and distant metastasis, involved in various steps of embryonic morphogenesis and
the associated cancer cells typically developed alterations in wound healing, carcinoma cells can concomitantly acquire
their shape as well as in their attachment to other cells and to multiple attributes that enable invasion and metastasis. This
the extracellular matrix (ECM). The best characterized alteration multifaceted EMT program can be activated transiently or stably,
involved the loss by carcinoma cells of E-cadherin, a key cell-to- and to differing degrees, by carcinoma cells during the course of
cell adhesion molecule. By forming adherens junctions with invasion and metastasis.
adjacent epithelial cells, E-cadherin helps to assemble epithelial A set of pleiotropically acting transcriptional factors, including
cell sheets and maintain the quiescence of the cells within these Snail, Slug, Twist, and Zeb1/2, orchestrate the EMT and related
sheets. Increased expression of E-cadherin was well established migratory processes during embryogenesis; most were initially
as an antagonist of invasion and metastasis, whereas reduction identified by developmental genetics. These transcriptional
of its expression was known to potentiate these phenotypes. The regulators are expressed in various combinations in a number
frequently observed downregulation and occasional mutational of malignant tumor types and have been shown in experimental
inactivation of E-cadherin in human carcinomas provided strong models of carcinoma formation to be causally important for
support for its role as a key suppressor of this hallmark capability programming invasion; some have been found to elicit metas-
(Berx and van Roy, 2009; Cavallaro and Christofori, 2004). tasis when ectopically overexpressed (Micalizzi et al., 2010;
Additionally, expression of genes encoding other cell-to-cell Taube et al., 2010; Schmalhofer et al., 2009; Yang and Weinberg,
and cell-to-ECM adhesion molecules is demonstrably altered 2008). Included among the cell-biological traits evoked by such
in some highly aggressive carcinomas, with those favoring cyto- transcription factors are loss of adherens junctions and associ-
stasis typically being downregulated. Conversely, adhesion ated conversion from a polygonal/epithelial to a spindly/fibro-
molecules normally associated with the cell migrations that blastic morphology, expression of matrix-degrading enzymes,
occur during embryogenesis and inflammation are often upregu- increased motility, and heightened resistance to apoptosis—all
lated. For example, N-cadherin, which is normally expressed in traits implicated in the processes of invasion and metastasis.
migrating neurons and mesenchymal cells during organogen- Several of these transcription factors can directly repress E-cad-
esis, is upregulated in many invasive carcinoma cells. Beyond herin gene expression, thereby depriving neoplastic epithelial
the gain and loss of such cell-cell/matrix attachment proteins, cells of this key suppressor of motility and invasiveness (Peinado
the master regulators of invasion and metastasis were largely et al., 2004).
unknown or, when suspected, lacking in functional validation The available evidence suggests that these transcription
(Cavallaro and Christofori, 2004). factors regulate one another as well as overlapping sets of target
The multistep process of invasion and metastasis has been genes. No rules have yet been established to describe their inter-
schematized as a sequence of discrete steps, often termed the actions and the conditions that govern their expression.
invasion-metastasis cascade (Talmadge and Fidler, 2010; Fidler, Evidence from developmental genetics indicates that contextual
2003). This depiction envisions a succession of cell-biologic signals received from neighboring cells in the embryo are
changes, beginning with local invasion, then intravasation by involved in triggering expression of these transcription factors
cancer cells into nearby blood and lymphatic vessels, transit of in those cells destined to pass through an EMT (Micalizzi et al.,
cancer cells through the lymphatic and hematogenous systems, 2010); in an analogous fashion, increasing evidence suggests
followed by escape of cancer cells from the lumina of such that heterotypic interactions of cancer cells with adjacent
vessels into the parenchyma of distant tissues (extravasation), tumor-associated stromal cells can induce expression of the
the formation of small nodules of cancer cells (micrometasta- malignant cell phenotypes that are known to be choreographed
ses), and finally the growth of micrometastatic lesions into by one or more of these transcriptional regulators (Karnoub and
macroscopic tumors, this last step being termed ‘‘colonization.’’ Weinberg, 2006–2007; Brabletz et al., 2001). Moreover, cancer
Research into the capability for invasion and metastasis has cells at the invasive margins of certain carcinomas can be
accelerated dramatically over the past decade as powerful seen to have undergone an EMT, suggesting that these cancer
new research tools and refined experimental models have cells are subject to microenvironmental stimuli distinct from
become available, and as critical regulatory genes were identi- those received by cancer cells located in the cores of these
fied. While still an emerging field replete with major unanswered lesions (Hlubek et al., 2007).
questions, significant progress has been made in delineating Although the evidence is still incomplete, it would appear that
important features of this complex hallmark capability. An admit- EMT-inducing transcription factors are able to orchestrate most
tedly incomplete representation of these advances is highlighted steps of the invasion-metastasis cascade save the final step of
below. colonization. We still know rather little about the various manifes-
The EMT Program Broadly Regulates Invasion tations and temporal stability of the mesenchymal state
and Metastasis produced by an EMT. Although expression of EMT-inducing
A developmental regulatory program, referred to as the ‘‘epithe- transcription factors has been observed in certain nonepithelial
lial-mesenchymal transition’’ (EMT), has become prominently tumor types, such as sarcomas and neuroectodermal tumors,
implicated as a means by which transformed epithelial cells their roles in programming malignant traits in these tumors are

654 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


presently poorly documented. Additionally, it remains to be simplistic; instead, in many cases, cancer cells may enter into
determined whether invasive carcinoma cells necessarily an EMT program only partially, thereby acquiring new mesen-
acquire their capability through activation of parts of the EMT chymal traits while continuing to express residual epithelial traits.
program, or whether alternative regulatory programs can also Distinct Forms of Invasion May Underlie Different
enable this capability. Cancer Types
Heterotypic Contributions of Stromal Cells to Invasion The EMT program regulates a particular type of invasiveness
and Metastasis that has been termed ‘‘mesenchymal.’’ In addition, two other
It is increasingly apparent that crosstalk between cancer cells distinct modes of invasion have been identified and implicated
and cells of the neoplastic stroma is involved in the acquired in cancer cell invasion (Friedl and Wolf, 2008, 2010). ‘‘Collective
capability for invasive growth and metastasis (Egeblad et al., invasion’’ involves nodules of cancer cells advancing en masse
2010; Qian and Pollard, 2010; Joyce and Pollard, 2009; Kalluri into adjacent tissues and is characteristic of, for example,
and Zeisberg, 2006). Such signaling may impinge on carcinoma squamous cell carcinomas; interestingly, such cancers are
cells and act to alter their hallmark capabilities as suggested rarely metastatic, suggesting that this form of invasion lacks
above. For example, mesenchymal stem cells (MSCs) present certain functional attributes that facilitate metastasis. Less clear
in the tumor stroma have been found to secrete CCL5/RANTES is the prevalence of an ‘‘amoeboid’’ form of invasion (Madsen
in response to signals released by cancer cells; CCL5 then acts and Sahai, 2010; Sabeh et al., 2009), in which individual cancer
reciprocally on the cancer cells to stimulate invasive behavior cells show morphological plasticity, enabling them to slither
(Karnoub et al., 2007). through existing interstices in the extracellular matrix rather
Macrophages at the tumor periphery can foster local invasion than clearing a path for themselves, as occurs in both the mesen-
by supplying matrix-degrading enzymes such as metalloprotei- chymal and collective forms of invasion. It is presently unre-
nases and cysteine cathepsin proteases (Kessenbrock et al., solved whether cancer cells participating in the collective and
2010; Joyce and Pollard, 2009; Palermo and Joyce, 2008; Mo- amoeboid forms of invasion employ components of the EMT
hamed and Sloane, 2006); in one model system, the invasion- program, or whether entirely different cell-biological programs
promoting macrophages are activated by IL-4 produced by the are responsible for choreographing these alternative invasion
cancer cells (Gocheva et al., 2010). And in an experimental programs.
model of metastatic breast cancer, tumor-associated macro- Another emerging concept, noted above, involves the facilita-
phages (TAMs) supply epidermal growth factor (EGF) to breast tion of cancer cell invasion by inflammatory cells that assemble
cancer cells, while the cancer cells reciprocally stimulate the at the boundaries of tumors, producing the extracellular
macrophages with CSF-1; their concerted interactions facilitate matrix-degrading enzymes and other factors that enable inva-
intravasation into the circulatory system and metastatic dissem- sive growth (Kessenbrock et al., 2010; Qian and Pollard, 2010;
ination of the cancer cells (Qian and Pollard, 2010; Wyckoff et al., Joyce and Pollard, 2009); these functions may obviate the
2007). need of cancer cells to produce these proteins through activa-
Observations like these indicate that the phenotypes of high- tion of EMT programs. Thus, cancer cells may secrete the
grade malignancy do not arise in a strictly cell-autonomous chemoattractants that recruit the proinvasive inflammatory cells
manner, and that their manifestation cannot be understood rather than producing the matrix-degrading enzymes them-
solely through analyses of tumor cell genomes. One important selves.
implication, still untested, is that the ability to negotiate most of The Daunting Complexity of Metastatic Colonization
the steps of the invasion-metastasis cascade may be acquired Metastasis can be broken down into two major phases: the
in certain tumors without the requirement that the associated physical dissemination of cancer cells from the primary tumor
cancer cells undergo additional mutations beyond those that to distant tissues, and the adaptation of these cells to foreign
were needed for primary tumor formation. tissue microenvironments that results in successful colonization,
Plasticity in the Invasive Growth Program i.e., the growth of micrometastases into macroscopic tumors.
The role of contextual signals in inducing an invasive growth The multiple steps of dissemination would seem to be in the
capability (often via an EMT) implies the possibility of revers- purview of the EMT and similarly acting migratory programs.
ibility, in that cancer cells that have disseminated from a primary Colonization, however, is not strictly coupled with physical
tumor to a more distant tissue site may no longer benefit from the dissemination, as evidenced by the presence in many patients
activated stroma and invasion/EMT-inducing signals that they of myriad micrometastases that have successfully disseminated
experienced while residing in the primary tumor; in the absence but never progress to macroscopic metastatic tumors (Tal-
of ongoing exposure to these signals, carcinoma cells may revert madge and Fidler, 2010; McGowan et al., 2009; Aguirre-Ghiso,
in their new homes to a noninvasive state. Thus, carcinoma cells 2007; Townson and Chambers, 2006; Fidler, 2003).
that have undergone an EMT during initial invasion and meta- In some types of cancer, the primary tumor may release
static dissemination may pass through the reverse process, systemic suppressor factors that render such micrometastases
termed the mesenchymal-epithelial transition (MET). This plas- dormant, as revealed clinically by explosive metastatic growth
ticity may result in the formation of new tumor colonies of carci- soon after resection of the primary growth (Demicheli et al.,
noma cells exhibiting a histopathology similar to those of carci- 2008; Folkman, 2002). In others, however, such as breast cancer
noma cells in the primary tumor that never underwent an EMT and melanoma, macroscopic metastases may erupt decades
(Hugo et al., 2007). Moreover, the notion that cancer cells after a primary tumor has been surgically removed or pharmaco-
routinely pass through a complete EMT program is likely to be logically destroyed; these metastatic tumor growths evidently

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 655


reflect dormant micrometastases that have solved, after much trial that primary tumor cells entering the circulation are fortuitously
and error, the complex problem of tissue colonization (Barkan, endowed with the ability to colonize certain distant tissue sites
et al., 2010; Aguirre-Ghiso, 2007; Townson and Chambers, 2006). (Talmadge and Fidler, 2010). Alternatively, the ability to colonize
One can infer from such natural histories that micrometasta- specific tissues may only develop in response to the selective
ses may lack other hallmark capabilities necessary for vigorous pressure on already disseminated cancer cells to adapt to
growth, such as the ability to activate angiogenesis; indeed the growth in foreign tissue microenvironments.
inability of certain experimentally generated dormant microme- Having developed such tissue-specific colonizing ability, the
tastases to form macroscopic tumors has been ascribed to their cells in metastatic colonies may proceed to disseminate further,
failure to activate tumor angiogenesis (Naumov et al., 2008; not only to new sites in the body but also back to the primary
Aguirre-Ghiso, 2007). Additionally, recent experiments have tumors in which their ancestors arose. Accordingly, tissue-
shown that nutrient starvation can induce intense autophagy specific colonization programs that are evident among cells
that causes cancer cells to shrink and adopt a state of reversible within a primary tumor may originate not from classical tumor
dormancy; such cells may exit this state and resume active progression occurring within the primary lesion but instead
growth and proliferation when changes in tissue microenviron- from emigrants that have returned home (Kim et al., 2009).
ment, such as access to more nutrients, permit (Kenific et al., Such reseeding is consistent with the aforementioned studies
2010; Lu et al., 2008). Other mechanisms of micrometastatic of human pancreatic cancer metastasis (Campbell et al.,
dormancy may involve anti-growth signals embedded in normal 2010; Luebeck, 2010; Yachida et al., 2010). Stated differently,
tissue extracellular matrix (Barkan et al., 2010) and tumor-sup- the phenotypes and underlying gene expression programs of
pressing actions of the immune system (Teng et al., 2008; the populations of cancer cells (and of the cancer stem cells
Aguirre-Ghiso, 2007). discussed below) within primary tumors may be significantly
Most disseminated cancer cells are likely to be poorly adap- modified by reverse migration of their distant metastatic
ted, at least initially, to the microenvironment of the tissue in progeny.
which they have landed. Accordingly, each type of disseminated Implicit in this self-seeding process is another notion: the
cancer cell may need to develop its own set of ad hoc solutions supportive stroma that arises in a primary tumor and contributes
to the problem of thriving in the microenvironment of one or to its acquisition of malignant traits may intrinsically provide
another foreign tissue (Gupta et al., 2005). These adaptations a hospitable site for reseeding and colonization by circulating
might require hundreds of distinct colonization programs, each cancer cells emanating from metastatic lesions.
dictated by the type of disseminating cancer cell and the nature Clarifying the regulatory programs that enable metastatic
of the tissue microenvironment in which colonization is colonization represents an important agenda for future research.
proceeding. As further discussed below, however, certain tissue Substantial progress is being made, for example, in defining sets
microenviroments may be preordained to be intrinsically hospi- of genes (‘‘metastatic signatures’’) that correlate with and appear
table to disseminated cancer cells (Peinado et al., 2011; to facilitate the establishment of macroscopic metastases in
Talmadge and Fidler, 2010). specific tissues (Coghlin and Murray, 2010; Bos et al., 2009;
Metastatic dissemination has long been depicted as the last Olson et al., 2009; Nguyen et al., 2009; Gupta et al., 2005). The
step in multistep primary tumor progression, and indeed for challenge is considerable, given the apparent multitude of
many tumors that is likely the case, as illustrated by recent distinct colonization programs cited above. Moreover, coloniza-
genome sequencing studies that present genetic evidence for tion is unlikely to depend exclusively on cell-autonomous
clonal evolution of pancreatic ductal adenocarcinoma to metas- processes. Instead, it almost certainly requires the establish-
tasis (Campbell et al., 2010; Luebeck, 2010; Yachida et al., ment of a permissive tumor microenvironment composed of
2010). On the other hand, evidence has recently emerged critical stromal support cells. For these reasons, the process
indicating that cells can disseminate remarkably early, of colonization is likely to encompass a large number of cell-
dispersing from ostensibly noninvasive premalignant lesions in biological programs that are, in aggregate, considerably more
both mice and humans (Coghlin and Murray, 2010; Klein, complex and diverse than the preceding steps of metastatic
2009). Additionally, micrometastases can be spawned from dissemination.
primary tumors that are not obviously invasive but possess
a neovasculature lacking in lumenal integrity (Gerhardt and Programming of Hallmark Capabilities
Semb, 2008). Although cancer cells can clearly disseminate by Intracellular Circuitry
from such pre-neoplastic lesions and seed the bone marrow In 2000, we presented a metaphor, in which the numerous
and other tissues, their capability to colonize these sites and signaling molecules affecting cancer cells operate as nodes
develop into pathologically significant macrometastases and branches of elaborate integrated circuits that are reprog-
remains unproven. At present, we view this early metastatic rammed derivatives of the circuits operating in normal cells.
dissemination as a demonstrable phenomenon in mice and hu- The ensuing decade has both solidified the original depiction
mans whose clinical significance is yet to be established. of these circuits and expanded the catalog of signals and the
Beyond the timing of their dissemination, it also remains interconnections of their signaling pathways. It is difficult if not
unclear when and where cancer cells develop the ability to colo- impossible to graphically portray this circuit comprehensively
nize foreign tissues as macroscopic tumors. This capability may and coherently, as was already the case in 2000.
arise during primary tumor formation as a result of a tumor’s We now suggest a portrayal of this circuitry that is aligned with
particular developmental path prior to any dissemination, such individual hallmarks of cancer. Thus, the intracellular integrated

656 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


Figure 2. Intracellular Signaling Networks Regulate the Operations of the Cancer Cell
An elaborate integrated circuit operates within normal cells and is reprogrammed to regulate hallmark capabilities within cancer cells. Separate subcircuits,
depicted here in differently colored fields, are specialized to orchestrate the various capabilities. At one level, this depiction is simplistic, as there is considerable
crosstalk between such subcircuits. In addition, because each cancer cell is exposed to a complex mixture of signals from its microenvironment, each of these
subcircuits is connected with signals originating from other cells in the tumor microenvironment, as outlined in Figure 5.

circuit can be segmented into distinct subcircuits, each of which ENABLING CHARACTERISTICS AND EMERGING
is specialized to support a discrete cell-biological property in HALLMARKS
normal cells and is reprogrammed in order to implement
a hallmark capability in cancer cells (Figure 2). Only a subset of We have defined the hallmarks of cancer as acquired functional
hallmark capabilities are addressed in this figure, either because capabilities that allow cancer cells to survive, proliferate, and
their underlying control circuits remain poorly understood or disseminate; these functions are acquired in different tumor
because they overlap extensively with those portrayed here. types via distinct mechanisms and at various times during the
An additional dimension of complexity involves considerable course of multistep tumorigenesis. Their acquisition is made
interconnections and thus crosstalk between the individual sub- possible by two enabling characteristics. Most prominent is the
circuits. For example, certain oncogenic events can affect development of genomic instability in cancer cells, which
multiple capabilities, as illustrated by the diverse effects that generates random mutations including chromosomal rearrange-
prominent oncogenes, such as mutant RAS and upregulated ments; among these are the rare genetic changes that can
MYC, have on multiple hallmark capabilities (e.g., proliferative orchestrate hallmark capabilities. A second enabling character-
signaling, energy metabolism, angiogenesis, invasion, and istic involves the inflammatory state of premalignant and frankly
survival). We anticipate that future renditions of this integrated malignant lesions that is driven by cells of the immune system,
circuit will encompass subcircuits and associated hallmark some of which serve to promote tumor progression through
capabilities that are still not addressed here. various means.

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 657


Figure 3. Emerging Hallmarks and Enabling
Characteristics
An increasing body of research suggests that two
additional hallmarks of cancer are involved in the
pathogenesis of some and perhaps all cancers.
One involves the capability to modify, or repro-
gram, cellular metabolism in order to most effec-
tively support neoplastic proliferation. The second
allows cancer cells to evade immunological
destruction, in particular by T and B lymphocytes,
macrophages, and natural killer cells. Because
neither capability is yet generalized and fully vali-
dated, they are labeled as emerging hallmarks.
Additionally, two consequential characteristics of
neoplasia facilitate acquisition of both core and
emerging hallmarks. Genomic instability and thus
mutability endow cancer cells with genetic alter-
ations that drive tumor progression. Inflammation
by innate immune cells designed to fight infections
and heal wounds can instead result in their inad-
vertent support of multiple hallmark capabilities,
thereby manifesting the now widely appreciated
tumor-promoting consequences of inflammatory
responses.

Yet other distinct attributes of cancer cells have been The extraordinary ability of genome maintenance systems to
proposed to be functionally important for the development of detect and resolve defects in the DNA ensures that rates of
cancer and might therefore be added to the list of core hallmarks spontaneous mutation are usually very low during each cell
(Negrini et al., 2010; Luo et al., 2009; Colotta et al., 2009). Two generation. In the course of acquiring the roster of mutant genes
such attributes are particularly compelling. The first involves needed to orchestrate tumorigenesis, cancer cells often
major reprogramming of cellular energy metabolism in order to increase the rates of mutation (Negrini et al., 2010; Salk et al.,
support continuous cell growth and proliferation, replacing the 2010). This mutability is achieved through increased sensitivity
metabolic program that operates in most normal tissues and to mutagenic agents, through a breakdown in one or several
fuels the physiological operations of the associated cells. The components of the genomic maintenance machinery, or both.
second involves active evasion by cancer cells from attack and In addition, the accumulation of mutations can be accelerated
elimination by immune cells; this capability highlights the dichot- by compromising the surveillance systems that normally monitor
omous roles of an immune system that both antagonizes and genomic integrity and force genetically damaged cells into either
enhances tumor development and progression. Both of these senescence or apoptosis (Jackson and Bartek, 2009; Kastan,
capabilities may well prove to facilitate the development and 2008; Sigal and Rotter, 2000). The role of TP53 is central here,
progression of many forms of human cancer and therefore can leading to its being called the ‘‘guardian of the genome’’ (Lane,
be considered to be emerging hallmarks of cancer. These 1992).
enabling characteristics and emerging hallmarks, depicted in A diverse array of defects affecting various components of the
Figure 3, are discussed individually below. DNA-maintenance machinery—often referred to as the ‘‘care-
takers’’ of the genome (Kinzler and Vogelstein, 1997)—have
An Enabling Characteristic: Genome Instability been documented. The catalog of defects in these caretaker
and Mutation genes includes those whose products are involved in (1) detect-
Acquisition of the multiple hallmarks enumerated above depends ing DNA damage and activating the repair machinery, (2) directly
in large part on a succession of alterations in the genomes of repairing damaged DNA, and (3) inactivating or intercepting
neoplastic cells. Simply depicted, certain mutant genotypes mutagenic molecules before they have damaged the DNA
confer selective advantage on subclones of cells, enabling their (Negrini et al., 2010; Ciccia and Elledge, 2010; Jackson and
outgrowth and eventual dominance in a local tissue environment. Bartek, 2009; Kastan, 2008; Harper and Elledge, 2007; Friedberg
Accordingly, multistep tumor progression can be portrayed as et al., 2006). From a genetic perspective, these caretaker genes
a succession of clonal expansions, each of which is triggered behave much like tumor suppressor genes, in that their functions
by the chance acquisition of an enabling mutant genotype. can be lost during the course of tumor progression, with such
Because heritable phenotypes, e.g., inactivation of tumor losses being achieved either through inactivating mutations or
suppressor genes, can also be acquired through epigenetic via epigenetic repression. Mutant copies of many of these care-
mechanisms such as DNA methylation and histone modifications taker genes have been introduced into the mouse germline and
(Berdasco and Esteller, 2010; Esteller, 2007; Jones and Baylin, result, predictably, in increased cancer incidence, supporting
2007), some clonal expansions may well be triggered by nonmu- their potential involvement in human cancer development
tational changes affecting the regulation of gene expression. (Barnes and Lindahl, 2004).

658 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


In the decade since we first enumerated the cancer hallmarks, even by standard histochemical staining techniques (Pagès
another major source of tumor-associated genomic instability et al., 2010). Historically, such immune responses were largely
has been uncovered: as described earlier, the loss of telomeric thought to reflect an attempt by the immune system to eradicate
DNA in many tumors generates karyotypic instability and associ- tumors, and indeed, there is increasing evidence for antitumoral
ated amplification and deletion of chromosomal segments responses to many tumor types with an attendant pressure on
(Artandi and DePinho, 2010). When viewed in this light, telome- the tumor to evade immune destruction, as discussed below.
rase is more than an enabler of the hallmark capability for By 2000, there were already clues that the tumor-associated
unlimited replicative potential and must also be added to the inflammatory response had the unanticipated, paradoxical effect
list of critical caretakers responsible for maintaining genome of enhancing tumorigenesis and progression, in effect helping
integrity. incipient neoplasias to acquire hallmark capabilities. In the
Advances in the molecular-genetic analysis of cancer cell ensuing decade, research on the intersections between inflam-
genomes have provided the most compelling demonstrations mation and cancer pathogenesis has blossomed, producing
of function-altering mutations and of ongoing genomic instability abundant and compelling demonstrations of the functionally
during tumor progression. One type of analysis—comparative important tumor-promoting effects that immune cells—largely
genomic hybridization (CGH)—documents the gains and losses of the innate immune system—have on neoplastic progression
of gene copy number across the cell genome; in many tumors, (DeNardo et al., 2010; Grivennikov et al., 2010; Qian and Pollard,
the pervasive genomic aberrations revealed by CGH provide 2010; Colotta et al., 2009). Inflammation can contribute to
clear evidence for loss of control of genome integrity. Impor- multiple hallmark capabilities by supplying bioactive molecules
tantly, the recurrence of specific aberrations (both amplifications to the tumor microenvironment, including growth factors that
and deletions) at particular sites in the genome indicates that sustain proliferative signaling, survival factors that limit cell
such sites are likely to harbor genes whose alteration favors death, proangiogenic factors, extracellular matrix-modifying
neoplastic progression (Korkola and Gray, 2010). enzymes that facilitate angiogenesis, invasion, and metastasis,
More recently, with the advent of efficient and economical and inductive signals that lead to activation of EMT and other
DNA-sequencing technologies, higher-resolution analyses hallmark-facilitating programs (DeNardo et al., 2010;
have become possible. Early studies are revealing distinctive Grivennikov et al., 2010; Qian and Pollard, 2010; Karnoub and
patterns of DNA mutations in different tumor types (see http:// Weinberg, 2006–2007).
cancergenome.nih.gov/). In the not-too-distant future, the Importantly, inflammation is in some cases evident at the
sequencing of entire cancer cell genomes promises to clarify earliest stages of neoplastic progression and is demonstrably
the prevalence of ostensibly random mutations scattered across capable of fostering the development of incipient neoplasias
cancer cell genomes. Thus, recurring genetic alterations may into full-blown cancers (Qian and Pollard, 2010; de Visser et al.,
point to a causal role of particular mutations in tumor pathogen- 2006). Additionally, inflammatory cells can release chemicals,
esis. notably reactive oxygen species, that are actively mutagenic for
Although the specifics of genome alteration vary dramatically nearby cancer cells, accelerating their genetic evolution toward
between different tumor types, the large number of genome states of heightened malignancy (Grivennikov et al., 2010). As
maintenance and repair defects that have already been docu- such, inflammation can be considered an enabling characteristic
mented in human tumors, together with abundant evidence of for its contributions to the acquisition of core hallmark capabil-
widespread destabilization of gene copy number and nucleotide ities. The cells responsible for this enabling characteristic are
sequence, persuade us that instability of the genome is inherent described in the section below on the tumor microenvironment.
to the great majority of human cancer cells. This leads, in turn, to
the conclusion that the defects in genome maintenance and An Emerging Hallmark: Reprogramming Energy
repair are selectively advantageous and therefore instrumental Metabolism
for tumor progression, if only because they accelerate the rate The chronic and often uncontrolled cell proliferation that repre-
at which evolving premalignant cells can accumulate favorable sents the essence of neoplastic disease involves not only
genotypes. As such, genome instability is clearly an enabling deregulated control of cell proliferation but also corresponding
characteristic that is causally associated with the acquisition of adjustments of energy metabolism in order to fuel cell growth
hallmark capabilities. and division. Under aerobic conditions, normal cells process
glucose, first to pyruvate via glycolysis in the cytosol and there-
An Enabling Characteristic: Tumor-Promoting after to carbon dioxide in the mitochondria; under anaerobic
Inflammation conditions, glycolysis is favored and relatively little pyruvate is
Pathologists have long recognized that some tumors are densely dispatched to the oxygen-consuming mitochondria. Otto
infiltrated by cells of both the innate and adaptive arms of the Warburg first observed an anomalous characteristic of cancer
immune system and thereby mirror inflammatory conditions cell energy metabolism (Warburg, 1930, 1956a, 1956b): even in
arising in non-neoplastic tissues (Dvorak, 1986). With the advent the presence of oxygen, cancer cells can reprogram their
of better markers for accurately identifying the distinct cell types glucose metabolism, and thus their energy production, by
of the immune system, it is now clear that virtually every limiting their energy metabolism largely to glycolysis, leading to
neoplastic lesion contains immune cells present at densities a state that has been termed ‘‘aerobic glycolysis.’’
ranging from subtle infiltrations detectable only with cell type- The existence of this metabolic switch in cancer cells has been
specific antibodies to gross inflammations that are apparent substantiated in the ensuing decades. Such reprogramming of

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 659


energy metabolism is seemingly counterintuitive, in that cancer muscle (Kennedy and Dewhirst, 2010; Feron, 2009; Semenza,
cells must compensate for the 18-fold lower efficiency of 2008). Additionally, it is becoming apparent that oxygenation,
ATP production afforded by glycolysis relative to mitochondrial ranging from normoxia to hypoxia, is not necessarily static in
oxidative phosphorylation. They do so in part by upregulating tumors but instead fluctuates temporally and regionally (Hardee
glucose transporters, notably GLUT1, which substantially et al., 2009), likely as a result of the instability and chaotic organi-
increases glucose import into the cytoplasm (Jones and Thomp- zation of the tumor-associated neovasculature.
son, 2009; DeBerardinis et al., 2008; Hsu and Sabatini, 2008). Altered energy metabolism is proving to be as widespread in
Indeed, markedly increased uptake and utilization of glucose cancer cells as many of the other cancer-associated traits that
have been documented in many human tumor types, most have been accepted as hallmarks of cancer. This realization
readily by noninvasively visualizing glucose uptake using posi- raises the question of whether deregulating cellular energy
tron emission tomography (PET) with a radiolabeled analog of metabolism is therefore a core hallmark capability of cancer cells
glucose (18F-fluorodeoxyglucose, FDG) as a reporter. that is as fundamental as the six well-established core hallmarks.
Glycolytic fueling has been shown to be associated with In fact, the redirection of energy metabolism is largely orches-
activated oncogenes (e.g., RAS, MYC) and mutant tumor trated by proteins that are involved in one way or another in
suppressors (e.g., TP53) (DeBerardinis et al., 2008; Jones and programming the core hallmarks of cancer. When viewed in
Thompson, 2009), whose alterations in tumor cells have been this way, aerobic glycolysis is simply another phenotype that is
selected primarily for their benefits in conferring the hallmark programmed by proliferation-inducing oncogenes.
capabilities of cell proliferation, avoidance of cytostatic controls, Interestingly, activating (gain-of-function) mutations in the iso-
and attenuation of apoptosis. This reliance on glycolysis can be citrate dehydrogenase 1/2 (IDH) enzymes have been reported in
further accentuated under the hypoxic conditions that operate glioma and other human tumors (Yen et al., 2010). Although
within many tumors: the hypoxia response system acts pleio- these mutations may prove to have been clonally selected for
tropically to upregulate glucose transporters and multiple their ability to alter energy metabolism, there is confounding
enzymes of the glycolytic pathway (Semenza, 2010a; Jones data associating their activity with elevated oxidation and
and Thompson, 2009; DeBerardinis et al., 2008). Thus, both stability of the HIF-1 transcription factors (Reitman and Yan,
the Ras oncoprotein and hypoxia can independently increase 2010), which could in turn affect genome stability and angiogen-
the levels of the HIF1a and HIF2a transcription factors, which esis/invasion, respectively, thus blurring the lines of phenotypic
in turn upregulate glycolysis (Semenza, 2010a, 2010b; Kroemer demarcation. Currently, therefore, the designation of reprog-
and Pouyssegur, 2008). rammed energy metabolism as an emerging hallmark seems
A functional rationale for the glycolytic switch in cancer cells most appropriate, to highlight both its evident importance as
has been elusive, given the relatively poor efficiency of gener- well as the unresolved issues surrounding its functional indepen-
ating ATP by glycolysis relative to mitochondrial oxidative phos- dence from the core hallmarks.
phorylation. According to one long-forgotten (Potter, 1958) and
recently revived and refined hypothesis (Vander Heiden et al., An Emerging Hallmark: Evading Immune Destruction
2009), increased glycolysis allows the diversion of glycolytic A second, still-unresolved issue surrounding tumor formation
intermediates into various biosynthetic pathways, including involves the role that the immune system plays in resisting or
those generating nucleosides and amino acids; this facilitates, eradicating formation and progression of incipient neoplasias,
in turn, the biosynthesis of the macromolecules and organelles late-stage tumors, and micrometastases. The long-standing
required for assembling new cells. Moreover, Warburg-like theory of immune surveillance proposes that cells and tissues
metabolism seems to be present in many rapidly dividing em- are constantly monitored by an ever-alert immune system, and
bryonic tissues, once again suggesting a role in supporting the that such immune surveillance is responsible for recognizing
large-scale biosynthetic programs that are required for active and eliminating the vast majority of incipient cancer cells
cell proliferation. and thus nascent tumors. According to this logic, solid tumors
Interestingly, some tumors have been found to contain two that do appear have somehow managed to avoid detection
subpopulations of cancer cells that differ in their energy-gener- by the various arms of the immune system or have been able
ating pathways. One subpopulation consists of glucose-depen- to limit the extent of immunological killing, thereby evading
dent (‘‘Warburg-effect’’) cells that secrete lactate, whereas cells eradication.
of the second subpopulation preferentially import and utilize the The role of defective immunological monitoring of tumors
lactate produced by their neighbors as their main energy source, would seem to be validated by the striking increases of certain
employing part of the citric acid cycle to do so (Kennedy and Dew- cancers in immunocompromised individuals (Vajdic and van
hirst, 2010; Feron, 2009; Semenza, 2008). These two populations Leeuwen, 2009). However, the great majority of these are
evidently function symbiotically: the hypoxic cancer cells depend virus-induced cancers, suggesting that much of the control of
on glucose for fuel and secrete lactate as waste, which is im- this class of cancers normally depends on reducing viral burden
ported and preferentially used as fuel by their better-oxygenated in infected individuals, in part through eliminating virus-infected
brethren. Although this provocative mode of intratumoral symbi- cells. These observations, therefore, seem to shed little light
osis has yet to be generalized, the cooperation between lactate- on the possible role of the immune system in limiting formation
secreting and lactate-utilizing cells to fuel tumor growth is in fact of the >80% of tumors of nonviral etiology. In recent years,
not an invention of tumors but rather again reflects cooption of however, an increasing body of evidence, both from genetically
a normal physiological mechanism, in this case one operating in engineered mice and from clinical epidemiology, suggests that

660 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


the immune system operates as a significant barrier to tumor This might be taken as an argument against the importance of
formation and progression, at least in some forms of non-virus- immune surveillance as an effective barrier to tumorigenesis
induced cancer. and tumor progression. We note, however, that HIV and pharma-
When mice genetically engineered to be deficient for various cologically immunosuppressed patients are predominantly
components of the immune system were assessed for the devel- immunodeficient in the T and B cell compartments and thus do
opment of carcinogen-induced tumors, it was observed that not present with the multicomponent immunological deficiencies
tumors arose more frequently and/or grew more rapidly in the that have been produced in the genetically engineered mutant
immunodeficient mice relative to immunocompetent controls. mice lacking both NK cells and CTLs; this leaves open the possi-
In particular, deficiencies in the development or function of bility that such patients still have residual capability for an immu-
CD8+ cytotoxic T lymphocytes (CTLs), CD4+ Th1 helper T cells, nological defense against cancer that is mounted by NK and
or natural killer (NK) cells each led to demonstrable increases other innate immune cells.
in tumor incidence; moreover, mice with combined immunodefi- In truth, the above discussions of cancer immunology simplify
ciencies in both T cells and NK cells were even more susceptible tumor-host immunological interactions, as highly immunogenic
to cancer development. The results indicated that, at least in cancer cells may well evade immune destruction by disabling
certain experimental models, both the innate and adaptive components of the immune system that have been dispatched
cellular arms of the immune system are able to contribute signif- to eliminate them. For example, cancer cells may paralyze infil-
icantly to immune surveillance and thus tumor eradication (Teng trating CTLs and NK cells, by secreting TGF-b or other immuno-
et al., 2008; Kim et al., 2007). suppressive factors (Yang et al., 2010; Shields et al., 2010). More
In addition, transplantation experiments have shown that subtle mechanisms operate through the recruitment of inflam-
cancer cells that originally arose in immunodeficient mice are matory cells that are actively immunosuppressive, including
often inefficient at initiating secondary tumors in syngeneic regulatory T cells (Tregs) and myeloid-derived suppressor cells
immunocompetent hosts, whereas cancer cells from tumors (MDSCs). Both can suppress the actions of cytotoxic lympho-
arising in immunocompetent mice are equally efficient at initi- cytes (Mougiakakos et al., 2010; Ostrand-Rosenberg and Sinha,
ating transplanted tumors in both types of hosts (Teng et al., 2009).
2008; Kim et al., 2007). Such behavior has been interpreted as In light of these considerations and the still-rudimentary
follows: Highly immunogenic cancer cell clones are routinely demonstrations of antitumor immunity as a significant barrier
eliminated in immunocompetent hosts—a process that has to tumor formation and progression in humans, we present
been referred to as ‘‘immunoediting’’—leaving behind only immunoevasion as another emerging hallmark, whose gener-
weakly immunogenic variants to grow and generate solid ality as a core hallmark capability remains to be firmly estab-
tumors; such weakly immunogenic cells can thereafter colonize lished.
both immunodeficient and immunocompetent hosts. Con-
versely, when arising in immunodeficient hosts, the immuno- THE TUMOR MICROENVIRONMENT
genic cancer cells are not selectively depleted and can, instead,
prosper along with their weakly immunogenic counterparts. Over the past decade, tumors have increasingly been recog-
When cells from such nonedited tumors are serially transplanted nized as organs whose complexity approaches and may even
into syngeneic recipients, the immunogenic cancer cells are exceed that of normal healthy tissues. When viewed from this
rejected when they confront, for the first time, the competent perspective, the biology of a tumor can only be understood
immune systems of their secondary hosts (Smyth et al., 2006). by studying the individual specialized cell types within it
(Unanswered in these particular experiments is the question of (Figure 4, upper) as well as the ‘‘tumor microenvironment’’
whether the chemical carcinogens used to induce such tumors that they construct during the course of multistep tumorigenesis
are prone to generate cancer cells that are especially immuno- (Figure 4, lower). This depiction contrasts starkly with the
genic.) earlier, reductionist view of a tumor as nothing more than
Clinical epidemiology also increasingly supports the existence a collection of relatively homogeneous cancer cells, whose
of antitumoral immune responses in some forms of human entire biology could be understood by elucidating the cell-
cancer (Bindea et al., 2010; Ferrone and Dranoff, 2010; Nelson, autonomous properties of these cells. We enumerate here
2008). For example, patients with colon and ovarian tumors a set of cell types known to contribute in important ways to
that are heavily infiltrated with CTLs and NK cells have a better the biology of many tumors and discuss the regulatory signaling
prognosis than those that lack such abundant killer lymphocytes that controls their individual and collective functions. Most of
(Pagès et al., 2010; Nelson, 2008); the case for other cancers is these observations stem from the study of carcinomas, in which
suggestive but less compelling and is the subject of ongoing the neoplastic epithelial cells constitute a compartment (the
investigation. Additionally, some immunosuppressed organ parenchyma) that is clearly distinct from the mesenchymal cells
transplant recipients have been observed to develop donor- forming the tumor-associated stroma.
derived cancers, suggesting that in the ostensibly tumor-free
donors, the cancer cells were held in check, in a dormant state, Cancer Cells and Cancer Stem Cells
by a fully functional immune system (Strauss and Thomas, 2010). Cancer cells are the foundation of the disease; they initiate
Still, the epidemiology of chronically immunosuppressed tumors and drive tumor progression forward, carrying the
patients does not indicate significantly increased incidences of oncogenic and tumor suppressor mutations that define cancer
the major forms of nonviral human cancer, as noted above. as a genetic disease. Traditionally, the cancer cells within tumors

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 661


Figure 4. The Cells of the Tumor Microenviron-
ment
(Upper) An assemblage of distinct cell types constitutes
most solid tumors. Both the parenchyma and stroma of
tumors contain distinct cell types and subtypes that
collectively enable tumor growth and progression.
Notably, the immune inflammatory cells present in tumors
can include both tumor-promoting as well as tumor-killing
subclasses.
(Lower) The distinctive microenvironments of tumors. The
multiple stromal cell types create a succession of tumor
microenvironments that change as tumors invade normal
tissue and thereafter seed and colonize distant tissues.
The abundance, histologic organization, and phenotypic
characteristics of the stromal cell types, as well as of the
extracellular matrix (hatched background), evolve during
progression, thereby enabling primary, invasive, and then
metastatic growth. The surrounding normal cells of the
primary and metastatic sites, shown only schematically,
likely also affect the character of the various neoplastic
microenvironments. (Not shown are the premalignant
stages in tumorigenesis, which also have distinctive
microenvironments that are created by the abundance
and characteristics of the assembled cells.)

often-rare tumor-initiating cells proved to share


transcriptional profiles with certain normal
tissue stem cell populations, motivating their
designation as stem-like.
The origins of CSCs within a solid tumor have
not been clarified and indeed may well vary from
have been portrayed as reasonably homogeneous cell popula- one tumor type to another. In some tumors, normal tissue stem
tions until relatively late in the course of tumor progression, cells may serve as the cells-of-origin that undergo oncogenic
when hyperproliferation combined with increased genetic transformation to yield CSCs; in others, partially differentiated
instability spawn distinct clonal subpopulations. Reflecting transit-amplifying cells, also termed progenitor cells, may suffer
such clonal heterogeneity, many human tumors are histopatho- the initial oncogenic transformation thereafter assuming more
logically diverse, containing regions demarcated by various stem-like character. Once primary tumors have formed, the
degrees of differentiation, proliferation, vascularity, inflamma- CSCs, like their normal counterparts, may self-renew as well
tion, and/or invasiveness. In recent years, however, evidence as spawn more differentiated derivatives; in the case of
has accumulated pointing to the existence of a new dimension neoplastic CSCs, these descendant cells form the great bulk of
of intratumor heterogeneity and a hitherto-unappreciated many tumors. It remains to be established whether multiple
subclass of neoplastic cells within tumors, termed cancer stem distinct classes of increasingly neoplastic stem cells form during
cells (CSCs). inception and subsequent multistep progression of tumors, ulti-
Although the evidence is still fragmentary, CSCs may prove to mately yielding the CSCs that have been described in fully devel-
be a common constituent of many if not most tumors, albeit oped cancers.
being present with widely varying abundance. CSCs are defined Recent research has interrelated the acquisition of CSC traits
operationally through their ability to efficiently seed new tumors with the EMT transdifferentiation program discussed above
upon inoculation into recipient host mice (Cho and Clarke, 2008; (Singh and Settleman, 2010; Mani et al., 2008; Morel et al.,
Lobo et al., 2007). This functional definition is often comple- 2008). Induction of this program in certain model systems can
mented by including the expression in CSCs of markers that induce many of the defining features of stem cells, including
are also expressed by the normal stem cells in the tissue-of- self-renewal ability and the antigenic phenotypes associated
origin (Al-Hajj et al., 2003). with both normal and cancer stem cells. This concordance
CSCs were initially implicated in the pathogenesis of hemato- suggests that the EMT program not only may enable cancer cells
poietic malignancies (Reya et al., 2001; Bonnet and Dick, 1997) to physically disseminate from primary tumors but also can
and then years later were identified in solid tumors, in particular confer on such cells the self-renewal capability that is crucial
breast carcinomas and neuroectodermal tumors (Gilbertson and to their subsequent clonal expansion at sites of dissemination
Rich, 2007; Al-Hajj et al., 2003). Fractionation of cancer cells on (Brabletz et al., 2005). If generalized, this connection raises an
the basis of displayed cell-surface markers has yielded subpop- important corollary hypothesis: the heterotypic signals that
ulations of neoplastic cells with a greatly enhanced ability, rela- trigger an EMT, such as those released by an activated, inflam-
tive to the corresponding majority populations, to seed new matory stroma, may also be important in creating and maintain-
tumors upon implantation in immunodeficient mice. These ing CSCs.

662 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


An increasing number of human tumors are reported to tumor progression may drive rampant genetic diversification
contain subpopulations with the properties of CSCs, as defined that outpaces the process of Darwinian selection, generating
operationally through their efficient tumor-initiating capabilities genetically distinct subpopulations far more rapidly than they
upon xenotransplantation into mice. Nevertheless, the im- can be eliminated.
portance of CSCs as a distinct phenotypic subclass of Such thinking is increasingly supported by in-depth sequence
neoplastic cells remains a matter of debate, as does their oft- analysis of tumor cell genomes, which has become practical due
cited rarity within tumors (Boiko et al., 2010; Gupta et al., 2009; to recent major advances in DNA (and RNA) sequencing tech-
Quintana et al., 2008). Indeed, it is plausible that the phenotypic nology. Thus the sequencing of the genomes of cancer cells
plasticity operating within tumors may produce bidirectional microdissected from different sectors of the same tumor
interconversion between CSCs and non-CSCs, resulting in (Yachida et al., 2010) has revealed striking intratumoral genetic
dynamic variation in the relative abundance of CSCs. Such heterogeneity. Some of this genetic diversity may be reflected
plasticity could complicate definitive measurement of their prev- in the long-recognized histological heterogeneity within indi-
alence. Analogous plasticity is already implicated in the EMT vidual human tumors. Alternatively, this genetic diversification
program, which can be engaged reversibly (Thiery and Sleeman, may enable functional specialization, producing subpopulations
2006). of cancer cells that contribute distinct, complementary capabil-
These complexities notwithstanding, it is evident that this ities, which then accrue to the common benefit of overall tumor
new dimension of tumor heterogeneity holds important implica- growth as described above.
tions for successful cancer therapies. Increasing evidence in
a variety of tumor types suggests that cells with properties of Endothelial Cells
CSCs are more resistant to various commonly used chemother- Much of the cellular heterogeneity within tumors is found in
apeutic treatments (Singh and Settleman, 2010; Creighton their stromal compartments. Prominent among the stromal
et al., 2009; Buck et al., 2007). Their persistence may help to constituents are the cells forming the tumor-associated vascu-
explain the almost-inevitable disease recurrence following lature. Mechanisms of development, differentiation, and
apparently successful debulking of human solid tumors by radi- homeostasis of endothelial cells composing the arteries, veins,
ation and various forms of chemotherapy. Indeed, CSCs may and capillaries were already well understood in 2000. So too
well prove to underlie certain forms of tumor dormancy, was the concept of the ‘‘angiogenic switch,’’ which activates
whereby latent cancer cells persist for years or even decades quiescent endothelial cells, causing them to enter into a cell-
after surgical resection or radio/chemotherapy, only to biological program that allows them to construct new blood
suddenly erupt and generate life-threatening disease. Hence, vessels (see above). Over the last decade, a network of inter-
CSCs may represent a double-threat, in that they are more connected signaling pathways involving ligands of signal-trans-
resistant to therapeutic killing and, at the same time, endowed ducing receptors displayed by endothelial cells (e.g., Notch,
with the ability to regenerate a tumor once therapy has been Neuropilin, Robo, and Eph-A/B) has been added to the
halted. already-prominent VEGF, angiopoietin, and FGF signals. These
This phenotypic plasticity implicit in CSC state may also newly characterized pathways have been functionally impli-
enable the formation of functionally distinct subpopulations cated in developmental and tumor-associated angiogenesis
within a tumor that support overall tumor growth in various and illustrate the complex regulation of endothelial cell pheno-
ways. For example, an EMT can convert epithelial carcinoma types (Pasquale, 2010; Ahmed and Bicknell, 2009; Dejana
cells into mesenchymal, fibroblast-like cancer cells that may et al., 2009; Carmeliet and Jain, 2000).
well assume the duties of cancer-associated fibroblasts (CAFs) Other avenues of research are revealing distinctive gene
in some tumors. Remarkably, several recent reports have expression profiles of tumor-associated endothelial cells and
documented the ability of glioblastoma cells (or possibly their identifying cell-surface markers displayed on the lumenal
associated CSC subpopulations) to transdifferentiate into endo- surfaces of normal versus tumor endothelial cells (Nagy et al.,
thelial-like cells that can substitute for bona fide host-derived 2010; Ruoslahti et al., 2010; Ruoslahti, 2002). Differences in
endothelial cells in forming a tumor-associated neovasculature signaling, in transcriptome profiles, and in vascular ‘‘ZIP codes’’
(Soda et al., 2011; El Hallani et al., 2010; Ricci-Vitiani et al., will likely prove to be important for understanding the conversion
2010; Wang et al., 2010). Observations like these indicate that of normal endothelial cells into tumor-associated endothelial
certain tumors may acquire stromal support by inducing some cells. Such knowledge may lead, in turn, to opportunities to
of their own cancer cells to undergo various types of metamor- develop novel therapies that exploit these differences in order
phosis to produce stromal cell types rather than relying on to selectively target tumor-associated endothelial cells.
recruited host cells to provide their functions. Closely related to the endothelial cells of the general circula-
The discovery of CSCs and biological plasticity in tumors tion are those forming lymphatic vessels (Tammela and Alitalo,
indicates that a single, genetically homogeneous population of 2010). Their role in the tumor-associated stroma, specifically in
cells within a tumor may nevertheless be phenotypically hetero- supporting tumor growth, is poorly understood. Indeed, because
geneous due to the presence of cells in distinct states of differ- of high interstitial pressure within solid tumors, intratumoral
entiation. However, an equally important source of phenotypic lymphatic vessels are typically collapsed and nonfunctional; in
variability may derive from the genetic heterogeneity within contrast, however, there are often functional, actively growing
a tumor that accumulates as cancer progression proceeds. (‘‘lymphangiogenic’’) lymphatic vessels at the peripheries of
Thus, elevated genetic instability operating in later stages of tumors and in the adjacent normal tissues that cancer cells

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 663


invade. These associated lymphatics likely serve as channels for including fibrosis, aberrant angiogenesis, and neoplasia (Griven-
the seeding of metastases in the draining lymph nodes that are nikov et al., 2010; Karin et al., 2006).
commonly observed in a number of cancer types. Over the past decade, the manipulation of genes involved in
the determination or effector functions of various immune cell
Pericytes types, together with pharmacological inhibitors of such cells or
As noted earlier, pericytes represent a specialized mesenchymal their functions, has shown them to play diverse and critical roles
cell type (related to smooth muscle cells) with finger-like projec- in fostering tumorigenesis. The roster of tumor-promoting
tions that wrap around the endothelial tubing of blood vessels. In inflammatory cells now includes macrophage subtypes, mast
normal tissues, pericytes are known to provide paracrine cells, and neutrophils, as well as T and B lymphocytes (Coffelt
support signals to the normally quiescent endothelium. For et al., 2010; DeNardo et al., 2010; Egeblad et al., 2010; Johans-
example, Ang-1 secreted by pericytes conveys antiproliferative son et al., 2008; Murdoch et al., 2008; DePalma et al., 2007).
stabilizing signals that are received by the Tie2 receptors Such studies are yielding a growing list of signaling molecules
expressed on the surface of endothelial cells; some pericytes released by inflammatory cells that serve as effectors of their
also produce low levels of VEGF that serve a trophic function tumor-promoting actions. These include the tumor growth factor
in endothelial homeostasis (Gaengel et al., 2009; Bergers and EGF, the angiogenic growth factor VEGF, other proangiogenic
Song, 2005). Pericytes also collaborate with the endothelial cells factors such as FGF2, chemokines, and cytokines that amplify
to synthesize the vascular basement membrane that anchors the inflammatory state; in addition, these cells may produce
both pericytes and endothelial cells and helps vessel walls to proangiogenic and/or proinvasive matrix-degrading enzymes,
withstand the hydrostatic pressure of blood flow. including MMP-9 and other matrix metalloproteinases, cysteine
Genetic and pharmacological perturbation of the recruitment cathepsin proteases, and heparanase (Qian and Pollard, 2010;
and association of pericytes has demonstrated the functional Murdoch et al., 2008). Consistent with their expression of these
importance of these cells in supporting the tumor endothelium diverse effectors, tumor-infiltrating inflammatory cells have been
(Pietras and Ostman, 2010; Gaengel et al., 2009; Bergers and shown to induce and help sustain tumor angiogenesis, to stimu-
Song, 2005). For example, pharmacological inhibition of late cancer cell proliferation, to facilitate, via their presence at the
signaling through the PDGF receptor expressed by tumor peri- margins of tumors, tissue invasion, and to support the metastatic
cytes and bone marrow-derived pericyte progenitors results in dissemination and seeding of cancer cells (Coffelt et al., 2010;
reduced pericyte coverage of tumor vessels, which in turn desta- Egeblad et al., 2010; Qian and Pollard, 2010; Mantovani, 2010;
bilizes vascular integrity and function (Pietras and Ostman, 2010; Joyce and Pollard, 2009; Mantovani et al., 2008; Murdoch
Raza et al., 2010; Gaengel et al., 2009); interestingly, and in et al., 2008; DePalma et al., 2007).
contrast, the pericytes of normal vessels are not prone to such In addition to fully differentiated immune cells present in tumor
pharmacological disruption, providing another example of the stroma, a variety of partially differentiated myeloid progenitors
differences in regulation of normal quiescent and tumor vascula- have been identified in tumors (Murdoch et al., 2008). Such cells
ture. An intriguing hypothesis, still to be fully substantiated, is represent intermediaries between circulating cells of bone
that tumors with poor pericyte coverage of their vasculature marrow origin and the differentiated immune cells typically found
may be more prone to permit cancer cell intravasation into the in normal and inflamed tissues. Importantly, these progenitors,
circulatory system, enabling subsequent hematogenous like their more differentiated derivatives, have demonstrable
dissemination (Raza et al., 2010; Gerhardt and Semb, 2008). tumor-promoting activity. Of particular interest, a class of
tumor-infiltrating myeloid cells (defined as coexpressing the
Immune Inflammatory Cells macrophage marker CD11b and the neutrophil marker Gr1)
As also discussed above, infiltrating cells of the immune system has been shown to suppress CTL and NK cell activity, having
are increasingly accepted to be generic constituents of tumors. been independently identified as MDSCs (Qian and Pollard,
These inflammatory cells operate in conflicting ways: both 2010; Ostrand-Rosenberg and Sinha, 2009). This attribute raises
tumor-antagonizing and tumor-promoting leukocytes can be the possibility that recruitment of certain myeloid cells may be
found, in various proportions, in most if not all neoplastic lesions. doubly beneficial for the developing tumor, by directly promoting
Although the presence of tumor-antagonizing CTLs and NK cells angiogenesis and tumor progression while at the same time
is not surprising, the prevalence of immune cells that functionally affording a means to evade immune destruction.
enhance hallmark capabilities was largely unanticipated. The counterintuitive existence of both tumor-promoting and
Evidence began to accumulate in the late 1990s that the infiltra- tumor-antagonizing immune cells can be rationalized by
tion of neoplastic tissues by cells of the immune system serves, invoking the diverse roles of the immune system: On the one
perhaps counterintuitively, to promote tumor progression. Such hand, the immune system specifically detects and targets infec-
work traced its conceptual roots back to the association of sites tious agents with the adaptive immune response, which is sup-
of chronic inflammation with tumor formation, and to the obser- ported by cells of the innate immune system. On the other, the
vation that tumors could be portrayed as wounds that never heal innate immune system is involved in wound healing and clearing
(Schäfer and Werner, 2008: Dvorak, 1986). In the course of dead cells and cellular debris. These specialized tasks are
normal wound healing and fighting infections, immune inflamma- accomplished by distinct subclasses of inflammatory cells,
tory cells appear transiently and then disappear, in contrast to namely a class of conventional macrophages and neutrophils
their persistence in sites of chronic inflammation, where their (engaged in supporting adaptive immunity), and subclasses of
presence has been associated with various tissue pathologies, ‘‘alternatively activated’’ macrophages, neutrophils, and

664 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


myeloid progenitors that are engaged in wound healing and years, the bone marrow has increasingly been implicated as
tissue housecleaning (Egeblad et al., 2010; Mantovani, 2010; a key source of tumor-associated stromal cells (Bergfeld and
Qian and Pollard, 2010; Johansson et al., 2008). The latter DeClerck, 2010; Fang and Salven, 2011; Giaccia and Schipani,
subtypes of immune cells are one of the major sources of the 2010; Patenaude et al., 2010; Lamagna and Bergers, 2006).
angiogenic, epithelial, and stromal growth factors and matrix-re- Mesenchymal stem and progenitor cells have been found to
modeling enzymes that are needed for wound healing, and it is transit into tumors from the marrow, where they may differentiate
these cells that are recruited and subverted to support into the various well-characterized stromal cell types. Some of
neoplastic progression. Similarly, subclasses of B and T these recent arrivals may also persist in an undifferentiated or
lymphocytes may facilitate the recruitment, activation, and partially differentiated state, exhibiting functions that their more
persistence of such wound-healing and tumor-promoting differentiated progeny lack.
macrophages and neutrophils (DeNardo et al., 2010; Egeblad The bone marrow origins of stromal cell types have
et al., 2010; Biswas and Mantovani, 2010). Of course, other been demonstrated using tumor-bearing mice in which the
subclasses of B and T lymphocytes and innate immune cell bone marrow cells and thus their disseminated progeny have
types can mount demonstrable tumor-killing responses. The been selectively labeled with reporters such as green fluorescent
balance between the conflicting inflammatory responses in protein (GFP). While immune inflammatory cells have been long
tumors is likely to prove instrumental in prognosis and, quite known to derive from the bone marrow, more recently the
possibly, in therapies designed to redirect these cells toward progenitors of pericytes and of various subtypes of cancer-asso-
tumor destruction. ciated fibroblasts originating from the bone marrow have been
described in various mouse models of cancer (Bergfeld and
Cancer-Associated Fibroblasts DeClerck, 2010; Fang and Salven, 2011; Giaccia and Schipani,
Fibroblasts are found in various proportions across the spectrum 2010; Lamagna and Bergers, 2006); the prevalence and func-
of carcinomas, constituting in many cases the preponderant cell tional importance of endothelial progenitors for tumor angiogen-
population of the tumor stroma. The term ‘‘cancer-associated esis is currently unresolved (Fang and Salven, 2011; Patenaude
fibroblast’’ subsumes at least two distinct cell types: (1) cells et al., 2010). Taken together, these various lines of evidence indi-
with similarities to the fibroblasts that create the structural foun- cate that tumor-associated stromal cells may be supplied to
dation supporting most normal epithelial tissues and (2) myofi- growing tumors by proliferation of preexisting stromal cells, by
broblasts, whose biological roles and properties differ markedly differentiation in situ of local stem/progenitor cells originating
from those of tissue-derived fibroblasts. Myofibroblasts are in the neighboring normal tissue, or via recruitment of bone
identifiable by their expression of a-smooth muscle actin marrow-derived stem/progenitor cells.
(SMA). They are rare in most healthy epithelial tissues, although
certain tissues, such as the liver and pancreas, contain appre- Heterotypic Signaling Orchestrates
ciable numbers of a-SMA-expressing cells. Myofibroblasts tran- the Cells of the Tumor Microenvironment
siently increase in abundance in wounds and are also found in Depictions of the intracellular circuitry governing cancer cell
sites of chronic inflammation. Although beneficial to tissue biology (e.g., Figure 2) will need to be complemented by similar
repair, myofibroblasts are problematic in chronic inflammation, diagrams charting the complex interactions between the
contributing to the pathological fibrosis observed in tissues neoplastic and stromal cells within a tumor and the dynamic
such as lung, kidney, and liver. extracellular matrix that they collectively erect and remodel (Ege-
Recruited myofibroblasts and reprogrammed variants of blad et al., 2010; Kessenbrock et al., 2010; Pietras and Ostman,
normal tissue-derived fibroblastic cells have been demonstrated 2010; Polyak et al., 2009). A reasonably complete, graphic
to enhance tumor phenotypes, notably cancer cell proliferation, depiction of the network of microenvironmental signaling inter-
angiogenesis, and invasion and metastasis; their tumor- actions is still far beyond our reach, as the great majority of
promoting activities have largely been defined by transplantation signaling molecules and pathways remain to be identified. We
of cancer-associated fibroblasts admixed with cancer cells into provide instead a hint of such interactions in Figure 5, upper.
mice, and more recently by genetic and pharmacologic pertur- These few well-established examples are intended to exemplify
bation of their functions in tumor-prone mice (Dirat et al., 2010; a signaling network of remarkable complexity that is of critical
Pietras and Ostman, 2010; Räsänen and Vaheri, 2010; Shimoda importance to tumor pathogenesis.
et al., 2010; Kalluri and Zeisberg, 2006; Bhowmick et al., 2004). Another dimension of complexity is not represented in this
Because they secrete a variety of extracellular matrix compo- simple schematic: both neoplastic cells and the stromal cells
nents, cancer-associated fibroblasts are implicated in the forma- around them change progressively during the multistep transfor-
tion of the desmoplastic stroma that characterizes many mation of normal tissues into high-grade malignancies. This
advanced carcinomas. The full spectrum of functions contrib- histopathological progression must reflect underlying changes
uted by both subtypes of cancer-associated fibroblasts to tumor in heterotypic signaling between tumor parenchyma and stroma.
pathogenesis remains to be elucidated. Such stepwise progression is likely to depend on back-and-
forth reciprocal interactions between the neoplastic cells and
Stem and Progenitor Cells of the Tumor Stroma the supporting stromal cells, as depicted in Figure 5, lower.
The various stromal cell types that constitute the tumor microen- Thus, incipient neoplasias begin the interplay by recruiting and
vironment may be recruited from adjacent normal tissue—the activating stromal cell types that assemble into an initial preneo-
most obvious reservoir of such cell types. However, in recent plastic stroma, which in turn responds reciprocally by enhancing

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 665


Figure 5. Signaling Interactions in the Tumor Microenvironment during Malignant Progression
(Upper) The assembly and collective contributions of the assorted cell types constituting the tumor microenvironment are orchestrated and maintained by
reciprocal heterotypic signaling interactions, of which only a few are illustrated.
(Lower) The intracellular signaling depicted in the upper panel within the tumor microenvironment is not static but instead changes during tumor progression as
a result of reciprocal signaling interactions between cancer cells of the parenchyma and stromal cells that convey the increasingly aggressive phenotypes that
underlie growth, invasion, and metastatic dissemination. Importantly, the predisposition to spawn metastatic lesions can begin early, being influenced by the
differentiation program of the normal cell-of-origin or by initiating oncogenic lesions. Certain organ sites (sometimes referred to as ‘‘fertile soil’’ or ‘‘metastatic
niches’’) can be especially permissive for metastatic seeding and colonization by certain types of cancer cells, as a consequence of local properties that are either
intrinsic to the normal tissue or induced at a distance by systemic actions of primary tumors. Cancer stem cells may be variably involved in some or all of the
different stages of primary tumorigenesis and metastasis.

666 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


the neoplastic phenotypes of the nearby cancer cells. The The rapidly growing armamentarium of targeted therapeutics
cancer cells, which may further evolve genetically, again feed can be categorized according to their respective effects on
signals back to the stroma, continuing the reprogramming of one or more hallmark capabilities, as illustrated in the examples
normal stromal cells to serve the budding neoplasm; ultimately presented in Figure 6. Indeed, the observed efficacy of these
signals originating in the tumor stroma enable cancer cells to drugs represents, in each case, a validation of a particular capa-
invade normal adjacent tissues and disseminate. bility: if a capability is truly important for the biology of tumors,
This model of reciprocal heterotypic signaling must be then its inhibition should impair tumor growth and progression.
extended to encompass the final stage of multistep tumor We note that most of the hallmark-targeting cancer drugs
progression—metastasis (Figure 5, lower right). The circulating developed to date have been deliberately directed toward
cancer cells that are released from primary tumors leave a micro- specific molecular targets that are involved in one way or another
environment created by the supportive stroma of such tumors. in enabling particular capabilities. Such specificity of action has
However, upon landing in a distant organ, these cancer cells been considered a virtue, as it presents inhibitory activity against
encounter a naive, fully normal, tissue microenvironment. a target while having, in principle, relatively fewer off-target
Consequently, many of the heterotypic signals that shaped their effects and thus less nonspecific toxicity. In fact, resulting clin-
phenotype while they resided within primary tumors may be ical responses have generally been transitory, being followed
absent in sites of dissemination, constituting a barrier to growth by almost-inevitable relapses.
of the seeded cancer cells. Thus, the succession of reciprocal One interpretation of this history, supported by growing exper-
cancer cell to stromal cell interactions that defined multistep imental evidence, is that each of the core hallmark capabilities is
progression in the primary tumor now must be repeated anew regulated by partially redundant signaling pathways. Conse-
in distant tissues as disseminated cancer cells proceed to colo- quently, a targeted therapeutic agent inhibiting one key pathway
nize their newfound organ sites. in a tumor may not completely shut off a hallmark capability, al-
Although this logic applies in some cases of metastasis, in lowing some cancer cells to survive with residual function until
others, as mentioned earlier, certain tissue microenvironments they or their progeny eventually adapt to the selective pressure
may, for various reasons, already be supportive of freshly imposed by the therapy being applied. Such adaptation, which
seeded cancer cells; such permissive sites have been referred can be accomplished by mutation, epigenetic reprogramming,
to as ‘‘metastatic niches’’ (Peinado et al., 2011; Coghlin and or remodeling of the stromal microenvironment, can reestablish
Murray, 2010). Implicit in this term is the notion that cancer cells the functional capability, permitting renewed tumor growth and
seeded in such sites may not need to begin by inducing clinical relapse. Given that the number of parallel signaling path-
a supportive stroma because it already preexists, at least in ways supporting a given hallmark must be limited, it may
part. Such permissivity may be intrinsic to the tissue site become possible to target all of these supporting pathways ther-
(Talmadge and Fidler, 2010) or preinduced by circulating factors apeutically, thereby preventing the development of adaptive
released by the primary tumor (Peinado et al., 2011). The most resistance.
well-documented components of induced premetastatic niches In response to therapy, cancer cells may also reduce their
are tumor-promoting inflammatory cells, although other cell dependence on a particular hallmark capability, becoming
types and the ECM may well prove to play important roles in more dependent on another; this represents a quite different
different metastatic contexts. form of acquired drug resistance. This concept is exemplified
The likelihood that signaling interactions between cancer cells by recent discoveries of unexpected responses to antiangio-
and their supporting stroma evolve during the course of multi- genic therapies. Some have anticipated that effective inhibition
stage tumor development clearly complicates the goal of fully of angiogenesis would render tumors dormant and might even
elucidating the mechanisms of cancer pathogenesis. For lead to their dissolution (Folkman and Kalluri, 2004). Instead,
example, this reality poses challenges to systems biologists the clinical responses to antiangiogenic therapies have been
seeking to chart the crucial regulatory networks than orchestrate found to be transitory (Azam et al., 2010; Ebos et al., 2009; Berg-
malignant progression. Moreover, it seems likely that under- ers and Hanahan, 2008).
standing these dynamic variations will become crucial to the In certain preclinical models, where potent angiogenesis inhib-
development of novel therapies designed to successfully target itors succeed in suppressing this hallmark capability, tumors
both primary and metastatic tumors. adapt and shift from a dependence upon continuing angiogen-
esis to heightening the activity of another instead—invasiveness
THERAPEUTIC TARGETING and metastasis (Azam et al., 2010: Ebos et al., 2009; Bergers and
Hanahan, 2008). By invading nearby tissues, initially hypoxic
The introduction of mechanism-based targeted therapies to cancer cells evidently gain access to normal, preexisting tissue
treat human cancers has been heralded as one of the fruits of vasculature. Initial clinical validation of this adaptive/evasive
three decades of remarkable progress of research into the resistance is apparent in the increased invasion and local metas-
mechanisms of cancer pathogenesis. We do not attempt here tasis seen when human glioblastomas are treated with antian-
to enumerate the myriad therapies that are under development giogenic therapies (Ellis and Reardon, 2009; Norden et al.,
or have been introduced of late into the clinic. Instead, we 2009; Verhoeff et al., 2009). The applicability of this lesson to
consider how the description of hallmark principles is beginning other human cancers has yet to be established.
to inform therapeutic development at present and may increas- Analogous adaptive shifts in dependence on other hallmark
ingly do so in the future. traits may also limit efficacy of analogous hallmark-targeting

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 667


Figure 6. Therapeutic Targeting of the Hallmarks of Cancer
Drugs that interfere with each of the acquired capabilities necessary for tumor growth and progression have been developed and are in clinical trials or in some
cases approved for clinical use in treating certain forms of human cancer. Additionally, the investigational drugs are being developed to target each of the
enabling characteristics and emerging hallmarks depicted in Figure 3, which also hold promise as cancer therapeutics. The drugs listed are but illustrative
examples; there is a deep pipeline of candidate drugs with different molecular targets and modes of action in development for most of these hallmarks.

therapies. For example, the deployment of apoptosis-inducing The six acquired capabilities—the hallmarks of cancer—have
drugs may induce cancer cells to hyperactivate mitogenic stood the test of time as being integral components of most
signaling, enabling them to compensate for the initial attrition forms of cancer. Further refinement of these organizing princi-
triggered by such treatments. Such considerations suggest ples will surely come in the foreseeable future, continuing the
that drug development and the design of treatment protocols remarkable conceptual progress of the last decade.
will benefit from incorporating the concepts of functionally Looking ahead, we envision significant advances during the
discrete hallmark capabilities and of the multiple biochemical coming decade in our understanding of invasion and metastasis.
pathways involved in supporting each of them. Thus, in partic- Similarly, the role of aerobic glycolysis in malignant growth will
ular, we can envisage that selective cotargeting of multiple be elucidated, including a resolution of whether this metabolic
core and emerging hallmark capabilities and enabling character- reprogramming is a discrete capability separable from the core
istics (Figure 6) in mechanism-guided combinations will result in hallmark of chronically sustained proliferation. We remain
more effective and durable therapies for human cancer. perplexed as to whether immune surveillance is a barrier that
virtually all tumors must circumvent, or only an idiosyncrasy of
CONCLUSION AND FUTURE VISION an especially immunogenic subset of them; this issue too will
be resolved in one way or another.
We have sought here to revisit, refine, and extend the concept of Yet other areas are currently in rapid flux. In recent years, elab-
cancer hallmarks, which has provided a useful conceptual orate molecular mechanisms controlling transcription through
framework for understanding the complex biology of cancer. chromatin modifications have been uncovered, and there are

668 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


clues that specific shifts in chromatin configuration occur during Al-Hajj, M., Wicha, M.S., Benito-Hernandez, A., Morrison, S.J., and Clarke,
the acquisition of certain hallmark capabilities (Berdasco and Es- M.F. (2003). Prospective identification of tumorigenic breast cancer cells.
Proc. Natl. Acad. Sci. USA 100, 3983–3988.
teller, 2010). Functionally significant epigenetic alterations seem
likely to be factors not only in the cancer cells but also in the Amaravadi, R.K., and Thompson, C.B. (2007). The roles of therapy-induced
autophagy and necrosis in cancer treatment. Clin. Cancer Res. 13, 7271–7279.
altered cells of the tumor-associated stroma. It is unclear at
Amit, I., Citri, A., Shay, T., Lu, Y., Katz, M., Zhang, F., Tarcic, G., Siwak, D., La-
present whether an elucidation of these epigenetic mechanisms
had, J., Jacob-Hirsch, J., et al. (2007). A module of negative feedback regula-
will materially change our overall understanding of the means by tors defines growth factor signaling. Nat. Genet. 39, 503–512.
which hallmark capabilities are acquired or simply add additional
Apel, A., Zentgraf, H., Büchler, M.W., and Herr, I. (2009). Autophagy-A double-
detail to the regulatory circuitry that is already known to govern edged sword in oncology. Int. J. Cancer 125, 991–995.
them. Artandi, S.E., and DePinho, R.A. (2000). Mice without telomerase: what can
Similarly, the discovery of hundreds of distinct regulatory mi- they teach us about human cancer? Nat. Med. 6, 852–855.
croRNAs has already led to profound changes in our under- Artandi, S.E., and DePinho, R.A. (2010). Telomeres and telomerase in cancer.
standing of the genetic control mechanisms that operate in Carcinogenesis 31, 9–18.
health and disease. By now dozens of microRNAs have been Azam, F., Mehta, S., and Harris, A.L. (2010). Mechanisms of resistance to anti-
implicated in various tumor phenotypes (Garzon et al., 2010), angiogenesis therapy. Eur. J. Cancer 46, 1323–1332.
and yet these only scratch the surface of the real complexity, Baeriswyl, V., and Christofori, G. (2009). The angiogenic switch in carcinogen-
as the functions of hundreds of microRNAs known to be present esis. Semin. Cancer Biol. 19, 329–337.
in our cells and altered in expression in different forms of cancer Baluk, P., Hashizume, H., and McDonald, D.M. (2005). Cellular abnormalities
remain total mysteries. Here again, we are unclear as to whether of blood vessels as targets in cancer. Curr. Opin. Genet. Dev. 15, 102–111.
future progress will cause fundamental shifts in our under- Barkan, D., Green, J.E., and Chambers, A.F. (2010). Extracellular matrix:
standing of the pathogenetic mechanisms of cancer or only a gatekeeper in the transition from dormancy to metastatic growth. Eur. J.
add detail to the elaborate regulatory circuits that have already Cancer 46, 1181–1188.
been mapped out. Barnes, D.E., and Lindahl, T. (2004). Repair and genetic consequences of endog-
Finally, the circuit diagrams of heterotypic interactions enous DNA base damage in mammalian cells. Annu. Rev. Genet. 38, 445–476.
between the multiple distinct cell types that assemble and Barrallo-Gimeno, A., and Nieto, M.A. (2005). The Snail genes as inducers of cell
collaborate to produce different forms and progressively malig- movement and survival: implications in development and cancer. Develop-
ment 132, 3151–3161.
nant stages of cancer are currently rudimentary. In another
Berdasco, M., and Esteller, M. (2010). Aberrant epigenetic landscape in
decade, we anticipate that the signaling circuitry describing
cancer: How cellular identity goes awry. Dev. Cell 19, 698–711.
the intercommunication between these various cells within
Bergers, G., and Benjamin, L.E. (2003). Tumorigenesis and the angiogenic
tumors will be charted in far greater detail and clarity, eclipsing
switch. Nat. Rev. Cancer 3, 401–410.
our current knowledge. And, as before (Hanahan and Weinberg,
Bergers, G., and Hanahan, D. (2008). Modes of resistance to anti-angiogenic
2000), we continue to foresee cancer research as an increasingly therapy. Nat. Rev. Cancer 8, 592–603.
logical science, in which myriad phenotypic complexities are
Bergers, G., and Song, S. (2005). The role of pericytes in blood-vessel forma-
manifestations of a small set of underlying organizing principles. tion and maintenance. Neuro-oncol. 7, 452–464.
Bergfeld, S.A., and DeClerck, Y.A. (2010). Bone marrow-derived mesen-
SUPPLEMENTAL INFORMATION chymal stem cells and the tumor microenvironment. Cancer Metastasis Rev.
29, 249–261.
Supplemental Information includes six figures that are downloadable for Berx, G., and van Roy, F. (2009). Involvement of members of the cadherin
presentations and can be found with this article online at doi:10.1016/j.cell. superfamily in cancer. Cold Spring Harb. Perspect. Biol. 1, a003129.
2011.02.013.
Bhowmick, N.A., Neilson, E.G., and Moses, H.L. (2004). Stromal fibroblasts in
cancer initiation and progression. Nature 432, 332–337.
ACKNOWLEDGMENTS
Bierie, B., and Moses, H.L. (2006). Tumour microenvironment: TGFbeta: the
molecular Jekyll and Hyde of cancer. Nat. Rev. Cancer 6, 506–520.
We thank Terry Schoop (OFC Graphics, Kensington, CA, USA) for exceptional
efforts in preparing the figures. And we thank Gerard Evan (Cambridge, UK), Bindea, G., Mlecnik, B., Fridman, W.H., Pagès, F., and Galon, J. (2010). Natural
Erwin Wagner (Madrid, ESP), and Zena Werb (San Francisco, USA) for valu- immunity to cancer in humans. Curr. Opin. Immunol. 22, 215–222.
able comments and suggestions on the manuscript. D.H. and R.A.W. are Biswas, S.K., and Mantovani, A. (2010). Macrophage plasticity and interaction
American Cancer Society Research Professors. Research in the authors’ labo- with lymphocyte subsets: cancer as a paradigm. Nat. Immunol. 11, 889–896.
ratories has been largely supported by the U.S. National Cancer Institute. Due Blasco, M.A. (2005). Telomeres and human disease: ageing, cancer and
to space limitations, many primary and historical publications have not been beyond. Nat. Rev. Genet. 6, 611–622.
cited, particularly in cases where topical reviews are available.
Boiko, A.D., Razorenova, O.V., van de Rijn, M., Swetter, S.M., Johnson, D.L.,
Ly, D.P., Butler, P.D., Yang, G.P., Joshua, B., Kaplan, M.J., et al. (2010).
REFERENCES Human melanoma-initiating cells express neural crest nerve growth factor
receptor CD271. Nature 466, 133–137.
Adams, J.M., and Cory, S. (2007). The Bcl-2 apoptotic switch in cancer devel- Bonnet, D., and Dick, J.E. (1997). Human acute myeloid leukemia is organized
opment and therapy. Oncogene 26, 1324–1337. as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3,
Aguirre-Ghiso, J.A. (2007). Models, mechanisms and clinical evidence for 730–737.
cancer dormancy. Nat. Rev. Cancer 7, 834–846. Bos, P.D., Zhang, X.H., Nadal, C., Shu, W., Gomis, R.R., Nguyen, D.X., Minn, A.J.,
Ahmed, Z., and Bicknell, R. (2009). Angiogenic signalling pathways. Methods van de Vijver, M.J., Gerald, W.L., Foekens, J.A., and Massagué, J. (2009). Genes
Mol. Biol. 467, 3–24. that mediate breast cancer metastasis to the brain. Nature 459, 1005–1009.

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 669


Brabletz, T., Jung, A., Reu, S., Porzner, M., Hlubek, F., Kunz-Schughart, L.A., DeBerardinis, R.J., Lum, J.J., Hatzivassiliou, G., and Thompson, C.B. (2008).
Knuechel, R., and Kirchner, T. (2001). Variable beta-catenin expression in The biology of cancer: Metabolic reprogramming fuels cell growth and prolif-
colorectal cancers indicates tumor progression driven by the tumor environ- eration. Cell Metab. 7, 11–20.
ment. Proc. Natl. Acad. Sci. USA 98, 10356–10361. Dejana, E., Orsenigo, F., Molendini, C., Baluk, P., and McDonald, D.M. (2009).
Brabletz, T., Jung, A., Spaderna, S., Hlubek, F., and Kirchner, T. (2005). Organization and signaling of endothelial cell-to-cell junctions in various
Opinion: migrating cancer stem cells - an integrated concept of malignant regions of the blood and lymphatic vascular trees. Cell Tissue Res. 335, 17–25.
tumour progression. Nat. Rev. Cancer 5, 744–749. Demicheli, R., Retsky, M.W., Hrushesky, W.J., Baum, M., and Gukas, I.D.
Buck, E., Eyzaguirre, A., Barr, S., Thompson, S., Sennello, R., Young, D., (2008). The effects of surgery on tumor growth: a century of investigations.
Iwata, K.K., Gibson, N.W., Cagnoni, P., and Haley, J.D. (2007). Loss of homo- Ann. Oncol. 19, 1821–1828.
typic cell adhesion by epithelial-mesenchymal transition or mutation limits DeNardo, D.G., Andreu, P., and Coussens, L.M. (2010). Interactions between
sensitivity to epidermal growth factor receptor inhibition. Mol. Cancer Ther. lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity.
6, 532–541. Cancer Metastasis Rev. 29, 309–316.
Burkhart, D.L., and Sage, J. (2008). Cellular mechanisms of tumour suppres- De Palma, M., Murdoch, C., Venneri, M.A., Naldini, L., and Lewis, C.E. (2007).
sion by the retinoblastoma gene. Nat. Rev. Cancer 8, 671–682. Tie2-expressing monocytes: regulation of tumor angiogenesis and therapeutic
Cabrita, M.A., and Christofori, G. (2008). Sprouty proteins, masterminds of implications. Trends Immunol. 28, 519–524.
receptor tyrosine kinase signaling. Angiogenesis 11, 53–62. Deshpande, A., Sicinski, P., and Hinds, P.W. (2005). Cyclins and cdks in devel-
Campbell, P.J., Yachida, S., Mudie, L.J., Stephens, P.J., Pleasance, E.D., opment and cancer: a perspective. Oncogene 24, 2909–2915.
Stebbings, L.A., Morsberger, L.A., Latimer, C., McLaren, S., Lin, M.L., et al. de Visser, K.E., Eichten, A., and Coussens, L.M. (2006). Paradoxical roles of
(2010). The patterns and dynamics of genomic instability in metastatic pancre- the immune system during cancer development. Nat. Rev. Cancer 6, 24–37.
atic cancer. Nature 467, 1109–1113. Dirat, B., Bochet, L., Escourrou, G., Valet, P., and Muller, C. (2010). Unraveling
Cao, Y. (2010). Adipose tissue angiogenesis as a therapeutic target for obesity the obesity and breast cancer links: a role for cancer-associated adipocytes?
and metabolic diseases. Nat. Rev. Drug Discov. 9, 107–115. Endocr. Dev. 19, 45–52.
Carmeliet, P. (2005). VEGF as a key mediator of angiogenesis in cancer. Dvorak, H.F. (1986). Tumors: wounds that do not heal. Similarities between
Oncology 69 (Suppl 3), 4–10. tumor stroma generation and wound healing. N. Engl. J. Med. 315, 1650–1659.
Carmeliet, P., and Jain, R.K. (2000). Angiogenesis in cancer and other Ebos, J.M., Lee, C.R., and Kerbel, R.S. (2009). Tumor and host-mediated path-
diseases. Nature 407, 249–257. ways of resistance and disease progression in response to antiangiogenic
therapy. Clin. Cancer Res. 15, 5020–5025.
Cavallaro, U., and Christofori, G. (2004). Cell adhesion and signalling by cad-
herins and Ig-CAMs in cancer. Nat. Rev. Cancer 4, 118–132. Egeblad, M., Nakasone, E.S., and Werb, Z. (2010). Tumors as organs: complex
tissues that interface with the entire organism. Dev. Cell 18, 884–901.
Cheng, N., Chytil, A., Shyr, Y., Joly, A., and Moses, H.L. (2008). Transforming
growth factor-beta signaling-deficient fibroblasts enhance hepatocyte growth El Hallani, S., Boisselier, B., Peglion, F., Rousseau, A., Colin, C., Idbaih, A.,
factor signaling in mammary carcinoma cells to promote scattering and inva- Marie, Y., Mokhtari, K., Thomas, J.L., Eichmann, A., et al. (2010). A new alter-
sion. Mol. Cancer Res. 6, 1521–1533. native mechanism in glioblastoma vascularization: tubular vasculogenic
mimicry. Brain 133, 973–982.
Chin, K., de Solorzano, C.O., Knowles, D., Jones, A., Chou, W., Rodriguez,
E.G., Kuo, W.L., Ljung, B.M., Chew, K., Myambo, K., et al. (2004). In situ anal- Ellis, L.M., and Reardon, D.A. (2009). Cancer: The nuances of therapy. Nature
yses of genome instability in breast cancer. Nat. Genet. 36, 984–988. 458, 290–292.

Cho, R.W., and Clarke, M.F. (2008). Recent advances in cancer stem cells. Esteller, M. (2007). Cancer epigenomics: DNA methylomes and histone-modi-
Curr. Opin. Genet. Dev. 18, 1–6. fication maps. Nat. Rev. Genet. 8, 286–298.
Evan, G.I., and d’Adda di Fagagna, F. (2009). Cellular senescence: hot or
Ciccia, A., and Elledge, S.J. (2010). The DNA damage response: making it safe
what? Curr. Opin. Genet. Dev. 19, 25–31.
to play with knives. Mol. Cell 40, 179–204.
Evan, G., and Littlewood, T. (1998). A matter of life and cell death. Science 281,
Coffelt, S.B., Lewis, C.E., Naldini, L., Brown, J.M., Ferrara, N., and De Palma,
1317–1322.
M. (2010). Elusive identities and overlapping phenotypes of proangiogenic
myeloid cells in tumors. Am. J. Pathol. 176, 1564–1576. Fang, S., and Salven, P. (2011). Stem cells in tumor angiogenesis. J. Mol. Cell.
Cardiol. 50, 290–295.
Coghlin, C., and Murray, G.I. (2010). Current and emerging concepts in tumour
metastasis. J. Pathol. 222, 1–15. Feron, O. (2009). Pyruvate into lactate and back: from the Warburg effect to
symbiotic energy fuel exchange in cancer cells. Radiother. Oncol. 92, 329–
Collado, M., and Serrano, M. (2010). Senescence in tumours: evidence from
333.
mice and humans. Nat. Rev. Cancer 10, 51–57.
Feldser, D.M., and Greider, C.W. (2007). Short telomeres limit tumor progres-
Colotta, F., Allavena, P., Sica, A., Garlanda, C., and Mantovani, A. (2009). sion in vivo by inducing senescence. Cancer Cell 11, 461–469.
Cancer-related inflammation, the seventh hallmark of cancer: links to genetic
instability. Carcinogenesis 30, 1073–1081. Ferrara, N. (2009). Vascular endothelial growth factor. Arterioscler. Thromb.
Vasc. Biol. 29, 789–791.
Cong, Y., and Shay, J.W. (2008). Actions of human telomerase beyond telo-
Ferrara, N. (2010). Pathways mediating VEGF-independent tumor angiogen-
meres. Cell Res. 18, 725–732.
esis. Cytokine Growth Factor Rev. 21, 21–26.
Creighton, C.J., Li, X., Landis, M., Dixon, J.M., Neumeister, V.M., Sjolund, A.,
Ferrone, C., and Dranoff, G. (2010). Dual roles for immunity in gastrointestinal
Rimm, D.L., Wong, H., Rodriguez, A., Herschkowitz, J.I., et al. (2009). Residual
cancers. J. Clin. Oncol. 28, 4045–4051.
breast cancers after conventional therapy display mesenchymal as well as
tumor-initiating features. Proc. Natl. Acad. Sci. USA 106, 13820–13825. Fidler, I.J. (2003). The pathogenesis of cancer metastasis: the ‘seed and soil’
hypothesis revisited. Nat. Rev. Cancer 3, 453–458.
Curto, M., Cole, B.K., Lallemand, D., Liu, C.H., and McClatchey, A.I. (2007).
Contact-dependent inhibition of EGFR signaling by Nf2/Merlin. J. Cell Biol. Folkman, J. (2002). Role of angiogenesis in tumor growth and metastasis.
177, 893–903. Semin. Oncol. 29(6, Suppl 16), 15–18.

Davies, M.A., and Samuels, Y. (2010). Analysis of the genome to personalize Folkman, J. (2006). Angiogenesis. Annu. Rev. Med. 57, 1–18.
therapy for melanoma. Oncogene 29, 5545–5555. Folkman, J., and Kalluri, R. (2004). Cancer without disease. Nature 427, 787.

670 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


Friedberg, E.C., Aguilera, A., Gellert, M., Hanawalt, P.C., Hays, J.B., Lehmann, Ikushima, H., and Miyazono, K. (2010). TGFbeta signalling: a complex web in
A.R., Lindahl, T., Lowndes, N., Sarasin, A., and Wood, R.D. (2006). DNA repair: cancer progression. Nat. Rev. Cancer 10, 415–424.
from molecular mechanism to human disease. DNA Repair (Amst.) 5, 986–996. Ince, T.A., Richardson, A.L., Bell, G.W., Saitoh, M., Godar, S., Karnoub, A.E.,
Friedl, P., and Wolf, K. (2008). Tube travel: the role of proteases in individual Iglehart, J.D., and Weinberg, R.A. (2007). Transformation of different human
and collective cancer cell invasion. Cancer Res. 68, 7247–7249. breast epithelial cell types leads to distinct tumor phenotypes. Cancer Cell
Friedl, P., and Wolf, K. (2010). Plasticity of cell migration: a multiscale tuning 12, 160–170.
model. J. Cell Biol. 188, 11–19. Jackson, S.P., and Bartek, J. (2009). The DNA-damage response in human
biology and disease. Nature 461, 1071–1078.
Gaengel, K., Genové, G., Armulik, A., and Betsholtz, C. (2009). Endothelial-
mural cell signaling in vascular development and angiogenesis. Arterioscler. Jiang, B.H., and Liu, L.Z. (2009). PI3K/PTEN signaling in angiogenesis and
Thromb. Vasc. Biol. 29, 630–638. tumorigenesis. Adv. Cancer Res. 102, 19–65.
Galluzzi, L., and Kroemer, G. (2008). Necroptosis: a specialized pathway of Johansson, M., Denardo, D.G., and Coussens, L.M. (2008). Polarized immune
programmed necrosis. Cell 135, 1161–1163. responses differentially regulate cancer development. Immunol. Rev. 222,
145–154.
Garzon, R., Marcucci, G., and Croce, C.M. (2010). Targeting microRNAs in
cancer: rationale, strategies and challenges. Nat. Rev. Drug Discov. 9, 775– Jones, P.A., and Baylin, S.B. (2007). The epigenomics of cancer. Cell 128, 683–
789. 692.

Gerhardt, H., and Semb, H. (2008). Pericytes: gatekeepers in tumour cell Jones, R.G., and Thompson, C.B. (2009). Tumor suppressors and cell metab-
metastasis? J. Mol. Med. 86, 135–144. olism: a recipe for cancer growth. Genes Dev. 23, 537–548.
Joyce, J.A., and Pollard, J.W. (2009). Microenvironmental regulation of metas-
Ghebranious, N., and Donehower, L.A. (1998). Mouse models in tumor
tasis. Nat. Rev. Cancer 9, 239–252.
suppression. Oncogene 17, 3385–3400.
Junttila, M.R., and Evan, G.I. (2009). p53—a Jack of all trades but master of
Giaccia, A.J., and Schipani, E. (2010). Role of carcinoma-associated fibro-
none. Nat. Rev. Cancer 9, 821–829.
blasts and hypoxia in tumor progression. Curr. Top. Microbiol. Immunol.
345, 31–45. Kalluri, R., and Zeisberg, M. (2006). Fibroblasts in cancer. Nat. Rev. Cancer 6,
392–401.
Gilbertson, R.J., and Rich, J.N. (2007). Making a tumour’s bed: glioblastoma
stem cells and the vascular niche. Nat. Rev. Cancer 7, 733–736. Kang, H.J., Choi, Y.S., Hong, S.B., Kim, K.W., Woo, R.S., Won, S.J., Kim, E.J.,
Jeon, H.K., Jo, S.Y., Kim, T.K., et al. (2004). Ectopic expression of the catalytic
Gocheva, V., Wang, H.W., Gadea, B.B., Shree, T., Hunter, K.E., Garfall, A.L.,
subunit of telomerase protects against brain injury resulting from ischemia and
Berman, T., and Joyce, J.A. (2010). IL-4 induces cathepsin protease activity
NMDA-induced neurotoxicity. J. Neurosci. 24, 1280–1287.
in tumor-associated macrophages to promote cancer growth and invasion.
Genes Dev. 24, 241–255. Karin, M., Lawrence, T., and Nizet, V. (2006). Innate immunity gone awry: link-
ing microbial infections to chronic inflammation and cancer. Cell 124, 823–835.
Grivennikov, S.I., Greten, F.R., and Karin, M. (2010). Immunity, inflammation,
Karnoub, A.E., and Weinberg, R.A. (2006–2007). Chemokine networks and
and cancer. Cell 140, 883–899.
breast cancer metastasis. Breast Dis. 26, 75–85.
Gupta, G.P., Minn, A.J., Kang, Y., Siegel, P.M., Serganova, I., Cordón-Cardo,
Karnoub, A.E., Dash, A.B., Vo, A.P., Sullivan, A., Brooks, M.W., Bell, G.W.,
C., Olshen, A.B., Gerald, W.L., and Massagué, J. (2005). Identifying site-
Richardson, A.L., Polyak, K., Tubo, R., and Weinberg, R.A. (2007). Mesen-
specific metastasis genes and functions. Cold Spring Harb. Symp. Quant.
chymal stem cells within tumour stroma promote breast cancer metastasis.
Biol. 70, 149–158.
Nature 449, 557–563.
Gupta, P.B., Chaffer, C.L., and Weinberg, R.A. (2009). Cancer stem cells:
Kastan, M.B. (2008). DNA damage responses: mechanisms and roles in
mirage or reality? Nat. Med. 15, 1010–1012.
human disease: 2007 G.H.A. Clowes Memorial Award Lecture. Mol. Cancer
Hanahan, D., and Folkman, J. (1996). Patterns and emerging mechanisms of Res. 6, 517–524.
the angiogenic switch during tumorigenesis. Cell 86, 353–364.
Kawai, T., Hiroi, S., Nakanishi, K., and Meeker, A.K. (2007). Telomere length
Hanahan, D., and Weinberg, R.A. (2000). The hallmarks of cancer. Cell 100, and telomerase expression in atypical adenomatous hyperplasia and small
57–70. bronchioloalveolar carcinoma of the lung. Am. J. Clin. Pathol. 127, 254–262.
Hansel, D.E., Meeker, A.K., Hicks, J., De Marzo, A.M., Lillemoe, K.D., Schulick, Kazerounian, S., Yee, K.O., and Lawler, J. (2008). Thrombospondins in cancer.
R., Hruban, R.H., Maitra, A., and Argani, P. (2006). Telomere length variation in Cell. Mol. Life Sci. 65, 700–712.
biliary tract metaplasia, dysplasia, and carcinoma. Mod. Pathol. 19, 772–779.
Kenific, C.M., Thorburn, A., and Debnath, J. (2010). Autophagy and metas-
Hardee, M.E., Dewhirst, M.W., Agarwal, N., and Sorg, B.S. (2009). Novel tasis: another double-edged sword. Curr. Opin. Cell Biol. 22, 241–245.
imaging provides new insights into mechanisms of oxygen transport in tumors. Kennedy, K.M., and Dewhirst, M.W. (2010). Tumor metabolism of lactate: the
Curr. Mol. Med. 9, 435–441. influence and therapeutic potential for MCT and CD147 regulation. Future
Harper, J.W., and Elledge, S.J. (2007). The DNA damage response: Ten years Oncol. 6, 127–148.
after. Mol. Cell 28, 739–745. Kessenbrock, K., Plaks, V., and Werb, Z. (2010). Matrix metalloproteinases:
Hezel, A.F., and Bardeesy, N. (2008). LKB1; linking cell structure and tumor Regulators of the tumor microenvironment. Cell 141, 52–67.
suppression. Oncogene 27, 6908–6919. Kim, M.Y., Oskarsson, T., Acharyya, S., Nguyen, D.X., Zhang, X.H., Norton, L.,
Hlubek, F., Brabletz, T., Budczies, J., Pfeiffer, S., Jung, A., and Kirchner, T. and Massagué, J. (2009). Tumor self-seeding by circulating cancer cells. Cell
(2007). Heterogeneous expression of Wnt/beta-catenin target genes within 139, 1315–1326.
colorectal cancer. Int. J. Cancer 121, 1941–1948. Kim, R., Emi, M., and Tanabe, K. (2007). Cancer immunoediting from immune
Hugo, H., Ackland, M.L., Blick, T., Lawrence, M.G., Clements, J.A., Williams, surveillance to immune escape. Immunology 121, 1–14.
E.D., and Thompson, E.W. (2007). Epithelial—mesenchymal and mesen- Kinzler, K.W., and Vogelstein, B. (1997). Cancer-susceptibility genes. Gate-
chymal—epithelial transitions in carcinoma progression. J. Cell. Physiol. keepers and caretakers. Nature 386, 761–763.
213, 374–383. Klein, C.A. (2009). Parallel progression of primary tumours and metastases.
Hsu, P.P., and Sabatini, D.M. (2008). Cancer cell metabolism: Warburg and Nat. Rev. Cancer 9, 302–312.
beyond. Cell 134, 703–707. Klymkowsky, M.W., and Savagner, P. (2009). Epithelial-mesenchymal transi-
Hynes, N.E., and MacDonald, G. (2009). ErbB receptors and signaling path- tion: a cancer researcher’s conceptual friend and foe. Am. J. Pathol. 174,
ways in cancer. Curr. Opin. Cell Biol. 21, 177–184. 1588–1593.

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 671


Korkola, J., and Gray, J.W. (2010). Breast cancer genomes—form and func- Mooi, W.J., and Peeper, D.S. (2006). Oncogene-induced cell senescence—
tion. Curr. Opin. Genet. Dev. 20, 4–14. halting on the road to cancer. N. Engl. J. Med. 355, 1037–1046.
Kovacic, J.C., and Boehm, M. (2009). Resident vascular progenitor cells: an Morel, A.-P., Lièvre, M., Thomas, C., Hinkal, G., Ansieau, S., and Puisieux, A.
emerging role for non-terminally differentiated vessel-resident cells in vascular (2008). Generation of breast cancer stem cells through epithelial-mesen-
biology. Stem Cell Res. (Amst.) 2, 2–15. chymal transition. PLoS ONE 3, e2888.
Kroemer, G., and Pouyssegur, J. (2008). Tumor cell metabolism: Cancer’s Mosesson, Y., Mills, G.B., and Yarden, Y. (2008). Derailed endocytosis: an
Achilles’ heel. Cancer Cell 13, 472–482. emerging feature of cancer. Nat. Rev. Cancer 8, 835–850.
Lamagna, C., and Bergers, G. (2006). The bone marrow constitutes a reservoir Mougiakakos, D., Choudhury, A., Lladser, A., Kiessling, R., and Johansson,
of pericyte progenitors. J. Leukoc. Biol. 80, 677–681. C.C. (2010). Regulatory T cells in cancer. Adv. Cancer Res. 107, 57–117.
Lane, D.P. (1992). Cancer. p53, guardian of the genome. Nature 358, 15–16. Murdoch, C., Muthana, M., Coffelt, S.B., and Lewis, C.E. (2008). The role of myeloid
Lemmon, M.A., and Schlessinger, J. (2010). Cell signaling by receptor tyrosine cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer 8, 618–631.
kinases. Cell 141, 1117–1134. Nagy, J.A., Chang, S.H., Shih, S.C., Dvorak, A.M., and Dvorak, H.F. (2010).
Heterogeneity of the tumor vasculature. Semin. Thromb. Hemost. 36,
Levine, B., and Kroemer, G. (2008). Autophagy in the pathogenesis of disease.
321–331.
Cell 132, 27–42.
Naumov, G.N., Folkman, J., Straume, O., and Akslen, L.A. (2008). Tumor-
Lipinski, M.M., and Jacks, T. (1999). The retinoblastoma gene family in differ-
vascular interactions and tumor dormancy. APMIS 116, 569–585.
entiation and development. Oncogene 18, 7873–7882.
Negrini, S., Gorgoulis, V.G., and Halazonetis, T.D. (2010). Genomic insta-
Lobo, N.A., Shimono, Y., Qian, D., and Clarke, M.F. (2007). The biology of
bility—an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11, 220–228.
cancer stem cells. Annu. Rev. Cell Dev. Biol. 23, 675–699.
Nelson, B.H. (2008). The impact of T-cell immunity on ovarian cancer
Lowe, S.W., Cepero, E., and Evan, G. (2004). Intrinsic tumour suppression.
outcomes. Immunol. Rev. 222, 101–116.
Nature 432, 307–315.
Nguyen, D.X., Bos, P.D., and Massagué, J. (2009). Metastasis: from dissemi-
Luebeck, E.G. (2010). Cancer: Genomic evolution of metastasis. Nature 467,
nation to organ-specific colonization. Nat. Rev. Cancer 9, 274–284.
1053–1055.
Norden, A.D., Drappatz, J., and Wen, P.Y. (2009). Antiangiogenic therapies for
Lu, Z., Luo, R.Z., Lu, Y., Zhang, X., Yu, Q., Khare, S., Kondo, S., Kondo, Y., Yu,
high-grade glioma. Nat. Rev. Neurol. 5, 610–620.
Y., Mills, G.B., et al. (2008). The tumor suppressor gene ARHI regulates au-
tophagy and tumor dormancy in human ovarian cancer cells. J. Clin. Invest. Nyberg, P., Xie, L., and Kalluri, R. (2005). Endogenous inhibitors of angiogen-
118, 3917–3929. esis. Cancer Res. 65, 3967–3979.

Luo, J., Solimini, N.L., and Elledge, S.J. (2009). Principles of cancer therapy: Okada, T., Lopez-Lago, M., and Giancotti, F.G. (2005). Merlin/NF-2 mediates
Oncogene and non-oncogene addiction. Cell 136, 823–837. contact inhibition of growth by suppressing recruitment of Rac to the plasma
membrane. J. Cell Biol. 171, 361–371.
Mac Gabhann, F., and Popel, A.S. (2008). Systems biology of vascular endo-
thelial growth factors. Microcirculation 15, 715–738. Olive, K.P., Jacobetz, M.A., Davidson, C.J., Gopinathan, A., McIntyre, D., Hon-
ess, D., Madhu, B., Goldgraben, M.A., Caldwell, M.E., Allard, D., et al. (2009).
Madsen, C.D., and Sahai, E. (2010). Cancer dissemination—Lessons from
Inhibition of Hedgehog signaling enhances delivery of chemotherapy in
leukocytes. Dev. Cell 19, 13–26.
a mouse model of pancreatic cancer. Science 324, 1457–1461.
Maida, Y., Yasukawa, M., Furuuchi, M., Lassmann, T., Possemato, R., Oka-
Olson, P., Lu, J., Zhang, H., Shai, A., Chun, M.G., Wang, Y., Libutti, S.K., Na-
moto, N., Kasim, V., Hayashizaki, Y., Hahn, W.C., and Masutomi, K. (2009).
kakura, E.K., Golub, T.R., and Hanahan, D. (2009). MicroRNA dynamics in the
An RNA-dependent RNA polymerase formed by TERT and the RMRP RNA.
stages of tumorigenesis correlate with hallmark capabilities of cancer. Genes
Nature 461, 230–235.
Dev. 23, 2152–2165.
Mani, S.A., Guo, W., Liao, M.J., Eaton, E.N., Ayyanan, A., Zhou, A.Y., Brooks, M.,
O’Reilly, K.E., Rojo, F., She, Q.B., Solit, D., Mills, G.B., Smith, D., Lane, H., Hof-
Reinhard, F., Zhang, C.C., Shipitsin, M., et al. (2008). The epithelial-mesen-
mann, F., Hicklin, D.J., Ludwig, D.L., et al. (2006). mTOR inhibition induces
chymal transition generates cells with properties of stem cells. Cell 133, 704–715.
upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res.
Mantovani, A. (2010). Molecular pathways linking inflammation and cancer. 66, 1500–1508.
Curr. Mol. Med. 10, 369–373.
Ostrand-Rosenberg, S., and Sinha, P. (2009). Myeloid-derived suppressor
Mantovani, A., Allavena, P., Sica, A., and Balkwill, F. (2008). Cancer-related cells: linking inflammation and cancer. J. Immunol. 182, 4499–4506.
inflammation. Nature 454, 436–444.
Pagès, F., Galon, J., Dieu-Nosjean, M.C., Tartour, E., Sautès-Fridman, C., and
Massagué, J. (2008). TGFb in cancer. Cell 134, 215–230. Fridman, W.H. (2010). Immune infiltration in human tumors: a prognostic factor
Masutomi, K., Possemato, R., Wong, J.M., Currier, J.L., Tothova, Z., Manola, that should not be ignored. Oncogene 29, 1093–1102.
J.B., Ganesan, S., Lansdorp, P.M., Collins, K., and Hahn, W.C. (2005). The Palermo, C., and Joyce, J.A. (2008). Cysteine cathepsin proteases as pharma-
telomerase reverse transcriptase regulates chromatin state and DNA damage cological targets in cancer. Trends Pharmacol. Sci. 29, 22–28.
responses. Proc. Natl. Acad. Sci. USA 102, 8222–8227. Park, J.I., Venteicher, A.S., Hong, J.Y., Choi, J., Jun, S., Shkreli, M., Chang, W.,
Mathew, R., Karantza-Wadsworth, V., and White, E. (2007). Role of autophagy Meng, Z., Cheung, P., Ji, H., et al. (2009). Telomerase modulates Wnt signalling
in cancer. Nat. Rev. Cancer 7, 961–967. by association with target gene chromatin. Nature 460, 66–72.
McGowan, P.M., Kirstein, J.M., and Chambers, A.F. (2009). Micrometastatic Partanen, J.I., Nieminen, A.I., and Klefstrom, J. (2009). 3D view to tumor
disease and metastatic outgrowth: clinical issues and experimental suppression: Lkb1, polarity and the arrest of oncogenic c-Myc. Cell Cycle 8,
approaches. Future Oncol. 5, 1083–1098. 716–724.
Micalizzi, D.S., Farabaugh, S.M., and Ford, H.L. (2010). Epithelial-mesen- Pasquale, E.B. (2010). Eph receptors and ephrins in cancer: bidirectional sig-
chymal transition in cancer: parallels between normal development and tumor nalling and beyond. Nat. Rev. Cancer 10, 165–180.
progression. J. Mammary Gland Biol. Neoplasia 15, 117–134. Passos, J.F., Saretzki, G., and von Zglinicki, T. (2007). DNA damage in telo-
Mizushima, N. (2007). Autophagy: process and function. Genes Dev. 21, 2861– meres and mitochondria during cellular senescence: is there a connection?
2873. Nucleic Acids Res. 35, 7505–7513.
Mohamed, M.M., and Sloane, B.F. (2006). Cysteine cathepsins: multifunc- Patenaude, A., Parker, J., and Karsan, A. (2010). Involvement of endothelial
tional enzymes in cancer. Nat. Rev. Cancer 6, 764–775. progenitor cells in tumor vascularization. Microvasc. Res. 79, 217–223.

672 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


Peinado, H., Lavothskin, S., and Lyden, D. (2011). The secreted factors Semenza, G.L. (2010b). Defining the role of hypoxia-inducible factor 1 in
responsible for pre-metastatic niche formation: Old sayings and new thoughts. cancer biology and therapeutics. Oncogene 29, 625–634.
Semin. Cancer Biol. Published online January 18, 2011. 10.1016/j.semcancer. Seppinen, L., Sormunen, R., Soini, Y., Elamaa, H., Heljasvaara, R., and Pihla-
2011.01.002. janiemi, T. (2008). Lack of collagen XVIII accelerates cutaneous wound healing,
Peinado, H., Marin, F., Cubillo, E., Stark, H.J., Fusenig, N., Nieto, M.A., and while overexpression of its endostatin domain leads to delayed healing. Matrix
Cano, A. (2004). Snail and E47 repressors of E-cadherin induce distinct inva- Biol. 27, 535–546.
sive and angiogenic properties in vivo. J. Cell Sci. 117, 2827–2839.
Shaw, R.J. (2009). Tumor suppression by LKB1: SIK-ness prevents metas-
Perona, R. (2006). Cell signalling: growth factors and tyrosine kinase recep- tasis. Sci. Signal. 2, pe55.
tors. Clin. Transl. Oncol. 8, 77–82.
Shay, J.W., and Wright, W.E. (2000). Hayflick, his limit, and cellular ageing. Nat.
Pietras, K., and Ostman, A. (2010). Hallmarks of cancer: interactions with the Rev. Mol. Cell Biol. 1, 72–76.
tumor stroma. Exp. Cell Res. 316, 1324–1331.
Sherr, C.J., and DePinho, R.A. (2000). Cellular senescence: Mitotic clock or
Polyak, K., and Weinberg, R.A. (2009). Transitions between epithelial and
culture shock? Cell 102, 407–410.
mesenchymal states: acquisition of malignant and stem cell traits. Nat. Rev.
Cancer 9, 265–273. Sherr, C.J., and McCormick, F. (2002). The RB and p53 pathways in cancer.
Cancer Cell 2, 103–112.
Polyak, K., Haviv, I., and Campbell, I.G. (2009). Co-evolution of tumor cells and
their microenvironment. Trends Genet. 25, 30–38. Shields, J.D., Kourtis, I.C., Tomei, A.A., Roberts, J.M., and Swartz, M.A.
(2010). Induction of lymphoidlike stroma and immune escape by tumors that
Potter, V.R. (1958). The biochemical approach to the cancer problem. Fed.
express the chemokine CCL21. Science 328, 749–752.
Proc. 17, 691–697.
Qian, B.Z., and Pollard, J.W. (2010). Macrophage diversity enhances tumor Shimoda, M., Mellody, K.T., and Orimo, A. (2010). Carcinoma-associated
progression and metastasis. Cell 141, 39–51. fibroblasts are a rate-limiting determinant for tumour progression. Semin.
Cell Dev. Biol. 21, 19–25.
Quintana, E., Shackleton, M., Sabel, M.S., Fullen, D.R., Johnson, T.M., and
Morrison, S.J. (2008). Efficient tumour formation by single human melanoma Sigal, A., and Rotter, V. (2000). Oncogenic mutations of the p53 tumor
cells. Nature 456, 593–598. suppressor: the demons of the guardian of the genome. Cancer Res. 60,
6788–6793.
Raica, M., Cimpean, A.M., and Ribatti, D. (2009). Angiogenesis in pre-malig-
nant conditions. Eur. J. Cancer 45, 1924–1934. Singh, A., and Settleman, J. (2010). EMT, cancer stem cells and drug resis-
Räsänen, K., and Vaheri, A. (2010). Activation of fibroblasts in cancer stroma. tance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751.
Exp. Cell Res. 316, 2713–2722. Sinha, S., and Levine, B. (2008). The autophagy effector Beclin 1: a novel BH3-
Raynaud, C.M., Hernandez, J., Llorca, F.P., Nuciforo, P., Mathieu, M.C., only protein. Oncogene 27 (Suppl 1), S137–S148.
Commo, F., Delaloge, S., Sabatier, L., André, F., and Soria, J.C. (2010). DNA Smyth, M.J., Dunn, G.P., and Schreiber, R.D. (2006). Cancer immunosurveil-
damage repair and telomere length in normal breast, preneoplastic lesions, lance and immunoediting: the roles of immunity in suppressing tumor develop-
and invasive cancer. Am. J. Clin. Oncol. 33, 341–345. ment and shaping tumor immunogenicity. Adv. Immunol. 90, 1–50.
Raza, A., Franklin, M.J., and Dudek, A.Z. (2010). Pericytes and vessel matura- Soda, Y., Marumoto, T., Friedmann-Morvinski, D., Soda, M., Liu, F., Michiue,
tion during tumor angiogenesis and metastasis. Am. J. Hematol. 85, 593–598. H., Pastorino, S., Yang, M., Hoffman, R.M., Kesari, S., and Verma, I.M. (2011).
Reitman, Z.J., and Yan, H. (2010). Isocitrate dehydrogenase 1 and 2 mutations Feature Article: Transdifferentiation of glioblastoma cells into vascular endo-
in cancer: alterations at a crossroads of cellular metabolism. J. Natl. Cancer thelial cells. Proc. Natl. Acad. Sci. USA. Published online January 24, 2011.
Inst. 102, 932–941. Sudarsanam, S., and Johnson, D.E. (2010). Functional consequences of
Reya, T., Morrison, S.J., Clarke, M.F., and Weissman, I.L. (2001). Stem cells, mTOR inhibition. Curr. Opin. Drug Discov. Devel. 13, 31–40.
cancer, and cancer stem cells. Nature 414, 105–111. Strauss, D.C., and Thomas, J.M. (2010). Transmission of donor melanoma by
Ribatti, D. (2009). Endogenous inhibitors of angiogenesis: a historical review. organ transplantation. Lancet Oncol. 11, 790–796.
Leuk. Res. 33, 638–644.
Talmadge, J.E., and Fidler, I.J. (2010). AACR centennial series: the biology of
Ricci-Vitiani, L., Pallini, R., Biffoni, M., Todaro, M., Invernici, G., Cenci, T., cancer metastasis: historical perspective. Cancer Res. 70, 5649–5669.
Maira, G., Parati, E.A., Stassi, G., Larocca, L.M., and De Maria, R. (2010).
Tammela, T., and Alitalo, K. (2010). Lymphangiogenesis: Molecular mecha-
Tumour vascularization via endothelial differentiation of glioblastoma stem-
nisms and future promise. Cell 140, 460–476.
like cells. Nature 468, 824–828.
Ruoslahti, E. (2002). Specialization of tumour vasculature. Nat. Rev. Cancer 2, Taube, J.H., Herschkowitz, J.I., Komurov, K., Zhou, A.Y., Gupta, S., Yang, J.,
83–90. Hartwell, K., Onder, T.T., Gupta, P.B., Evans, K.W., et al. (2010). Core epithe-
lial-to-mesenchymal transition interactome gene-expression signature is
Ruoslahti, E., Bhatia, S.N., and Sailor, M.J. (2010). Targeting of drugs and
associated with claudin-low and metaplastic breast cancer subtypes. Proc.
nanoparticles to tumors. J. Cell Biol. 188, 759–768.
Natl. Acad. Sci. USA 107, 15449–15454.
Sabeh, F., Shimizu-Hirota, R., and Weiss, S.J. (2009). Protease-dependent
Teng, M.W.L., Swann, J.B., Koebel, C.M., Schreiber, R.D., and Smyth, M.J.
versus -independent cancer cell invasion programs: three-dimensional amoe-
(2008). Immune-mediated dormancy: an equilibrium with cancer. J. Leukoc.
boid movement revisited. J. Cell Biol. 185, 11–19.
Biol. 84, 988–993.
Salk, J.J., Fox, E.J., and Loeb, L.A. (2010). Mutational heterogeneity in human
Thiery, J.P., and Sleeman, J.P. (2006). Complex networks orchestrate epithe-
cancers: origin and consequences. Ann. Rev. Pathol. 5, 51–75.
lial-mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 7, 131–142.
Schäfer, M., and Werner, S. (2008). Cancer as an overhealing wound: an old
hypothesis revisited. Nat. Rev. Mol. Cell Biol. 9, 628–638. Thiery, J.P., Acloque, H., Huang, R.Y., and Nieto, M.A. (2009). Epithelial-
mesenchymal transitions in development and disease. Cell 139, 871–890.
Schmalhofer, O., Brabletz, S., and Brabletz, T. (2009). E-cadherin, beta-cate-
nin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev. 28, Townson, J.L., and Chambers, A.F. (2006). Dormancy of solitary metastatic
151–166. cells. Cell Cycle 5, 1744–1750.
Semenza, G.L. (2008). Tumor metabolism: cancer cells give and take lactate. Turner, H.E., Harris, A.L., Melmed, S., and Wass, J.A. (2003). Angiogenesis in
J. Clin. Invest. 118, 3835–3837. endocrine tumors. Endocr. Rev. 24, 600–632.
Semenza, G.L. (2010a). HIF-1: upstream and downstream of cancer metabo- Vajdic, C.M., and van Leeuwen, M.T. (2009). Cancer incidence and risk factors
lism. Curr. Opin. Genet. Dev. 20, 51–56. after solid organ transplantation. Int. J. Cancer 125, 1747–1754.

Cell 144, March 4, 2011 ª2011 Elsevier Inc. 673


Vander Heiden, M.G., Cantley, L.C., and Thompson, C.B. (2009). Under- phage-assisted tumor cell intravasation in mammary tumors. Cancer Res.
standing the Warburg effect: the metabolic requirements of cell proliferation. 67, 2649–2656.
Science 324, 1029–1033. Yachida, S., Jones, S., Bozic, I., Antal, T., Leary, R., Fu, B., Kamiyama, M., Hru-
Verhoeff, J.J., van Tellingen, O., Claes, A., Stalpers, L.J., van Linde, M.E., Richel, ban, R.H., Eshleman, J.R., Nowak, M.A., et al. (2010). Distant metastasis
D.J., Leenders, W.P., and van Furth, W.R. (2009). Concerns about anti-angio- occurs late during the genetic evolution of pancreatic cancer. Nature 467,
genic treatment in patients with glioblastoma multiforme. BMC Cancer 9, 444. 1114–1117.
Wang, R., Chadalavada, K., Wilshire, J., Kowalik, U., Hovinga, K.E., Geber, A., Yang, J., and Weinberg, R.A. (2008). Epithelial-mesenchymal transition: At the
Fligelman, B., Leversha, M., Brennan, C., and Tabar, V. (2010). Glioblastoma crossroads of development and tumor metastasis. Dev. Cell 14, 818–829.
stem-like cells give rise to tumour endothelium. Nature 468, 829–833. Yang, L., Pang, Y., and Moses, H.L. (2010). TGF-beta and immune cells: an
Warburg, O. (1956a). On the origin of cancer cells. Science 123, 309–314. important regulatory axis in the tumor microenvironment and progression.
Trends Immunol. 31, 220–227.
Warburg, O. (1956b). On respiratory impairment in cancer cells. Science 124,
269–270. Yen, K.E., Bittinger, M.A., Su, S.M., and Fantin, V.R. (2010). Cancer-associ-
ated IDH mutations: biomarker and therapeutic opportunities. Oncogene 29,
Warburg, O.H. (1930). The Metabolism of Tumours: Investigations from the
6409–6417.
Kaiser Wilhelm Institute for Biology, Berlin-Dahlem (London, UK: Arnold
Constable). Yilmaz, M., and Christofori, G. (2009). EMT, the cytoskeleton, and cancer cell
invasion. Cancer Metastasis Rev. 28, 15–33.
Wertz, I.E., and Dixit, V.M. (2010). Regulation of death receptor signaling by the
Yuan, T.L., and Cantley, L.C. (2008). PI3K pathway alterations in cancer: vari-
ubiquitin system. Cell Death Differ. 17, 14–24.
ations on a theme. Oncogene 27, 5497–5510.
White, E., Karp, C., Strohecker, A.M., Guo, Y., and Mathew, R. (2010). Role of
Zee, Y.K., O’Connor, J.P., Parker, G.J., Jackson, A., Clamp, A.R., Taylor, M.B.,
autophagy in suppression of inflammation and cancer. Curr. Opin. Cell Biol.
Clarke, N.W., and Jayson, G.C. (2010). Imaging angiogenesis of genitourinary
22, 212–217.
tumors. Nat. Rev. Urol. 7, 69–82.
White, E., and DiPaola, R.S. (2009). The double-edged sword of autophagy
Zhang, H., Herbert, B.S., Pan, K.H., Shay, J.W., and Cohen, S.N. (2004).
modulation in cancer. Clin. Cancer Res. 15, 5308–5316.
Disparate effects of telomere attrition on gene expression during replicative
Willis, S.N., and Adams, J.M. (2005). Life in the balance: how BH3-only senescence of human mammary epithelial cells cultured under different condi-
proteins induce apoptosis. Curr. Opin. Cell Biol. 17, 617–625. tions. Oncogene 23, 6193–6198.
Witsch, E., Sela, M., and Yarden, Y. (2010). Roles for growth factors in cancer Zong, W.X., and Thompson, C.B. (2006). Necrotic death as a cell fate. Genes
progression. Physiology (Bethesda) 25, 85–101. Dev. 20, 1–15.
Wyckoff, J.B., Wang, Y., Lin, E.Y., Li, J.F., Goswami, S., Stanley, E.R., Segall, Zumsteg, A., and Christofori, G. (2009). Corrupt policemen: inflammatory cells
J.E., Pollard, J.W., and Condeelis, J. (2007). Direct visualization of macro- promote tumor angiogenesis. Curr. Opin. Oncol. 21, 60–70.

674 Cell 144, March 4, 2011 ª2011 Elsevier Inc.


REVIEW

Hallmarks of Cancer: New Dimensions


Douglas Hanahan

ABSTRACT The hallmarks of cancer conceptualization is a heuristic tool for distilling the vast
complexity of cancer phenotypes and genotypes into a provisional set of underly-
ing principles. As knowledge of cancer mechanisms has progressed, other facets of the disease have
emerged as potential refinements. Herein, the prospect is raised that phenotypic plasticity and dis-
rupted differentiation is a discrete hallmark capability, and that nonmutational epigenetic reprogram-
ming and polymorphic microbiomes both constitute distinctive enabling characteristics that facilitate
the acquisition of hallmark capabilities. Additionally, senescent cells, of varying origins, may be added
to the roster of functionally important cell types in the tumor microenvironment.

Significance: Cancer is daunting in the breadth and scope of its diversity, spanning genetics, cell and
tissue biology, pathology, and response to therapy. Ever more powerful experimental and computa-
tional tools and technologies are providing an avalanche of “big data” about the myriad manifestations
of the diseases that cancer encompasses. The integrative concept embodied in the hallmarks of cancer
is helping to distill this complexity into an increasingly logical science, and the provisional new dimen-
sions presented in this perspective may add value to that endeavor, to more fully understand mecha-
nisms of cancer development and malignant progression, and apply that knowledge to cancer medicine.

INTRODUCTION steps of tumor pathogenesis. Certainly, the diversity of malig-


nant pathogenesis spanning multiple tumor types and an
The Hallmarks of Cancer were proposed as a set of func-
increasing plethora of subtypes includes various aberrations
tional capabilities acquired by human cells as they make
(and hence acquired capabilities and characteristics) that are
their way from normalcy to neoplastic growth states, more
the result of tissue-specific barriers necessarily circumvented
specifically capabilities that are crucial for their ability to
during particular tumorigenesis pathways. While appreciat-
form malignant tumors. In these articles (1, 2), Bob Weinberg
ing that such specialized mechanisms can be instrumental,
and I enumerated what we imagined were shared commonali-
we limited the hallmarks designation to parameters having
ties that unite all types of cancer cells at the level of cellular
broad engagement across the spectrum of human cancers.
phenotype. The intent was to provide a conceptual scaffold
The eight hallmarks currently comprise (Fig.  1, left) the
that would make it possible to rationalize the complex phe-
acquired capabilities for sustaining proliferative signaling,
notypes of diverse human tumor types and variants in terms
evading growth suppressors, resisting cell death, enabling
of a common set of underlying cellular parameters. Initially
replicative immortality, inducing/accessing vasculature,
we envisaged the complementary involvement of six distinct
activating invasion and metastasis, reprogramming cellular
hallmark capabilities and later expanded this number to
metabolism, and avoiding immune destruction. In the most
eight. This formulation was influenced by the recognition
recent elaboration of this concept (2), deregulating cellular
that human cancers develop as products of multistep pro-
metabolism and avoiding immune destruction were segre-
cesses, and that the acquisition of these functional capabili-
gated as “emerging hallmarks,” but now, eleven years later, it
ties might be mapped in some fashion to the distinguishable
is evident that they, much like the original six, can be consid-
ered core hallmarks of cancer, and are included as such in the
current depiction (Fig. 1, left).
Ludwig Institute for Cancer Research – Lausanne Branch, Lausanne, Swit- As we noted at the time, these hallmark traits, on their
zerland. The Swiss Institute for Experimental Cancer Research (ISREC)
own, fail to address the complexities of cancer pathogenesis,
within the School of Life Sciences at the Swiss Federal Institute of Tech-
nology Lausanne (EPFL), Lausanne, Switzerland. The Swiss Cancer Center that is, the precise molecular and cellular mechanisms that
Leman (SCCL), Lausanne, Switzerland. allow evolving preneoplastic cells to develop and acquire
Corresponding Author: Douglas Hanahan, Agora Translational Cancer these aberrant phenotypic capabilities in the course of tumor
Research Center, Rue du Bugnon 25A, Lausanne CH-1011, Switzerland. development and malignant progression. Accordingly, we
Phone: 41-21-545-1119; E-mail: douglas.hanahan@epfl.ch added another concept to the discussion, portrayed as “ena-
Cancer Discov 2022;12:31–46 bling characteristics,” consequences of the aberrant condi-
doi: 10.1158/2159-8290.CD-21-1059 tion of neoplasia that provide means by which cancer cells
©2021 American Association for Cancer Research and tumors can adopt these functional traits. As such, the

JANUARY 2022 CANCER DISCOVERY | 31

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

Sustaining Evading Emerging hallmarks &


proliferative signaling growth suppressors enabling characteristics
Unlocking Nonmutational
phenotypic epigenetic
Deregulating Avoiding plasticity reprogramming
cellular immune
metabolism destruction

Resisting Enabling
cell death replicative
immortality

Genome
instability & Tumor-promoting
mutation inflammation
Senescent Polymorphic
cells microbiomes
Inducing or accessing Activating invasion
vasculature & metastasis

Figure 1.  In essence: the Hallmarks of Cancer, circa 2022. Left, the Hallmarks of Cancer currently embody eight hallmark capabilities and two enabling
characteristics. In addition to the six acquired capabilities—Hallmarks of Cancer—proposed in 2000 (1), the two provisional “emerging hallmarks” intro-
duced in 2011 (2)—cellular energetics (now described more broadly as “reprogramming cellular metabolism”) and “avoiding immune destruction”—have
been sufficiently validated to be considered part of the core set. Given the growing appreciation that tumors can become sufficiently vascularized either
by switching on angiogenesis or by co-opting normal tissue vessels (128), this hallmark is also more broadly defined as the capability to induce or oth-
erwise access, principally by invasion and metastasis, vasculature that supports tumor growth. The 2011 sequel further incorporated “tumor-promoting
inflammation” as a second enabling characteristic, complementing overarching “genome instability and mutation,” which together were fundamentally
involved in activating the eight hallmark (functional) capabilities necessary for tumor growth and progression. Right, this review incorporates additional
proposed emerging hallmarks and enabling characteristics involving “unlocking phenotypic plasticity,” “nonmutational epigenetic reprogramming,” “poly-
morphic microbiomes,” and “senescent cells.”

enabling characteristics reflected upon molecular and cel- UNLOCKING PHENOTYPIC PLASTICITY
lular mechanisms by which hallmarks are acquired rather
During organogenesis, the development, determination,
than the aforementioned eight capabilities themselves.
and organization of cells into tissues in order to assume
These two enabling processes were genome instability and
homeostatic functions is accompanied by terminal differen-
tumor-promoting inflammation.
We further recognized that the tumor microenvironment tiation, whereby progenitor cells—sometimes irrevocably—
(TME), herein defined to be composed of heterogeneous and stop growing upon culmination of these processes. As such,
interactive populations of cancer cells and cancer stem cells the end result of cellular differentiation is in most cases
along with a multiplicity of recruited stromal cell types—the antiproliferative and constitutes a clear barrier to the con-
transformed parenchyma and the associated stroma—is now tinuing proliferation that is necessary for neoplasia. There
widely appreciated to play an integral role in tumorigenesis is increasing evidence that unlocking the normally restricted
and malignant progression. capability for phenotypic plasticity in order to evade or
Given the continued interest in these formulations and our escape from the state of terminal differentiation is a criti-
enduring intent to encourage ongoing discussion and refine- cal component of cancer pathogenesis (3). This plasticity
ment of the Hallmarks scheme, it is appropriate to consider can operate in several manifestations (Fig. 2). Thus, nascent
a frequently posed question: are there additional features of cancer cells originating from a normal cell that had advanced
this conceptual model that might be incorporated, respecting down a pathway approaching or assuming a fully differenti-
the need to ensure that they are broadly applicable across the ated state may reverse their course by dedifferentiating back
spectrum of human cancers? Accordingly, I present several to progenitor-like cell states. Conversely, neoplastic cells
prospective new hallmarks and enabling characteristics, ones arising from a progenitor cell that is destined to follow a
that might in due course become incorporated as core com- pathway leading to end-stage differentiation may short-
ponents of the hallmarks of cancer conceptualization. These circuit the process, maintaining the expanding cancer cells in
parameters are “unlocking phenotypic plasticity,” “nonmu- a partially differentiated, progenitor-like state. Alternatively,
tational epigenetic reprogramming,” “polymorphic microbi- transdifferentiation may operate, in which cells that were
omes,” and “senescent cells” (Fig. 1, right). Importantly, the initially committed into one differentiation pathway switch
examples presented in support of these propositions are illus- to an entirely different developmental program, thereby
trative but by no means comprehensive, as there is a growing acquiring tissue-specific traits that were not preordained by
and increasingly persuasive body of published evidence in their normal cells-of-origin. The following examples sup-
support of each vignette. port the argument that differing forms of cellular plasticity,

32 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

Unlocking
phenotypic Progenitor cell Differentiated cell
plasticity
Normal differentiation

Dedifferentiation

Blocked
differentiation

Transdifferentiation

Figure 2.  Unlocking phenotypic plasticity. Left, phenotypic plasticity is arguably an acquired hallmark capability that enables various disruptions of
cellular differentiation, including (i) dedifferentiation from mature to progenitor states, (ii) blocked (terminal) differentiation from progenitor cell states,
and (iii) transdifferentiation into different cell lineages. Right, depicted are three prominent modes of disrupted differentiation integral to cancer patho-
genesis. By variously corrupting the normal differentiation of progenitor cells into mature cells in developmental lineages, tumorigenesis and malignant
progression arising from cells of origin in such pathways is facilitated.

when taken together, constitute a functionally distinct the strict definition of this provisional hallmark as separable
hallmark capability. and independent.
Another line of evidence involves suppressed expression
Dedifferentiation of the MITF master regulator of melanocyte differentiation,
Colon carcinogenesis exemplifies disrupted differentiation, which is evidently involved in the genesis of aggressive forms
in that there is a teleological necessity for incipient cancer of malignant melanoma. Loss of this developmental TF is
cells to escape from the conveyer belt of terminal differen- associated with the reactivation of neural crest progenitor
tiation and exfoliation, which could in principle occur via genes and the downregulation of genes that characterize
dedifferentiation of not yet irrevocably terminally differenti- fully differentiated melanocytes. The reappearance of the
ated colonic epithelial cells, or via blocked differentiation neural crest genes indicates that these cells revert to the pro-
of progenitor/stem cells in the crypts that spawn these dif- genitor state from which melanocytes arise developmentally.
ferentiating cells. Both differentiated cells and stem cells Moreover, a lineage tracing study of BRAF-induced melano-
have been implicated as cell-of-origin for colon cancer (4–6). mas established mature pigmented melanocytes as the cells
Two developmental transcription factors (TF), the homeobox of origin, which undergo dedifferentiation during the course
of tumorigenesis (9). Of note, the mutant BRAF oncogene,
protein HOXA5 and SMAD4, the latter involved in BMP
which is found in more than half of cutaneous melanomas,
signal transmission, are highly expressed in differentiating
induces hyperproliferation that precedes and hence is mecha-
colonic epithelial cells, and typically lost in advanced colon
nistically separable from the subsequent dedifferentiation
carcinomas, which characteristically express markers of stem
arising from downregulation of MITF. Another study func-
and progenitor cells. Functional perturbations in mouse
tionally implicated upregulation of the developmental TF
models have shown that forced expression of HOXA5 in ATF2, whose characteristic expression in mouse and human
colon cancer cells restores differentiation markers, suppresses melanomas indirectly suppresses MITF1, concomitant with
stem cell phenotypes, and impairs invasion and metastasis, malignant progression of the consequently dedifferentiated
providing a rationale for its characteristic downregulation melanoma cells (10). Conversely, expression in melanomas
(7, 8). SMAD4, by contrast, both enforces differentiation and of mutant forms of ATF2 that fail to repress MITF results in
thereby suppresses proliferation driven by oncogenic WNT well-differentiated melanomas (11).
signaling, revealed by the engineered loss of SMAD4 expres- Additionally, a recent study (12) has associated lineage
sion, providing an explanation for its loss of expression so as dedifferentiation with malignant progression from pancre-
to enable dedifferentiation and, subsequently, WNT-driven atic islet cell neoplasias into metastasis-prone carcinomas;
hyperproliferation (5). Notably, the loss of both of these “dif- these neuroendocrine cells and derivative tumors arise from
ferentiation suppressors” with consequent dedifferentiation a developmental lineage that is distinct from the one gen-
is associated with acquisition of other hallmark capabilities, erating the far larger number of adjacent cells that form
as are other hallmark-inducing regulators, which complicates the exocrine and pancreas and the ductal adenocarcinomas

JANUARY 2022 CANCER DISCOVERY | 33

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

that arise therefrom. Notably, the multistep differentiation Other examples of differentiation modulators involve the
pathway of islet progenitor cells into mature  β cells has been metabolite alpha-ketoglutarate (αKG), a necessary cofactor
thoroughly characterized (13). Comparative transcriptome for a number of chromatin-modifying enzymes, which is
profiling reveals that adenoma-like islet tumors are most sim- demonstrably involved in stimulating certain differentiated
ilar to immature but differentiated insulin-producing β cells, cell states. In pancreas cancer, the tumor suppressor p53
whereas the invasive carcinomas are most similar to embry- stimulates the production of αKG and maintenance of a more
onic islet cell precursors. The progression toward poorly well-differentiated cell state, whereas prototypical loss of p53
differentiated carcinomas involves a first step of dedifferen- function results in reductions in  αKG levels and consequent
tiation that does not initially involve increased proliferation dedifferentiation associated with malignant progression (20).
or reduced apoptosis when compared with the well-differen- In one form of liver cancer, mutation of an isocitrate dehy-
tiated adenomas, both of which rather occur later. Thus, the drogenase gene (IDH1/2) results in the production not of
discrete step of dedifferentiation is not driven by observable differentiation-inducing  αKG but rather a related “onco-
alterations in the hallmark traits of sustained proliferation metabolite,” D-2-hydroxygluterate (D2HG), which has been
and resistance to apoptosis. Rather, upregulation of a miRNA shown to block hepatocyte differentiation from liver progeni-
previously implicated in specifying the islet progenitor state, tor cells by D2HG-mediated repression of a master regulator
one that is downregulated during terminal differentiation of hepatocyte differentiation and quiescence, HNF4a. The
of  β cells, has been shown to orchestrate the observed dedif- D2HG-mediated suppression of HNF4a function elicits a
ferentiation occurring during malignant progression (12). proliferative expansion of the hepatocyte progenitor cells in
the liver, which become susceptible to oncogenic transforma-
Blocked Differentiation tion upon subsequent mutational activation of the KRAS
While the above examples illustrate how suppression of oncogene that drives malignant progression to liver cholan-
differentiation factor expression can facilitate tumorigenesis giocarcinoma (21). Mutant IDH1/2 and their oncometabolite
by enabling more well-differentiated cells to dedifferentiate D2HG are also operative in a variety of myeloid and other
into progenitors, in other cases incompletely differentiated solid tumor types, where D2HG inhibits  αKG-dependent
progenitor cells can suffer regulatory changes that actively dioxygenases necessary for histone and DNA methylation
block their continued advance into fully differentiated, typi- events that mediate alterations in chromatin structure dur-
cally nonproliferative states. ing developmental lineage differentiation, thereby freezing
Acute promyelocytic leukemia (APL) has long been docu- incipient cancer cells in a progenitor state (22, 23).
mented to result from a chromosomal translocation that An additional, related concept is “circumvented differen-
fuses the PML locus with the gene encoding the retinoic tiation,” wherein partially or undifferentiated progenitor/
acid  α  nuclear receptor (RARα). Myeloid progenitor cells stem cells exit the cell cycle and become dormant, residing in
bearing such translocations are evidently unable to continue protective niches, with the potential to reinitiate proliferative
their usual terminal differentiation into granulocytes, result- expansion (24), albeit still with the selective pressure to dis-
ing in cells trapped in a proliferative, promyelocytic progeni- rupt their programmed differentiation in one way or another.
tor stage (14). Proof-of-concept of this scheme comes from
treating cultured APL cells, mouse models of this disease, Transdifferentiation
as well as afflicted patients, with retinoic acid, the ligand of The concept of transdifferentiation has long been rec-
RARα; this therapeutic treatment causes the neoplastic APL ognized by pathologists in the form of tissue metaplasia,
cells to differentiate into ostensibly mature nonproliferating wherein cells of a particular differentiated phenotype mark-
granulocytes, short-circuiting their continuing proliferative edly change their morphology to become clearly recognizable
expansion (14–16). as elements of another tissue, of which one prominent exam-
A variation on this theme involves another form of acute ple is Barrett’s esophagus, where chronic inflammation of
myeloid leukemia, this one carrying the t(8;21) translocation, the stratified squamous epithelium of the esophagus induces
which produces the AML1–ETO fusion protein. This protein transdifferentiation into a simple columnar epithelium that
can, on its own, transform myeloid progenitors, at least in is characteristic of the intestine, thereby facilitating the sub-
part by blocking their differentiation. Therapeutic interven- sequent development of adenocarcinomas, and not the squa-
tion in mouse models and in patients with a pharmaco- mous cell carcinomas that would be anticipated to arise from
logic inhibitor of a chromatin-modifying histone deacetylase this squamous epithelium (3). Now, molecular determinants
(HDAC) causes the myeloid leukemia cells to recommence are revealing mechanisms of transdifferentiation in various
their differentiation into cells with a more mature myeloid cancers, both for cases where gross tissue metaplasia is evi-
cell morphology. Concomitant with this response is a reduc- dent and for others where it is rather more subtle, as the
tion in proliferative capacity, thereby impairing the progres- following examples illustrate.
sion of this leukemia (17, 18). One illuminating case for transdifferentiation as a dis-
A third example, in melanoma, involves a developmental crete event in tumorigenesis involves pancreatic ductal ade-
TF, SOX10, which is normally downregulated during mel- nocarcinoma (PDAC), wherein one of the implicated cells
anocyte differentiation. Gain- and loss-of-function studies of origin, the pancreatic acinar cell, can become transdiffer-
in a zebrafish model of BRAF-induced melanoma have dem- entiated into a ductal cell phenotype during the initiation
onstrated that aberrantly maintained expression of SOX10 of neoplastic development. Two TFs—PTF1a and MIST1—
blocks differentiation of neural progenitor cells into melano- govern, via their expression in the context of self-sustaining,
cytes, enabling BRAF-driven melanomas to form (19). “feed-forward” regulatory loops, the specification and

34 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

maintenance of the differentiated pancreatic acinar cell oncogenic signaling pathway known to drive the neoplastic
state (25). Both of these TFs are frequently downregulated growth of these cells (33). Drug-resistant cancer cells switch,
during neoplastic development and malignant progression via broad epigenetic shifts in specific chromatin domains and
of human and mouse PDAC. Functional genetic studies in the altered accessibility of two superenhancers, to a devel-
mice and cultured human PDAC cells have demonstrated opmentally related but distinct cell type. The newly gained
that experimentally forced expression of PTF1a impairs phenotypic state of the BCC cells enables them to sustain
KRAS-induced transdifferentiation and proliferation, and expression of the WNT oncogenic signaling pathway, which
can also force the redifferentiation of already neoplastic in turn imparts independence from the drug-suppressed HH/
cells into a quiescent acinar cell phenotype (26). Conversely, SMO signaling pathway (34). As might be anticipated from
suppression of PTF1a expression elicits acinar-to-ductal this transdifferentiation, the transcriptome of the cancer cells
metaplasia, namely transdifferentiation, and thereby sensi- shifts from a gene signature reflecting the implicated cell-of-
tizes the duct-like cells to oncogenic KRAS transformation, origin of BCCs, namely the stem cells of hair follicle bulge,
accelerating subsequent development of invasive PDAC to one indicative of the basal stem cells that populate the
(27). Similarly, forced expression of MIST1 in KRAS-express- interfollicular epidermis. Such transdifferentiation to enable
ing pancreas also blocks transdifferentiation and impairs drug resistance is being increasingly documented in different
the initiation of pancreatic tumorigenesis otherwise facili- forms of cancer (35).
tated by the formation of premalignant duct-like (PanIN) Developmental lineage plasticity also appears to be
lesions, whereas genetic deletion of MIST1 enhances their prevalent among the major subtypes of lung carcinomas,
formation and the initiation of KRAS-driven neoplastic that is, neuroendocrine carcinomas [small-cell lung cancer
progression (28). Loss of either PTF1 or MIST1 expression (SCLC)] and adenocarcinomas + squamous cell carcinomas
during tumorigenesis is associated with elevated expression [collectively non–small cell lung cancer (NSCLC)]. Single-
of another developmental regulatory TF, SOX9, which is cell RNA sequencing has revealed remarkably dynamic
normally operative in the specification of ductal cells (27, and heterogeneous interconversion among these subtypes
28). Forced upregulation of SOX9, obviating the need to as well as distinct variations thereof during the stages in
downregulate PTF1a and MIST1, has also been shown to lung tumorigenesis, subsequent malignant progression,
stimulate transdifferentiation of acinar cells into a ductal and responses to therapy (36–38). Thus, rather than the
cell phenotype that is sensitive to KRAS-induced neoplasia simple conceptualization of a pure clonal switch from
(29), implicating SOX9 as a key functional effector of their one lineage into another, these studies paint a much more
downregulation in the genesis of human PDAC. Thus, complex picture, of dynamically interconverting subpopu-
three TFs that regulate pancreatic differentiation can be lations of cancer cells exhibiting characteristics of multiple
variously altered to induce a transdifferentiated state that developmental lineages and stages of differentiation, a
facilitates—in the context of mutational activation of KRAS— sobering realization in regard to lineage-based therapeutic
oncogenic transformation and the initiation of tumorigen- targeting of human lung cancer. Regulatory determinants
esis and malignant progression. of this dynamic phenotypic plasticity are beginning to be
Additional members of the SOX family of chromatin- identified (37, 39, 40).
associated regulatory factors are on the one hand broadly
associated both with cell fate specification and lineage switch- Synopsis
ing in development (30), and on the other with multiple The three classes of mechanism described above highlight
tumor-associated phenotypes (31). Another salient example selective regulators of cellular plasticity that are separable—at
of SOX-mediated transdifferentiation involves a mechanism least in part—from core oncogenic drivers and other hallmark
of therapeutic resistance in prostate carcinomas. In this case, capabilities. Beyond these examples lies a considerable body
loss of the RB and p53 tumor suppressors—whose absence of evidence associating many forms of cancer with disrupted
is characteristic of neuroendocrine tumors—in response to differentiation concomitant with the acquisition of tran-
antiandrogen therapy is necessary but not sufficient for the scriptome signatures and other phenotypes—for example,
frequently observed conversion of well-differentiated pros- histologic morphology—associated with progenitor or stem
tate cancer cells into carcinoma cells that have entered a dif- cell stages observed in the corresponding normal tissue-of-
ferentiation lineage with molecular and histologic features origin or in other more distantly related cell types and line-
of neuroendocrine cells, which notably do not express the ages (41–43). As such, these three subclasses of phenotypic
androgen receptor. In addition to loss of RB and p53, the plasticity—dedifferentiation of mature cells back to progeni-
acquired resistance to antiandrogen therapy requires upregu- tor states, blocked differentiation to freeze developing cells
lated expression of the SOX2 developmental regulatory gene, in progenitor/stem cell states, and transdifferentiation to
which is demonstrably instrumental in inducing transdif- alternative cell lineages—appear to be operative in multiple
ferentiation of the therapy-responsive adenocarcinoma cells cancer types during primary tumor formation, malignant
into derivatives that reside in a neuroendocrine cell state that progression, and/or response to therapy. There are, however,
is refractory to the therapy (32). two conceptual considerations. First, dedifferentiation and
A third example also reveals transdifferentiation as a strat- blocked differentiation are likely intertwined, being indis-
egy employed by carcinoma cells to avoid elimination by tinguishable in many tumor types where the cell-of-origin—
a lineage-specific therapy, in this case involving basal cell differentiated cell or progenitor/stem cell—is either unknown
carcinomas (BCC) of the skin treated with a pharmaco- or alternatively involved. Second, the acquisition or mainte-
logic inhibitor of the Hedgehog-Smoothened (HH/SMO) nance of progenitor cell phenotypes and loss of differentiated

JANUARY 2022 CANCER DISCOVERY | 35

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

Nonmutational
epigenetic reprogramming

Figure 3.  Nonmutational epigenetic reprogramming. Much as during embryogenesis and tissue differentiation and homeostasis, growing evidence
makes the case that instrumental gene-regulatory circuits and networks in tumors can be governed by a plethora of corrupted and co-opted mechanisms
that are independent from genome instability and gene mutation.

features is in most cases an imprecise reflection of the normal including metastatic lesions, and during the development
developmental stage, being immersed in a milieu of other of adaptive resistance to therapy. One result is the now
hallmark-enabling changes in the cancer cell that are not widespread appreciation that mutations in genes that organ-
present in naturally developing cells. In addition, yet another ize, modulate, and maintain chromatin architecture, and
form of phenotypic plasticity involves cell senescence, dis- thereby globally regulate gene expression, are increasingly
cussed more generally below, wherein cancer cells induced detected and functionally associated with cancer hallmarks
to undergo ostensibly irreversible senescence are instead able (46–48).
to escape and resume proliferative expansion (44). Finally, There is, in addition, a case to be made for another appar-
as with other hallmark capabilities, cellular plasticity is not ently independent mode of genome reprogramming that
a novel invention or aberration of cancer cells, but rather involves purely epigenetically regulated changes in gene
the corruption of latent but activatable capabilities that expression, one that might be termed “nonmutational epi-
various normal cells use to support homeostasis, repair, and genetic reprogramming” (Fig.  3). Indeed, the proposition
regeneration (45). of mutation-less cancer evolution and purely epigenetic
Collectively, these illustrative examples encourage consid- programming of hallmark cancer phenotypes was raised
eration of the proposition that unlocking cellular plasticity almost a decade ago (49) and is increasingly discussed
to enable various forms of disrupted differentiation consti- (46, 50–52).
tutes a discrete hallmark capability, distinguishable in regu- The concept of nonmutational epigenetic regulation of
lation and cellular phenotype from the well-validated core gene expression is of course well established as the central
hallmarks of cancer (Fig. 2). mechanism mediating embryonic development, differentia-
tion, and organogenesis (53–55). In the adult, for example,
long-term memory involves changes in gene and histone
NONMUTATIONAL EPIGENETIC
modification, in chromatin structure, and in the triggering of
REPROGRAMMING gene expression switches that are stably maintained over time
The enabling characteristic of genome (DNA) instabil- by positive and negative feedback loops (56, 57). Growing
ity and mutation is a fundamental component of cancer evidence supports the proposition that analogous epigenetic
formation and pathogenesis. At present, multiple interna- alterations can contribute to the acquisition of hallmark
tional consortia are cataloging mutations across the genome capabilities during tumor development and malignant pro-
of human cancer cells, doing so in virtually every type of gression. A few examples are presented below in support of
human cancer, at different stages of malignant progression, this hypothesis.

36 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

Microenvironmental Mechanisms within, has broad effects on the invasive and other phe-
of Epigenetic Reprogramming notypic characteristics of cancer cells. Compared with the
If not solely by consequence of oncogenic mutations, how normal tissue ECM from which tumors originate, the tumor
then is the cancer cell genome reprogrammed? A growing ECM is typically characterized by increased cross-linking and
body of evidence indicates that the aberrant physical proper- density, enzymatic modifications, and altered molecular com-
position, which collectively orchestrate—in part via integrin
ties of the tumor microenvironment can cause broad changes
receptors for ECM motifs—stiffness-induced signaling and
in the epigenome, from which changes beneficial to the phe-
gene-expression networks that elicit invasiveness and other
notypic selection of hallmark capabilities can result in clonal
hallmark characteristics (71).
outgrowth of cancer cells with enhanced fitness for prolif-
In addition to such regulatory mechanisms endowed by
erative expansion. One common characteristic of tumors (or
the physical tumor microenvironment, paracrine signaling
regions within tumors) is hypoxia, consequent to insufficient
involving soluble factors released into the extracellular milieu
vascularization. Hypoxia, for example, reduces the activity of
by the various cell types populating solid tumors can also con-
the TET demethylases, resulting in substantive changes in
tribute to the induction of several morphologically distinct
the methylome, in particular hypermethylation (58). Insuf-
invasive growth programs (72), only one of which—dubbed
ficient vascularization likely also limits the bioavailability of
“mesenchymal”—seems to involve the aforementioned EMT
critical blood-borne nutrients, and nutrient deprivation has
epigenetic regulatory mechanism.
been shown for example to alter translational control and
consequently enhance the malignant phenotype of breast Epigenetic Regulatory Heterogeneity
cancer cells (59).
A growing knowledge base is heightening appreciation of
A persuasive example of hypoxia-mediated epigenetic regu-
the importance of intratumoral heterogeneity in generating
lation involves a form of invariably lethal pediatric epend- the phenotypic diversity where the fittest cells for prolif-
ymoma. Like many embryonic and pediatric tumors, this erative expansion and invasion outgrow their brethren and
form lacks recurrent mutations, in particular a dearth of hence are selected for malignant progression. Certainly, one
driver mutations in oncogenes and tumor suppressors. facet of this phenotypic heterogeneity is founded in chronic
Rather, the aberrant growth of these cancer cells is demon- or episodic genomic instability and consequent genetic het-
strably governed by a gene regulatory program induced by erogeneity in the cells populating a tumor. In addition, it
hypoxia (60, 61). Notably, the putative cell-of-origin of this is increasingly evident that there can be non–mutationally
cancer resides in a hypoxic compartment, likely sensitizing based epigenetic heterogeneity. A salient example involves
cells resident therein to the initiation of tumorigenesis by as the linker histone H1.0, which is dynamically expressed and
yet unknown cofactors. repressed in subpopulations of cancer cells within a number
Another persuasive line of evidence for microenvironmen- of tumor types, with consequent sequestration or accessibil-
tally mediated epigenetic regulation involves the invasive ity, respectively, of megabase-sized domains, including ones
growth capability of cancer cells. A classic example involves conveying hallmark capabilities (73). Notably, the population
the reversible induction of invasiveness of cancer cells at the of cancer cells with repressed H1.0 were found to have stem-
margins of many solid tumors, orchestrated by the devel- like characteristics, enhanced tumor-initiating capability, and
opmental regulatory program known as the epithelial-to- an association with poor prognosis in patients.
mesenchymal transition (EMT; refs. 62–64). Notably, a mas- Another example of epigenetically regulated plasticity has
ter regulator of the EMT, ZEB1, has been recently shown to been described in human oral squamous cell carcinomas
induce expression of a histone methyltransferase, SETD1B, (SCC), wherein cancer cells at the invasive margins adopt a
that in turn sustains ZEB1 expression in a positive feedback partial EMT (p-EMT) state lacking the aforementioned mes-
loop that maintains the (invasive) EMT regulatory state enchymal TFs but expressing other EMT-defining genes that
(65). A previous study similarly documented that induction are not expressed in the central core of the tumors (74). The
of EMT by upregulated expression of a related TF, SNAIL1, p-EMT cells evidently do not represent a clonal compartmen-
caused marked alterations in the chromatin landscape con- talization of mutationally altered cells: cultures of primary
sequent to induction of a number of chromatin modifiers, tumor-derived cancer cells contain dynamic mixtures of both
whose activity was demonstrably necessary for the mainte- p-EMThi and p-EMTlo cells, and when p-EMThi/lo cells were
nance of the phenotypic state (66). Furthermore, a roster of FACS-purified and cultured, both reverted to mixed popula-
conditions and factors to which cancer cells at the margins of tions of p-EMThi and p-EMTlo cells within 4 days. Moreover,
tumors are exposed, including hypoxia and cytokines secreted although paracrine signals from the adjacent stroma could
by stromal cells, can evidently induce the EMT and in turn be envisaged as deterministic for the p-EMThi state, the sta-
invasiveness (67, 68). ble presence and regeneration of the two epigenetic states in
A distinctive example of microenvironmental program- culture argues for a cancer cell–intrinsic mechanism. Notably,
ming of invasiveness, ostensibly unrelated to the EMT pro- this conclusion is supported by analysis of 198 cell lines rep-
gram, involves autocrine activation, in pancreas cancer cells resenting 22 cancer types, including SCC, wherein 12 stably
and others, via interstitial pressure–driven fluid flow, of a heterogeneous epigenetic states (including the p-EMT in
neuronal signaling circuit involving secreted glutamate and SCC) were variously detected in the cell line models as well
its receptor NMDAR (69, 70). Notably, the prototypical stiff- as their cognate primary tumors (75). Again, the heteroge-
ness of many solid tumors, embodied in extensive alterations neous phenotypic states could not be linked to detectable
to the extracellular matrix (ECM) that envelop the cells genetic differences, and in several cases FACS-sorted cells of

JANUARY 2022 CANCER DISCOVERY | 37

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

a particular state were shown to dynamically reequilibrate and mutation. Notably, it can be anticipated that nonmuta-
upon culture, recapitulating a stable balance among the het- tional epigenetic reprogramming will prove to be integrally
erogeneous states seen in the original cell lines. involved in enabling the provisional new hallmark capability
Additionally, technologies for genome-wide profiling of of phenotypic plasticity discussed above, in particular being
diverse attributes—beyond DNA sequence and its mutational a driving force in the dynamic transcriptomic heterogeneity
variation—are illuminating influential elements of the cancer that is increasingly well documented in cancer cells populat-
cell genome’s annotation and organization that correlate ing malignant TMEs. The advance of single cell multi-omic
with patient prognosis, and increasingly with hallmark capa- profiling technologies is envisaged to illuminate the respec-
bilities (76–78). Epigenomic heterogeneity is being revealed tive contributions of and interplay between mutation-driven
by increasingly powerful technologies for profiling genome- versus nonmutational epigenetic regulation to the evolution
wide DNA methylation (79, 80), histone modification (81), of tumors during malignant progression and metastasis.
chromatin accessibility (82), and posttranscriptional modifi-
cation and translation of RNA (83, 84). A challenge in regard
to the postulate being considered herein will be to ascertain
POLYMORPHIC MICROBIOMES
which epigenomic modifications in particular cancer types An expansive frontier in biomedicine is unfolding via
(i) have regulatory significance and (ii) are representative of illumination of the diversity and variability of the plethora
purely nonmutational reprogramming, as opposed to being of microorganisms, collectively termed the microbiota, that
mutation-driven and thus explainable by genome instability. symbiotically associate with the barrier tissues of the body
exposed to the external environment—the epidermis and the
Epigenetic Regulation of the Stromal Cell Types internal mucosa, in particular the gastrointestinal tract, as
Populating the Tumor Microenvironment well as the lung, the breast, and the urogenital system. There
In general, the accessory cells in the tumor microenvi- is growing appreciation that the ecosystems created by resi-
ronment that functionally contribute to the acquisition dent bacteria and fungi—the microbiomes—have profound
of hallmark capabilities are not thought to suffer genetic impact on health and disease (87), a realization fueled by
instability and mutational reprogramming to enhance their the capability to audit the populations of microbial species
tumor-promoting activities; rather it is inferred that these using next-generation sequencing and bioinformatic tech-
cells—cancer-associated fibroblasts, innate immune cells, and nologies. For cancer, the evidence is increasingly compelling
endothelial cells and pericytes of the tumor vasculature— that polymorphic variability in the microbiomes between
are epigenetically reprogrammed upon their recruitment by individuals in a population can have a profound impact on
soluble and physical factors that define the solid tumor micro- cancer phenotypes (88, 89). Association studies in human
environment (2, 85). It can be anticipated the multi-omic pro- and experimental manipulation in mouse models of can-
filing technologies currently being applied to cancer cells will cer are revealing particular microorganisms, principally but
increasingly be used to interrogate the accessory (stromal) not exclusively bacteria, which can have either protective
cells in tumors to elucidate how normal cells are corrupted or deleterious effects on cancer development, malignant
to functionally support tumor development and progression. progression, and response to therapy. So too can the global
For example, a recent study (86) suggests that such reprogram- complexity and constitution of a tissue microbiome at large.
ming can involve modifications of the epigenome in addition Indeed, while the gut microbiome has been the pioneer of
to the inductive interchange of cytokines, chemokines, and this new frontier, multiple tissues and organs have associ-
growth factors that alter intracellular signaling networks in ated microbiomes, which have distinctive characteristics in
all of these cell types: when mouse models of metastasis to regard to population dynamics and diversity of microbial
lung were treated with a combination of a DNA methyltrans- species and subspecies. This growing appreciation of the
ferase inhibitor (5-azacytidine) and an inhibitor of histone importance of polymorphically variable microbiomes in
modification (an HDAC), the infiltrating myeloid cells were health and disease posits the question: is the microbiome
found to have switched from an immature (tumor-promot- a discrete enabling characteristic that broadly affects, both
ing) progenitor state into cells resembling mature interstitial positively and negatively, the acquisition of hallmark capa-
(tumor-antagonizing) macrophages, which, in contrast to bilities for cancer? I reflect on this possibility below, illustrat-
their counterparts in untreated tumors, were incapable of ing evidence for some of the prominent tissue microbiomes
supporting the hallmark capabilities necessary for efficient implicated in cancer hallmarks (Fig.  4), beginning with the
metastatic colonization (86). It can be envisaged that multi- most prominent and evidently impactful microbiome, that
omic profiling and pharmacologic perturbation will serve to of the intestinal tract.
elucidate the reprogrammed epigenetic state in such myeloid
cells as well as other hallmark-enabling accessory cell types
populating tumor microenvironments. Diverse Modulatory Effects of the Gut Microbiome
It has long been recognized that the gut microbiome is fun-
Synopsis damentally important for the function of the large intestine
Collectively, these illustrative snapshots support the prop- (colon) in degrading and importing nutrients into the body
osition that nonmutational epigenetic reprograming will as part of metabolic homeostasis, and that distortions in the
come to be accepted as a bona fide enabling characteristic microbial populations—dysbiosis—in the colon can cause a
that serves to facilitate the acquisition of hallmark capa- spectrum of physiologic maladies (87). Among these has
bilities (Fig. 3), distinct from that of genomic DNA instability been the suspicion that the susceptibility, development, and

38 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

Gut

Skin Lung
Modulating
tumor
Growth
Inflammation
Immune
evasions
Genome
instability
Vaginal/ Therapy
Oral resistance
cervical

Polymorphic
microbiomes Tumor

Figure 4.  Polymorphic microbiomes. Left, while intersecting with the enabling characteristics of tumor-promoting inflammation and genomic instabil-
ity and mutation, there is growing reason to conclude that polymorphic microbiomes in one individual versus another, being resident in the colon, other
mucosa and connected organs, or in tumors themselves, can diversely influence—by either inducing or inhibiting—many of the hallmark capabilities,
and thus are potentially an instrumental and quasi-independent variable in the puzzle of how cancers develop, progress, and respond to therapy. Right,
multiple tissue microbiomes are implicated in modulating tumor phenotypes. In addition to the widely studied gut microbiome, other distinctive tissue
microbiomes, as well as the tumor microbiome, are implicated in modulating the acquisition—both positively and negatively—of the illustrated hallmark
capabilities in certain tumor types.

pathogenesis of colon cancer is influenced by the gut micro- bacteria; the connection between butyrate-induced senes-
biome. In recent years, persuasive functional studies, involv- cence and enhanced colon tumorigenesis was demonstrated
ing fecal transplants from colon tumor–bearing patients and by the use of a senolytic drug that kills senescent cells,
mice into recipient mice predisposed to develop colon cancer which impaired tumor growth (92). In addition, bacterial-
has established a principle: there are both cancer-protective produced butyrate has pleiotropic and paradoxical effects on
and tumor-promoting microbiomes, involving particular differentiated cells versus undifferentiated (stem) cells in the
bacterial species, which can modulate the incidence and colonic epithelium in conditions where the intestinal barrier
pathogenesis of colon tumors (90). is disrupted (dysbiosis) and the bacteria are invasive, affect-
The mechanisms by which microbiota impart these modu- ing, for example, cellular energetics and metabolism, histone
latory roles are still being elucidated, but two general effects modification, cell-cycle progression, and (tumor-promoting)
are increasingly well established for tumor-promoting micro- innate immune inflammation that is immunosuppressive of
biomes and in some cases for specific tumor-promoting adaptive immune responses (93).
bacterial species. The first effect is mutagenesis of the colonic Indeed, a broad effect of polymorphic microbiomes
epithelium, consequent to the production of bacterial tox- involves the modulation of the adaptive and innate immune
ins and other molecules that either damage DNA directly, systems via multifarious routes, including the production by
or disrupt the systems that maintain genomic integrity, or bacteria of “immunomodulatory” factors that activate dam-
stress cells in other ways that indirectly impair the fidel- age sensors on epithelial or resident immune cells, resulting
ity of DNA replication and repair. A case in point is E. coli in the expression of a diverse repertoire of chemokines and
carrying the PKS locus, which demonstrably mutagenizes cytokines that can sculpt the abundance and characteristics
the human genome and is implicated in conveying hallmark- of immune cells populating the colonic epithelia and its
enabling mutations (91). underlying stroma and draining lymph nodes. In addition,
Additionally, bacteria have been reported to bind to the certain bacteria can breach both the protective biofilm and
surface of colonic epithelial cells and produce ligand mimet- the mucus lining the colonic epithelia and proceed to dis-
ics that stimulate epithelial proliferation, contributing in rupt the epithelial cell–cell tight junctions that collectively
neoplastic cells to the hallmark capability for proliferative maintain the integrity of the physical barrier that normally
signaling (88). Another mechanism by which specific bacterial compartmentalizes the intestinal microbiome. Upon invad-
species promote tumorigenesis involves butyrate-producing ing the stroma, bacteria can trigger both innate and adap-
bacteria, whose abundance is elevated in patients with colo- tive immune responses, eliciting secretion of a repertoire of
rectal cancer (92). The production of the metabolite butyrate cytokines and chemokines. One manifestation can be the
has complex physiologic effects, including the induction of creation of tumor-promoting or tumor-antagonizing immune
senescent epithelial and fibroblastic cells. A mouse model microenvironments, consequently protecting against or
of colon carcinogenesis populated with butyrate-producing facilitating tumorigenesis and malignant progression. Con-
bacteria developed more tumors than mice lacking such cordantly, the modulation by distinctive microbiomes in

JANUARY 2022 CANCER DISCOVERY | 39

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

individual patients of the intertwined parameters of (i) elicit- organ/tissue-specific differences in the constitution of the
ing (innate) tumor promoting inflammation and (ii) escap- associated microbiomes in homeostasis, aging, and cancer,
ing (adaptive) immune destruction can be associated not with both overlapping and distinctive species and abundan-
only with prognosis, but also with responsiveness or resist- cies to that of the colon (104, 105). Moreover, association
ance to immunotherapies involving immune checkpoint studies are providing increasing evidence that local tumor-
inhibitors and other therapeutic modalities (89, 94–96). antagonizing/protective versus tumor-promoting tissue
Provisional proof-of-concept has come from recent stud- microbiomes, similarly to the gut microbiome, can modulate
ies demonstrating restored efficacy to immunotherapy susceptibility and pathogenesis to human cancers arising in
following transplants of fecal microbiota from therapy- their associated organs (106–109).
responsive patients into patients with melanoma who had
progressed during prior treatment with immune checkpoint Impact of Intratumoral Microbiota?
blockade (97, 98). Finally, pathologists have long recognized that bacteria can
An ongoing mystery has involved the molecular mecha- be detected within solid tumors, an observation that has now
nisms by which particular and variable constituents of the been substantiated with sophisticated profiling technologies.
gut microbiome systemically modulate the activity of the For example, in a survey of 1,526 tumors encompassing seven
adaptive immune system, either enhancing antitumoral human cancer types (bone, brain, breast, lung, melanoma,
immune responses evoked by immune checkpoint blockade, ovary, and pancreas), each type was characterized by a distinc-
or rather eliciting systemic or local (intratumoral) immu- tive microbiome that was largely localized inside cancer cells
nosuppression. A recent study has shed some light: certain and immune cells, and within each tumor type, variations in
strains of Enterococcus (and other bacteria) express a pepti- the tumor microbiome could be detected and inferred to be
doglycan hydrolyase called SagA that releases mucopeptides associated with clinicopathologic features (110). Microbiota
from the bacterial wall, which can then circulate systemi- have been similarly detected in genetically engineered de novo
cally and activate the NOD2 pattern receptor, which in turn mouse models of lung and pancreas cancer, and their absence
can enhance T-cell responses and the efficacy of checkpoint in germ-free mice and/or their abrogation with antibiotics can
immunotherapy (99). Other immunoregulatory molecules demonstrably impair tumorigenesis, functionally implicating
produced by specific bacterial subspecies are being identi-
the tumor microbiome as an enabler of tumor-promoting
fied and functionally evaluated, including bacteria-produced
inflammation and malignant progression (111, 112). Associa-
inosine, a rate-limiting metabolite for T-cell activity (100).
tion studies in human pancreatic ductal adenocarcinoma and
These examples and others are beginning to chart the molec-
functional tests via fecal transplants into tumor-bearing mice
ular mechanisms by which polymorphic microbiomes are
have established that variations in the tumor microbiome—
indirectly and systemically modulating tumor immunobiol-
and the associated gut microbiome—modulate immune phe-
ogy, above and beyond immune responses consequent to
notypes and survival (113). An important challenge for the
direct physical interactions of bacteria with the immune
future will be to extend these implications to other tumor
system (101, 102).
types, and to delineate the potentially separable contribu-
Beyond the causal links to colon cancer and melanoma, the
tions of constitution and variation in the tumor microbiome
gut microbiome’s demonstrable ability to elicit the expres-
to that of the gut (and local tissue of origin) microbiome,
sion of immunomodulatory chemokines and cytokines that
enter the systemic circulation is evidently also capable of potentially by identifying specific microbial species that are
affecting cancer pathogenesis and response to therapy in functionally influential in one location or the other.
other organs of the body (94, 95). An illuminating example Synopsis
involves the development of cholangiocarcinomas in the
liver: gut dysbiosis allows the entry and transport of bacteria Among the fascinating questions for the future is whether
and bacterial products through the portal vein to the liver, microbiota resident in different tissues or populating incipi-
where TLR4 expressed on hepatocytes is triggered to induce ent neoplasias have the capability to contribute to or interfere
expression of the chemokine CXCL1, which recruits CXCR2- with the acquisition of other hallmark capabilities beyond
expressing granulocytic myeloid cells (gMDSC) that serve to immunomodulation and genome mutation, thereby influ-
suppress natural killer cells so as to evade immune destruc- encing tumor development and progression. There are clues
tion (103), and likely convey other hallmark capabilities (85). that particular bacterial species can directly stimulate the
As such, the gut microbiome is unambiguously implicated as hallmark of proliferative signaling, for example, in colonic
an enabling characteristic that can alternatively facilitate or epithelium (88), and modulate growth suppression by alter-
protect against multiple forms of cancer. ing tumor suppressor activity in different compartments of
the intestine (114), whereas direct effects on other hallmark
Beyond the Gut: Implicating Distinctive capabilities, such as avoiding cell death, inducing angiogene-
Microbiomes in Other Barrier Tissues sis, and stimulating invasion and metastasis, remain obscure,
Virtually all tissues and organs exposed, directly or indi- as does the generalizability of these observations to multiple
rectly, to the outside environment are also repositories for com- forms of human cancer. Irrespective, there is an increasingly
mensal microorganisms (104). Unlike the intestine, where the compelling case to be made that polymorphic variation in
symbiotic role of the microbiome in metabolism is well recog- microbiomes of the intestine and other organs constitutes
nized, the normal and pathogenic roles of resident microbiota a distinctive enabling characteristic for the acquisition of
in these diverse locations is still emerging. There are evidently hallmark capabilities (Fig.  4), albeit intersecting with and

40 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

complementing those of genome instability and mutation, from their SASP-expressing, nonproliferative condition, and
and tumor-promoting inflammation. resume cell proliferation and manifestation of the associ-
ated capabilities of fully viable oncogenic cells (44). Such
transitory senescence is most well documented in cases of
SENESCENT CELLS
therapy resistance (44), representing a form of dormancy
Cellular senescence is a typically irreversible form of pro- that circumvents therapeutic targeting of proliferating can-
liferative arrest, likely evolved as a protective mechanism for cer cells, but may well prove to be more broadly operative in
maintaining tissue homeostasis, ostensibly as a complemen- other stages of tumor development, malignant progression,
tary mechanism to programmed cell death that serves to and metastasis.
inactivate and in due course remove diseased, dysfunctional, Moreover, the hallmark-promoting capabilities of senes-
or otherwise unnecessary cells. In addition to shutting down cent cells are not limited to senescent cancer cells. Cancer-
the cell division cycle, the senescence program evokes changes associated fibroblasts (CAF) in tumors have been shown to
in cell morphology and metabolism and, most profoundly, undergo senescence, creating senescent CAFs that are demon-
the activation of a senescence-associated secretory phenotype strably tumor-promoting by virtue of conveying hallmark
(SASP) involving the release of a plethora of bioactive pro- capabilities to cancer cells in the TME (115, 116, 121). More-
teins, including chemokines, cytokines, and proteases whose over, senescent fibroblasts in normal tissues produced in
identity is dependent on the cell and tissue type from which a part by natural aging or environmental insults have similarly
senescent cell arises (115–117). Senescence can be induced in been implicated in remodeling tissue microenvironments via
cells by a variety of conditions, including microenvironmen- their SASP so as to provide paracrine support for local inva-
tal stresses such as nutrient deprivation and DNA damage, as sion (so-called “field effects”) and distant metastasis (116) of
well as damage to organelles and cellular infrastructure, and neoplasias developing in proximity. Additionally, senescent
imbalances in cellular signaling networks (115, 117), all of fibroblasts in aging skin have been shown to recruit—via their
which have been associated with the observed increase in the SASP—innate immune cells that are both immunosuppres-
abundance of senescent cells in various organs during aging sive of adaptive antitumoral immune responses anchored
(118, 119). by CD8 T cells, and stimulatory of skin tumor growth (123),
Cellular senescence has long been viewed as a protective with the latter effect potentially reflecting paracrine contribu-
mechanism against neoplasia, whereby cancerous cells are tions of such innate immune cells (myeloid cells, neutrophils,
induced to undergo senescence (120). Most of the afore- and macrophages) to other hallmark capabilities.
mentioned instigators of the senescent program are asso- While less well established, it seems likely that other abun-
ciated with malignancy, in particular DNA damage as a dant stromal cells populating particular tumor microen-
consequence of aberrant hyperproliferation, so-called onco- vironments will prove to undergo senescence, and thereby
gene-induced senescence due to hyperactivated signaling, modulate cancer hallmarks and consequent tumor phe-
and therapy-induced senescence consequent to cellular and notypes. For example, therapy-induced senescent tumor
genomic damage caused by chemotherapy and radiotherapy. endothelial cells can enhance proliferation, invasion, and
Indeed, there are well-established examples of the protective metastasis in breast cancer models (124, 125).
benefits of senescence in limiting malignant progression (118,
Certainly, such clues warrant investigation in other tumor
119). To the contrary, however, an increasing body of evidence
types to assess generality of fibroblastic, endothelial, and
reveals quite the opposite: in certain contexts, senescent cells
other stromal cell senescence as a driving force in tumor
variously stimulate tumor development and malignant pro-
evolution. Also currently unresolved are the regulatory
gression (119, 121). In one illuminating case study, senescent
mechanisms and functional determinants through which
cells were pharmacologically ablated in aging mice, in par-
a particular senescent cell type in a given TME evokes a
ticular depleting senescent cells characteristically expressing
tumor-promoting versus a tumor-antagonizing SASP, which
the cell-cycle inhibitor p16−INK4a: in addition to delaying mul-
can seeming be alternatively induced in the same senescing
tiple age-related symptoms, the depletion of senescent cells
cell type, perhaps by different instigators when immersed in
in aging mice resulted in reduced incidences of spontaneous
distinctive physiologic and neoplastic microenvironments.
tumorigenesis and cancer-associated death (122).
The principal mechanism by which senescent cells pro-
mote tumor phenotypes is thought to be the SASP, which is Synopsis
demonstrably capable of conveying, in paracrine fashion to The concept that tumors are composed of genetically
viable cancer cells in proximity, as well as to other cells in the transformed cancer cells interacting with and benefiting from
TME, signaling molecules (and proteases that activate and/ recruited and epigenetically/phenotypically corrupted acces-
or desequester them) so as to convey hallmark capabilities. sory (stromal) cells is well established as instrumental to the
Thus, in different experimental systems, senescent cancer pathogenesis of cancer. The considerations discussed above
cells have been shown to variously contribute to prolifera- and described in the reviews and reports cited herein (and
tive signaling, avoiding apoptosis, inducing angiogenesis, elsewhere) make a persuasive case for the proposition that
stimulating invasion and metastasis, and suppressing tumor senescent cells (of whatever cellular origin) should be consid-
immunity (116, 118, 120, 121). ered for addition to the roster of functionally significant cells
Yet another facet to the effects of senescent cancer cells in the tumor microenvironment (Fig.  5). As such, senescent
on cancer phenotypes involves transitory, reversible senes- cells warrant being factored into the quest for deep knowl-
cent cell states, whereby senescent cancer cells can escape edge of cancer mechanisms. Furthermore, the realization of

JANUARY 2022 CANCER DISCOVERY | 41

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

Senescent cells
(multiple origins)

Senescent cells

Figure 5.  Senescent cells. Heterogeneous cancer cell subtypes as well as stromal cell types and subtypes are functionally integrated into the manifes-
tations of tumors as outlaw organs. Clues are increasingly implicating senescent cell derivatives of many of these cellular constituents of the TME, and
their variable SASPs, in modulating hallmark capabilities and consequent tumor phenotypes.

their importance motivates the ancillary goal to therapeuti- (ii) MYC (https://cancer.sanger.ac.uk/cosmic/census-page/
cally target tumor-promoting senescent cells of all constitu- MYC),
tions, be it by pharmacologic or immunologic ablation, or by (iii) NOTCH (https://cancer.sanger.ac.uk/cosmic/census-
reprogramming the SASP into tumor-antagonizing variants page/NOTCH1; ref. 127), and
(115, 121, 126). (iv) TP53 (https://cancer.sanger.ac.uk/cosmic/census-page/
TP53),
CONCLUDING REMARKS highlighting the important challenge to more fully elu-
cidate the regulatory networks governing these acquired
While the eight hallmarks of cancer and their two ena-
capabilities.
bling characteristics have proved of enduring heuristic value in
In addition to adding cellular plasticity to the roster,
the conceptualization of cancer, the considerations presented
nonmutational epigenetic reprogramming and polymorphic
above suggest that there may be new facets of some general-
variations in organ/tissue microbiomes may come to be incor-
ity and hence of relevance to more fully understanding the
porated as mechanistic determinants—enabling characteris-
complexities, mechanisms, and manifestations of the disease.
tics—by which hallmark capabilities are acquired, along with
By applying the metric of discernable if not complete inde- tumor-promoting inflammation (itself partially intercon-
pendence from the 10 core attributes, it is arguable that these nected to the microbiome), above and beyond the mutations
four parameters may well—pursuant to further validation and and other aberrations that manifest the afore-mentioned
generalization beyond the case studies presented—become inte- oncogenic drivers.
grated into the hallmarks of cancer schematic (Fig. 6). Thus, cel- Finally, senescent cells of different origins—including can-
lular plasticity may come to be added to the roster of hallmark cer cells and various stromal cells—that functionally contrib-
capabilities. Notably, while the eight core and this nouveau ute to the development and malignant progression of cancer,
capability are each, by their definition as a hallmark, concep- albeit in markedly distinctive ways to those of their nonsenes-
tually distinguishable, aspects of their regulation are at least cent brethren, may become incorporated as generic compo-
partially interconnected in some and perhaps many cancers. nents of the TME. In conclusion, it is envisaged that raising
For example, multiple hallmarks are coordinately modulated these provisional “trial balloons” will stimulate debate, dis-
in some tumor types by canonical oncogenic drivers, including cussion, and continuing experimental investigation in the
(i) KRAS (https://cancer.sanger.ac.uk/cosmic/census-page/ cancer research community about the defining conceptual
KRAS), parameters of cancer biology, genetics, and pathogenesis.

42 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

Sustaining Evading
proliferative signaling growth suppressors

Unlocking Nonmutational
phenotypic plasticity epigenetic reprogramming

Deregulating Avoiding immune


cellular destruction
metabolism

Resisting cell Enabling


death replicative
immortality

Genome
Tumor-promoting
instability &
inflammation
mutation

Polymorphic
Senescent cells
microbiomes

Inducing or accessing Activating invasion &


vasculature metastasis

Figure 6.  Hallmarks of Cancer—new additions. Depicted are the canonical and prospective new additions to the “Hallmarks of Cancer.” This treatise
raises the possibility, aiming to stimulate debate, discussion, and experimental elaboration, that some or all of the four new parameters will come to be
appreciated as generic to multiple forms of human cancer and hence appropriate to incorporate into the core conceptualization of the hallmarks of cancer.

Author’s Disclosures 5. Perekatt AO, Shah PP, Cheung S, Jariwala N, Wu A, Gandhi V,


et  al. SMAD4 suppresses WNT-driven dedifferentiation and onco-
No disclosures were reported. genesis in the differentiated gut epithelium. Cancer Res 2018;78:
4878–90.
Acknowledgments 6. Shih IM, Wang TL, Traverso G, Romans K, Hamilton SR, Ben-Sasson S,
First and foremost, I profoundly thank Bob Weinberg for an et  al. Top-down morphogenesis of colorectal tumors. Proc Natl
exceptional tradition of insightful and formative discussions, and Acad Sci U S A 2001;98:2640–5.
for excellent comments and suggestions to the first vignette of this 7. Ordóñez-Morán P, Dafflon C, Imajo M, Nishida E, Huelsken J.
manuscript. Additionally, I wish to thank: Ben Stanger; Bradley HOXA5 counteracts stem cell traits by inhibiting Wnt signaling in
colorectal cancer. Cancer Cell 2015;28:815–29.
Bernstein, Giovanni Ciriello, and William Flavahan; Jennifer Wargo;
8. Tan SH, Barker N. Stemming colorectal cancer growth and metas-
and Sheila Stewart for their valuable comments and suggestions
tasis: HOXA5 forces cancer stem cells to differentiate. Cancer Cell
on the four vignettes, respectively, and SayoStudio for assistance in 2015;28:683–5.
crafting the figures. 9. Köhler C, Nittner D, Rambow F, Radaelli E, Stanchi F, Vandamme N,
et  al. Mouse cutaneous melanoma induced by mutant BRaf arises
Received August 4, 2021; revised November 17, 2021; accepted from expansion and dedifferentiation of mature pigmented mel-
November 18, 2021; published first January 12, 2022. anocytes. Cell Stem Cell 2017;21:679–93.
10. Shah M, Bhoumik A, Goel V, Dewing A, Breitwieser W, Kluger H,
et al. A role for ATF2 in regulating MITF and melanoma develop-
REFERENCES ment. PLoS Genet 2010;6:e1001258.
11. Claps G, Cheli Y, Zhang T, Scortegagna M, Lau E, Kim H, et al. A
1. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100: transcriptionally inactive ATF2 variant drives melanomagenesis.
57–70. Cell Rep 2016;15:1884–92.
2. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. 12. Saghafinia S, Homicsko K, Di Domenico A, Wullschleger S,
Cell 2011;144:646–74. Perren A, Marinoni I, et  al. Cancer cells retrace a stepwise differ-
3. Yuan S, Norgard RJ, Stanger BZ. Cellular plasticity in cancer. Cancer entiation program during malignant progression. Cancer Discov
Discov 2019;9:837–51. 2021;11:2638–57.
4. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, 13. Yu X-X, Qiu W-L, Yang L, Zhang Y, He M-Y, Li L-C, et al. Defining
van den Born M, et al. Crypt stem cells as the cells-of-origin of intes- multistep cell fate decision pathways during pancreatic develop-
tinal cancer. Nature 2009;457:608–11. ment at single-cell resolution. EMBO J 2019;38:e100164.

JANUARY 2022 CANCER DISCOVERY | 43

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

14. de Thé H. Differentiation therapy revisited. Nat Rev Cancer 2018; 37. LaFave LM, Kartha VK, Ma S, Meli K, Del Priore I, Lareau C, et al.
18:117–27. Epigenomic state transitions characterize tumor progression in
15. He LZ, Merghoub T, Pandolfi PP. In vivo analysis of the molecular mouse lung adenocarcinoma. Cancer Cell 2020;38:212–28.
pathogenesis of acute promyelocytic leukemia in the mouse and its 38. Marjanovic ND, Hofree M, Chan JE, Canner D, Wu K, Trakala M,
therapeutic implications. Oncogene 1999;18:5278–92. et  al. Emergence of a high-plasticity cell state during lung cancer
16. Warrell RP, de Thé H, Wang ZY, Degos L. Acute promyelocytic leu- evolution. Cancer Cell 2020;38:229–46.
kemia. N Engl J Med 1993;329:177–89. 39. Drapkin BJ, Minna JD. Studying lineage plasticity one cell at a time.
17. Bots M, Verbrugge I, Martin BP, Salmon JM, Ghisi M, Baker A, et al. Cancer Cell 2020;38:150–2.
Differentiation therapy for the treatment of t(8;21) acute myeloid leu- 40. Inoue Y, Nikolic A, Farnsworth D, Liu A, Ladanyi M, Somwar  R,
kemia using histone deacetylase inhibitors. Blood 2014;123:1341–52. et  al. Extracellular signal-regulated kinase mediates chromatin
18. Ferrara FF, Fazi F, Bianchini A, Padula F, Gelmetti V, Minucci S, rewiring and lineage transformation in lung cancer [Internet]. Can-
et  al. Histone deacetylase-targeted treatment restores retinoic acid cer Biology; 2020 Nov. Available from: http://biorxiv.org/lookup/
signaling and differentiation in acute myeloid leukemia. Cancer Res doi/10.1101/2020.11.12.368522.
2001;61:2–7. 41. Dravis C, Chung C-Y, Lytle NK, Herrera-Valdez J, Luna G, Trejo CL,
19. Kaufman CK, Mosimann C, Fan ZP, Yang S, Thomas AJ, Ablain J, et  al. Epigenetic and transcriptomic profiling of mammary gland
et al. A zebrafish melanoma model reveals emergence of neural crest development and tumor models disclose regulators of cell state
identity during melanoma initiation. Science 2016;351:aad2197. plasticity. Cancer Cell 2018;34:466–82.
20. Morris JP, Yashinskie JJ, Koche R, Chandwani R, Tian S, Chen C-C, 42. Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L,
et al. α-Ketoglutarate links p53 to cell fate during tumour suppres- Weinstein JN, et  al. Machine learning identifies stemness features
sion. Nature 2019;573:595–9. associated with oncogenic dedifferentiation. Cell 2018;173:338–54.
21. Saha SK, Parachoniak CA, Ghanta KS, Fitamant J, Ross KN, 43. Miao Z-F, Lewis MA, Cho CJ, Adkins-Threats M, Park D, Brown JW,
Najem MS, et al. Mutant IDH inhibits HNF-4α to block hepatocyte et  al. A dedicated evolutionarily conserved molecular network
differentiation and promote biliary cancer. Nature 2014;513:110–4. licenses differentiated cells to return to the cell cycle. Dev Cell
22. Dang L, Su S-SM. Isocitrate dehydrogenase mutation and (R)-2-hy- 2020;55:178–94.
droxyglutarate: from basic discovery to therapeutics development. 44. De Blander H, Morel A-P, Senaratne AP, Ouzounova M, Puisieux A.
Annu Rev Biochem 2017;86:305–31. Cellular plasticity: a route to senescence exit and tumorigenesis.
23. Waitkus MS, Diplas BH, Yan H. Biological role and therapeutic Cancers 2021;13:4561.
potential of IDH mutations in cancer. Cancer Cell 2018;34:186–95. 45. Merrell AJ, Stanger BZ. Adult cell plasticity in vivo: de-differentia-
24. Phan TG, Croucher PI. The dormant cancer cell life cycle. Nat Rev tion and transdifferentiation are back in style. Nat Rev Mol Cell Biol
Cancer 2020;20:398–411. 2016;17:413–25.
25. Jiang M, Azevedo-Pouly AC, Deering TG, Hoang CQ, DiRenzo D, 46. Baylin SB, Jones PA. Epigenetic determinants of cancer. Cold Spring
Hess DA, et al. MIST1 and PTF1 collaborate in feed-forward regula- Harb Perspect Biol 2016;8:a019505.
tory loops that maintain the pancreatic acinar phenotype in adult 47. Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the
mice. Mol Cell Biol 2016;36:2945–55. hallmarks of cancer. Science 2017;357:eaal2380.
26. Krah NM, Narayanan SM, Yugawa DE, Straley JA, Wright CVE, 48. Jones PA, Issa J-PJ, Baylin S. Targeting the cancer epigenome for
MacDonald RJ, et  al. Prevention and reversion of pancreatic tum- therapy. Nat Rev Genet 2016;17:630–41.
origenesis through a differentiation-based mechanism. Dev Cell 49. Huang S. Tumor progression: Chance and necessity in Darwinian
2019;50:744–54. and Lamarckian somatic (mutationless) evolution. Prog Biophys
27. Krah NM, De La O J-P, Swift GH, Hoang CQ, Willet SG, Chen Pan F, Mol Biol 2012;110:69–86.
et al. The acinar differentiation determinant PTF1A inhibits initia- 50. Darwiche N. Epigenetic mechanisms and the hallmarks of cancer:
tion of pancreatic ductal adenocarcinoma. eLife 2015;4:e07125. an intimate affair. Am J Cancer Res 2020;10:1954–78.
28. Shi G, DiRenzo D, Qu C, Barney D, Miley D, Konieczny SF. Main- 51. Feng Y, Liu X, Pauklin S. 3D chromatin architecture and epigenetic
tenance of acinar cell organization is critical to preventing Kras- regulation in cancer stem cells. Protein Cell 2021;12:440–54.
induced acinar-ductal metaplasia. Oncogene 2013;32:1950–8. 52. Nam AS, Chaligne R, Landau DA. Integrating genetic and non-
29. Kopp JL, von Figura G, Mayes E, Liu F-F, Dubois CL, Morris JP, genetic determinants of cancer evolution by single-cell multi-omics.
et al. Identification of Sox9-dependent acinar-to-ductal reprogram- Nat Rev Genet 2021;22:3–18.
ming as the principal mechanism for initiation of pancreatic ductal 53. Bitman-Lotan E, Orian A. Nuclear organization and regulation of
adenocarcinoma. Cancer Cell 2012;22:737–50. the differentiated state. Cell Mol Life Sci CMLS 2021;78:3141–58.
30. Julian LM, McDonald AC, Stanford WL. Direct reprogramming with 54. Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes
SOX factors: masters of cell fate. Curr Opin Genet Dev 2017;46:24–36. shape. Cell 2007;128:635–8.
31. Grimm D, Bauer J, Wise P, Krüger M, Simonsen U, Wehland M, et al. 55. Zeng Y, Chen T. DNA methylation reprogramming during mam-
The role of SOX family members in solid tumours and metastasis. malian development. Genes 2019;10:257.
Semin Cancer Biol 2020;67:122–53. 56. Hegde AN, Smith SG. Recent developments in transcriptional and
32. Mu P, Zhang Z, Benelli M, Karthaus WR, Hoover E, Chen C-C, et al. translational regulation underlying long-term synaptic plasticity
SOX2 promotes lineage plasticity and antiandrogen resistance in and memory. Learn Mem 2019;26:307–17.
TP53- and RB1-deficient prostate cancer. Science 2017;355:84–8. 57. Kim S, Kaang B-K. Epigenetic regulation and chromatin remodeling
33. Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, in learning and memory. Exp Mol Med 2017;49:e281.
Tibes  R, et  al. Inhibition of the hedgehog pathway in advanced 58. Thienpont B, Van Dyck L, Lambrechts D. Tumors smother their
basal-cell carcinoma. N Engl J Med 2009;361:1164–72. epigenome. Mol Cell Oncol 2016;3:e1240549.
34. Biehs B, Dijkgraaf GJP, Piskol R, Alicke B, Boumahdi S, Peale F, 59. Gameiro PA, Struhl K. Nutrient deprivation elicits a transcriptional
et al. A cell identity switch allows residual BCC to survive Hedgehog and translational inflammatory response coupled to decreased pro-
pathway inhibition. Nature 2018;562:429–33. tein synthesis. Cell Rep 2018;24:1415–24.
35. Boumahdi S, de Sauvage FJ. The great escape: tumour cell plasticity in 60. Lin GL, Monje M. Understanding the deadly silence of posterior
resistance to targeted therapy. Nat Rev Drug Discov 2020;19:39–56. fossa A ependymoma. Mol Cell 2020;78:999–1001.
36. Groves SM, Ireland A, Liu Q, Simmons AJ, Lau K, Iams WT, 61. Michealraj KA, Kumar SA, Kim LJY, Cavalli FMG, Przelicki D,
et al. Cancer Hallmarks Define a Continuum of Plastic Cell States Wojcik JB, et al. Metabolic regulation of the epigenome drives lethal
between Small Cell Lung Cancer Archetypes [Internet]. Systems infantile ependymoma. Cell 2020;181:1329–45.
Biology; 2021 Jan. Available from: http://biorxiv.org/lookup/doi/10. 62. Bakir B, Chiarella AM, Pitarresi JR, Rustgi AK. EMT, MET, plasticity,
1101/2021.01.22.427865. and tumor metastasis. Trends Cell Biol 2020;30:764–76.

44 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions REVIEW

63. Gupta PB, Pastushenko I, Skibinski A, Blanpain C, Kuperwasser C. 87. Thomas S, Izard J, Walsh E, Batich K, Chongsathidkiet P, Clarke G,
Phenotypic plasticity: driver of cancer initiation, progression, and et al. The host microbiome regulates and maintains human health:
therapy resistance. Cell Stem Cell 2019;24:65–78. a primer and perspective for non-microbiologists. Cancer Res
64. Lambert AW, Weinberg RA. Linking EMT programmes to normal 2017;77:1783–812.
and neoplastic epithelial stem cells. Nat Rev Cancer 2021;21:325–38. 88. Dzutsev A, Badger JH, Perez-Chanona E, Roy S, Salcedo R, Smith CK,
65. Lindner P, Paul S, Eckstein M, Hampel C, Muenzner JK, Erlenbach- et al. Microbes and cancer. Annu Rev Immunol 2017;35:199–228.
Wuensch K, et  al. EMT transcription factor ZEB1 alters the epige- 89. Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V, Wargo JA.
netic landscape of colorectal cancer cells. Cell Death Dis 2020;11:147. The microbiome, cancer, and cancer therapy. Nat Med 2019;25:
66. Javaid S, Zhang J, Anderssen E, Black JC, Wittner BS, Tajima K, 377–88.
et  al. Dynamic chromatin modification sustains epithelial-mesen- 90. Sears CL, Garrett WS. Microbes, microbiota, and colon cancer. Cell
chymal transition following inducible expression of Snail-1. Cell Host Microbe 2014;15:317–28.
Rep 2013;5:1679–89. 91. Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A, van
67. Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial- Hoeck A, Wood HM, Nomburg J, et  al. Mutational signature in
mesenchymal transition through epigenetic and post-translational colorectal cancer caused by genotoxic pks+ E. coli. Nature 2020;580:
modifications. Mol Cancer 2016;15:18. 269–73.
68. Skrypek N, Goossens S, De Smedt E, Vandamme N, Berx G. 92. Okumura S, Konishi Y, Narukawa M, Sugiura Y, Yoshimoto S,
Epithelial-to-mesenchymal transition: epigenetic reprogramming Arai  Y, et  al. Gut bacteria identified in colorectal cancer patients
driving cellular plasticity. Trends Genet TIG 2017;33:943–59. promote tumourigenesis via butyrate secretion. Nat Commun 2021;
69. Li L, Hanahan D. Hijacking the neuronal NMDAR signaling circuit 12:5674.
to promote tumor growth and invasion. Cell 2013;153:86–100. 93. Salvi PS, Cowles RA. Butyrate and the intestinal epithelium: modu-
70. Li L, Zeng Q, Bhutkar A, Galván JA, Karamitopoulou E, Noordermeer lation of proliferation and inflammation in homeostasis and dis-
D, et al. GKAP acts as a genetic modulator of NMDAR signaling to ease. Cells 2021;10:1775.
govern invasive tumor growth. Cancer Cell 2018;33:736–51. 94. Fessler J, Matson V, Gajewski TF. Exploring the emerging role of
71. Mohammadi H, Sahai E. Mechanisms and impact of altered tumour the microbiome in cancer immunotherapy. J Immunother Cancer
mechanics. Nat Cell Biol 2018;20:766–74. 2019;7:108.
72. Odenthal J, Takes R, Friedl P. Plasticity of tumor cell invasion: 95. Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA.
governance by growth factors and cytokines. Carcinogenesis 2016; The influence of the gut microbiome on cancer, immunity, and
37:1117–28. cancer immunotherapy. Cancer Cell 2018;33:570–80.
73. Torres CM, Biran A, Burney MJ, Patel H, Henser-Brownhill T, 96. Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. The micro-
Cohen A-HS, et al. The linker histone H1.0 generates epigenetic and biome in cancer immunotherapy: diagnostic tools and therapeutic
functional intratumor heterogeneity. Science 2016;353:aaf1644. strategies. Science 2018;359:1366–70.
74. Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, Gillespie S, et al. 97. Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L,
Single-cell transcriptomic analysis of primary and metastatic tumor et  al. Fecal microbiota transplant promotes response in immuno-
ecosystems in head and neck cancer. Cell 2017;171:1611–24. therapy-refractory melanoma patients. Science 2021;371:602–9.
75. Kinker GS, Greenwald AC, Tal R, Orlova Z, Cuoco MS, McFarland 98. Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J-M,
JM, et  al. Pan-cancer single-cell RNA-seq identifies recurring pro- Morrison RM, et  al. Fecal microbiota transplant overcomes resist-
grams of cellular heterogeneity. Nat Genet 2020;52:1208–18. ance to anti–PD-1 therapy in melanoma patients. Science 2021;
76. Murtha M, Esteller M. Extraordinary cancer epigenomics: think- 371:595–602.
ing outside the classical coding and promoter box. Trends Cancer 99. Griffin ME, Espinosa J, Becker JL, Luo J-D, Carroll TS, Jha JK,
2016;2:572–84. et al. Enterococcus peptidoglycan remodeling promotes checkpoint
77. Nebbioso A, Tambaro FP, Dell’Aversana C, Altucci L. Cancer epige- inhibitor cancer immunotherapy. Science 2021;373:1040–6.
netics: moving forward. PLoS Genet 2018;14:e1007362. 100. Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H,
78. Tavernari D, Battistello E, Dheilly E, Petruzzella AS, Mina M, et  al. Microbiome-derived inosine modulates response to check-
Sordet-Dessimoz J, et al. Non-genetic evolution drives lung adeno- point inhibitor immunotherapy. Science 2020;369:1481–9.
carcinoma spatial heterogeneity and progression. Cancer Discov 101. Ansaldo E, Belkaid Y. How microbiota improve immunotherapy.
2021;11:1490–507. Science 2021;373:966–7.
79. Heyn H, Vidal E, Ferreira HJ, Vizoso M, Sayols S, Gomez A, et  al. 102. Ansaldo E, Farley TK, Belkaid Y. Control of immunity by the micro-
Epigenomic analysis detects aberrant super-enhancer DNA methyla- biota. Annu Rev Immunol 2021;39:449–79.
tion in human cancer. Genome Biol 2016;17:11. 103. Zhang Q, Ma C, Duan Y, Heinrich B, Rosato U, Diggs LP, et al. Gut
80. Saghafinia S, Mina M, Riggi N, Hanahan D, Ciriello G. Pan-cancer microbiome directs hepatocytes to recruit MDSCs and promote
landscape of aberrant DNA methylation across human tumors. Cell cholangiocarcinoma. Cancer Discov 2021;11:1248–67.
Rep 2018;25:1066–80. 104. Ding T, Schloss PD. Dynamics and associations of microbial com-
81. Audia JE, Campbell RM. Histone modifications and cancer. Cold munity types across the human body. Nature 2014;509:357–60.
Spring Harb Perspect Biol 2016;8:a019521. 105. Byrd AL, Liu M, Fujimura KE, Lyalina S, Nagarkar DR, Charbit B,
82. Corces MR, Granja JM, Shams S, Louie BH, Seoane JA, Zhou W, et  al. Gut microbiome stability and dynamics in healthy donors
et al. The chromatin accessibility landscape of primary human can- and patients with non-gastrointestinal cancers. J Exp Med 2021;
cers. Science 2018;362:eaav1898. 218:e20200606.
83. Esteve-Puig R, Bueno-Costa A, Esteller M. Writers, readers and 106. Healy CM, Moran GP. The microbiome and oral cancer: more ques-
erasers of RNA modifications in cancer. Cancer Lett 2020;474: tions than answers. Oral Oncol 2019;89:30–3.
127–37. 107. Swaney MH, Kalan LR. Living in your skin: microbes, molecules and
84. Janin M, Coll-SanMartin L, Esteller M. Disruption of the RNA mechanisms. Infect Immun 2021;89:e00695–20.
modifications that target the ribosome translation machinery in 108. Willis JR, Gabaldón T. The human oral microbiome in health
human cancer. Mol Cancer 2020;19:70. and disease: from sequences to ecosystems. Microorganisms 2020;
85. Hanahan D, Coussens LM. Accessories to the crime: functions 8:308.
of cells recruited to the tumor microenvironment. Cancer Cell 109. Xu J, Peng J-J, Yang W, Fu K, Zhang Y. Vaginal microbiomes and
2012;21:309–22. ovarian cancer: a review. Am J Cancer Res 2020;10:743–56.
86. Lu Z, Zou J, Li S, Topper MJ, Tao Y, Zhang H, et al. Epigenetic ther- 110. Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, et al.
apy inhibits metastases by disrupting premetastatic niches. Nature The human tumor microbiome is composed of tumor type-specific
2020;579:284–90. intracellular bacteria. Science 2020;368:973–80.

JANUARY 2022 CANCER DISCOVERY | 45

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
REVIEW Hanahan

111. Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, et  al. 120. Lee S, Schmitt CA. The dynamic nature of senescence in cancer. Nat
Commensal microbiota promote lung cancer development via γδ T Cell Biol 2019;21:94–101.
cells. Cell 2019;176:998–1013. 121. Wang B, Kohli J, Demaria M. Senescent cells in cancer therapy:
112. Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, friends or foes? Trends Cancer 2020;6:838–57.
Mishra A, et al. The pancreatic cancer microbiome promotes onco- 122. Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, et al.
genesis by induction of innate and adaptive immune suppression. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifes-
Cancer Discov 2018;8:403–16. pan. Nature 2016;530:184–9.
113. McAllister F, Khan MAW, Helmink B, Wargo JA. The tumor micro- 123. Ruhland MK, Loza AJ, Capietto A-H, Luo X, Knolhoff BL,
biome in pancreatic cancer: bacteria and beyond. Cancer Cell Flanagan KC, et al. Stromal senescence establishes an immunosup-
2019;36:577–9. pressive microenvironment that drives tumorigenesis. Nat Com-
114. Kadosh E, Snir-Alkalay I, Venkatachalam A, May S, Lasry A, Elyada mun 2016;7:11762.
E, et  al. The gut microbiome switches mutant p53 from tumour- 124. Hwang HJ, Lee Y-R, Kang D, Lee HC, Seo HR, Ryu J-K, et  al.
suppressive to oncogenic. Nature 2020;586:133–8. Endothelial cells under therapy-induced senescence secrete CXCL11,
115. Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. which increases aggressiveness of breast cancer cells. Cancer Lett
Genes Dev 2020;34:1565–76. 2020;490:100–10.
116. Faget DV, Ren Q, Stewart SA. Unmasking senescence: context- 125. Wang D, Xiao F, Feng Z, Li M, Kong L, Huang L, et  al. Sunitinib
dependent effects of SASP in cancer. Nat Rev Cancer 2019;19: facilitates metastatic breast cancer spreading by inducing endothe-
439–53. lial cell senescence. Breast Cancer Res 2020;22:103.
117. Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, 126. Amor C, Feucht J, Leibold J, Ho Y-J, Zhu C, Alonso-Curbelo D, et al.
Bishop C, et  al. Cellular senescence: defining a path forward. Cell Senolytic CAR T cells reverse senescence-associated pathologies.
2019;179:813–27. Nature 2020;583:127–32.
118. He S, Sharpless NE. Senescence in health and disease. Cell 2017; 127. Aster JC, Pear WS, Blacklow SC. The varied roles of notch in cancer.
169:1000–11. Annu Rev Pathol 2017;12:245–75.
119. Kowald A, Passos JF, Kirkwood TBL. On the evolution of cellular 128. Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR.
senescence. Aging Cell 2020;19:e13270. Vessel co-option in cancer. Nat Rev Clin Oncol 2019;16:469–93.

46 | CANCER DISCOVERY JANUARY 2022 AACRJournals.org

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
Hallmarks of Cancer: New Dimensions
Douglas Hanahan

Cancer Discov 2022;12:31-46.

Updated version Access the most recent version of this article at:
http://cancerdiscovery.aacrjournals.org/content/12/1/31

Cited articles This article cites 126 articles, 28 of which you can access for free at:
http://cancerdiscovery.aacrjournals.org/content/12/1/31.full#ref-list-1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerdiscovery.aacrjournals.org/content/12/1/31.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from cancerdiscovery.aacrjournals.org on January 12, 2022. © 2022 American Association for Cancer
Research.
VOLUME 34 • NUMBER 7 • MARCH 1, 2016

JOURNAL OF CLINICAL ONCOLOGY O R I G I N A L R E P O R T

Impact of Ipsilateral Blood Draws, Injections, Blood Pressure


Measurements, and Air Travel on the Risk of Lymphedema for
Patients Treated for Breast Cancer
Chantal M. Ferguson, Meyha N. Swaroop, Nora Horick, Melissa N. Skolny, Cynthia L. Miller, Lauren S. Jammallo,
Cheryl Brunelle, Jean A. O’Toole, Laura Salama, Michelle C. Specht, and Alphonse G. Taghian
See accompanying article on page 655

All authors: Massachusetts General


Hospital, Harvard Medical School, Boston, A B S T R A C T
MA.
Purpose
Published online ahead of print at
www.jco.org on December 7, 2015.
The goal of this study was to investigate the association between blood draws, injections, blood
pressure readings, trauma, cellulitis in the at-risk arm, and air travel and increases in arm volume in a
Supported by Award No. R01CA139118
(A.G.T.) and Award No. P50CA089393
cohort of patients treated for breast cancer and screened for lymphedema.
(A.G.T.) from the National Cancer Institute, Patients and Methods
and the Adele McKinnon Research Fund Between 2005 and 2014, patients undergoing treatment of breast cancer at our institution were
for Breast Cancer-Related Lymphedema.
screened prospectively for lymphedema. Bilateral arm volume measurements were performed
Terms in blue are defined in the glossary, preoperatively and postoperatively using a Perometer. At each measurement, patients reported the
found at the end of this article and online
at www.jco.org
number of blood draws, injections, blood pressure measurements, trauma to the at-risk arm(s),
and number of flights taken since their last measurement. Arm volume was quantified using the
Presented at the San Antonio Breast
Cancer Symposium, San Antonio, TX,
relative volume change and weight-adjusted change formulas. Linear random effects models were
December 9-12, 2014. used to assess the association between relative arm volume (as a continuous variable) and non-
The content is solely the responsibility of
treatment risk factors, as well as clinical characteristics.
the authors and does not necessarily Results
represent the official views of the National In 3,041 measurements, there was no significant association between relative volume change
Cancer Institute or the National Institutes
of Health.
or weight-adjusted change increase and undergoing one or more blood draws (P = .62), injections
(P = .77), number of flights (one or two [P = .77] and three or more [P = .91] v none), or duration of
Authors’ disclosures of potential conflicts
of interest are found in the article online at
flights (1 to 12 hours [P = .43] and 12 hours or more [P = .54] v none). By multivariate analysis, factors
www.jco.org. Author contributions are significantly associated with increases in arm volume included body mass index $ 25 (P = .0236), axillary
found at the end of this article. lymph node dissection (P , .001), regional lymph node irradiation (P = .0364), and cellulitis (P , .001).
Corresponding author: Alphonse G. Conclusion
Taghian, MD, PhD, Department of This study suggests that although cellulitis increases risk of lymphedema, ipsilateral blood draws, injections,
Radiation Oncology, Massachusetts
General Hospital, 100 Blossom St,
blood pressure readings, and air travel may not be associated with arm volume increases. The results may
Boston, MA 02114; e-mail: ataghian@ help to educate clinicians and patients on posttreatment risk, prevention, and management of lymphedema.
partners.org.

© 2015 by American Society of Clinical


J Clin Oncol 34:691-698. © 2015 by American Society of Clinical Oncology
Oncology

0732-183X/16/3407w-691w/$20.00 the efficacy of such precautionary behaviors do


INTRODUCTION
DOI: 10.1200/JCO.2015.61.5948 not exist, highlighted in a recent statement by the
National Lymphedema Network.5 The guidelines
Clinicians and national guidelines strongly advise place a large amount of burden on patients and
patients with breast cancer to avoid blood draws, clinicians, who go to great lengths to exercise
injections, blood pressure readings, and trauma to precautionary behaviors and face high levels of
the at-risk arm during and after treatment to anxiety when they accidentally do not abide by the
reduce the risk of developing cellulitis and breast guidelines. Therefore, we sought to investigate the
cancer–related lymphedema (BCRL).1-6 Patients association between blood draws, injections,
are also advised to exercise caution when flying by blood pressure readings, trauma, and cellulitis in
wearing prophylactic compression sleeves.1,3,5 the at-risk arm and flying on increases in arm
These guidelines are based on anecdotal infor- volume in a large prospective cohort of patients
mation, and comprehensive data demonstrating undergoing treatment of breast cancer.

© 2015 by American Society of Clinical Oncology 691


Ferguson et al

Lymphedema is a potential adverse effect of breast cancer Patient Population


treatment characterized by arm swelling, discomfort, and impaired All 632 patients included in this study had a new diagnosis of invasive
upper extremity function in its later stages. The development of breast cancer at their baseline measurement, more than 6 months of
lymphedema is the result of a compromised lymphatic drainage postsurgical follow-up, and at least one follow-up arm measurement.
Patient and clinocopathologic characteristics were collected via medical
system after the removal of lymph nodes or radiation to the axillary record review. Patients who undergo sentinel lymph node biopsy are still at
lymph nodes. Patients with breast cancer carry a lifelong risk of risk for developing lymphedema and are often advised to exercise risk
developing lymphedema; the average time to onset is 14.4 months reduction practices just like those who undergo ALND. Therefore, both
after treatment completion.7,8 Consequently, if patients comply groups were included in the study. Episodes of cellulitis were defined as
with cautionary guidelines, they must exercise risk-reducing clinical signs of infection in the ipsilateral arm and/or breast that required
practices for the rest of their lives. antibiotics. Each side was evaluated independently for patients who
underwent bilateral breast surgery, resulting in a total of 760 at-risk arms
Well-defined risk factors for developing lymphedema include
available for analysis. In our hospital, the majority of patients who chose to
axillary lymph node dissection (ALND),9-15 regional lymph node undergo contralateral prophylactic mastectomy undergo sentinel lymph
irradiation (RLNR),8,16-19 higher body mass index (BMI), and node biopsy on their unaffected breast to stage an occult breast cancer if
older age at diagnosis.7,8,11,14,20-30 It remains unclear why, among discovered on final pathology. Therefore, the unaffected breasts for patients
patients with similar demographic and treatment-related charac- undergoing bilateral mastectomies were included in the analysis because
teristics, some go on to develop lymphedema. This variation has they are also at risk for developing lymphedema. Patients were censored
prompted speculation that events including blood draws, injec- from the analysis after a diagnosis of distant metastases and recurrence and
if they wore a compression sleeve while flying.
tions, blood pressure readings, trauma to the arm, air travel, and
cellulitis might incite lymphedema.6,31 Despite the prevalence and
persistence of recommendations to pursue precautionary behavior Statistical Analysis
after breast cancer treatment, few data exist to support these The Kaplan-Meier method was used to estimate the 2-year cumulative
incidence of lymphedema, defined by RVC/WAC $ 10%. Linear random
practices, and investigation into the topic is warranted.5 effects models were used to assess the association between relative arm volume
(as a continuous variable), clinical risk factors, and episodes of non-
precautionary behavior. These models account for the correlation between
arm volume measurements obtained from the same patient and, for patients
PATIENTS AND METHODS who underwent bilateral breast surgery, on the same side of the body. At each
measurement, the number of blood draws, injections, blood pressure
Study Design readings, trauma, and episodes of cellulitis were analyzed both as continuous
Patients with newly diagnosed breast cancer between 2009 and 2014 variables and as dichotomous variables, categorized according to whether
who were prospectively screened for lymphedema at our institution were patients reported having had one or more events versus none. Number of and
included in this analysis. Bilateral arm volume measurements were obtained hours spent on flights in total since last follow-up were analyzed as both
using an optoelectric Perometer (Pero-system, Wuppertal, Germany), which continuous and trichotomous variables. Model estimates give the mean
uses infrared light to measure limb circumference and calculate arm volume. change in RVC or WAC associated with a one-unit increase in a continuous
All patients had a preoperative baseline measurement, a postoperative risk factor and the mean difference in RVC or WAC between subgroups for
measurement, a measurement after chemotherapy and/or radio therapy, and categorical variables. Univariate model results were used to estimate and plot
measurements at regular follow-up intervals corresponding with oncology the mean RVC or WAC within each subgroup for categorical clinical and
visits.32-35 Regular follow-up intervals correspond to time periods of between nontreatment-related variables, along with the 95% CI for the mean and the P
3 and 7 months. Occasionally patients are measured more frequently at their value associated with the comparison of means. The multivariate model was
request. The protocol for lymphedema screening has been previously chosen by starting with a model that included all variables that were sig-
published and approved by the institutional review board33 (ClinicalTrials. nificant at the .10 significance level in the univariate analysis, as well as all
gov identification number NCT01521741). nonprecautionary behaviors, and removing one variable at a time until only
Arm volume changes were quantified using the relative volume change significant (P , .05) variables remained. Because cellulitis is in the causal
(RVC)33 formula after unilateral breast surgery or the weight-adjusted volume pathway between the risk events and arm volume increase, it was not included
change (WAC) formula after bilateral breast surgery.34 The RVC equation in the model selection process to avoid overadjustment bias.
accounts for preoperative differences in arm volume by using a baseline Cox proportional hazards models were used to assess the association
measurement and for nontreatment-related changes, including weight gain or between risk of cellulitis and risk events. Patients who developed cellulitis in the
loss, by comparing the surgical arm to the nonsurgical arm.33,34 Briefly, RVC = postoperative period were excluded from this analysis because of lack of
{[A(2)U(1)/U(2)A(1)} 2 1), where A(1) and A(2) are arm volumes on the information on risk events. In addition, in analyzing the association between risk
surgical or ipsilateral side at preoperative and postoperative measurements, and events and subsequent cellulitis, we evaluated the relationship between the risk
U(1) and U(2) are arm volumes on the contralateral side at corresponding time of infection and the number of risk events reported before the cellulitis episode.
points.33 The WAC calculates changes in arm volume compared with a
preoperative arm measurement for each arm independently and accounts for
weight changes that could influence arm size.34 RVC and WAC have been
RESULTS
shown to classify lymphedema similarly.34 Lymphedema was defined as RVC
or WAC $ 10%.7,15,36
At each Perometer measurement, patients were asked to complete a Patient Population
survey on which they reported the number of blood draws, injections, Six hundred thirty-two patients with a total of 3,041 post-
blood pressure readings, and trauma to the at-risk arm(s); number of operative measurements were included. Median age at diagnosis
flights and the length of the flight since their last measurement; and use of
compression sleeve while flying. Self-reported trauma ranged from
was 52 years (range, 28 to 81 years), and median BMI was 26 lb/in2
bruising to arm fractures. (range, 16 to 59 lb/in2; Table 1); 92.1% of the cohort was white, 0.6%
This analysis includes information from each patient visit at which Hispanic, 2% African American, 2.7% Asian, and 2.6% unknown.
both an arm volume measurement and a survey were obtained. Patients were followed for a median of 24 months (6 to 60 months)

692 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Risk Reduction Practices for Lymphedema

Table 1. Summary of Clinicopathologic Factors Stratified by Lymphedema Table 1. Summary of Clinicopathologic Factors Stratified by Lymphedema
(RVC/WAC $ 10%) (RVC/WAC $ 10%) (continued)
Lymphedema Lymphedema
No (n = 697) Yes (n = 63) Overall (N = 760) No (n = 697) Yes (n = 63) Overall (N = 760)
Age at diagnosis, years* 52 51 52 Chest wall only
BMI, lb/in2* 25.4 29.1 25.6 No. 316 16 332
Follow-up, months* 23.59 34.31 24.03 % 45.3 25.4 43.7
No. of postoperative visits* 4 5 4 RLNR
Months between visits* 7.14 6.45 7.09 No. 130 30 160
Tumor type % 18.7 47.6 21.1
None Not available
No. 112 13 125 No. 3 0 3
% 16.1 20.6 16.4 % 0.4 0 0.4
Invasive Cellulitis
No. 583 50 633 No
% 83.6 79.4 83.3 No. 659 56 715
Not available % 94.5 88.9 94.1
No. 2 0 2 Yes
% 0.3 0 0.3 No. 38 7 45
Laterality % 5.5 11.1 5.9
Unilateral
No. 473 31 504 Abbreviations: ALND, axillary lymph node dissection; BMI, body mass index;
RNLR, regional lymph node irradiation; RVC, relative volume change; SLNB,
% 67.9 49.2 66.3
sentinel lymph node biopsy; WAC, weight-adjusted volume change.
Bilateral *Values shown, median.
No. 224 32 256
% 32.1 50.8 33.7
Breast surgery
Lumpectomy and had a median of four postoperative visits (one to 16). The
No. 359 18 377
% 51.5 28.6 49.6
median time between measurements was 7 months (interquartile
Mastectomy range, 6.2 months). Thirteen percent of patients had one mea-
No. 338 45 383 surement, 34% had two to three measurements, 40% had four to six
% 48.5 71.4 50.4 measurements, and 13% had seven to 16 measurements. By medical
Axillary surgery
None
record review, 5.9% (45 of 760) of treated breasts had at least one
No. 53 7 60 episode of cellulitis any time after surgery requiring antibiotics
% 7.6 11.1 7.9 (Table 1); 47% (21 of 45) of cellulitis episodes occurred in the
SLNB
immediate postoperative period and therefore before the first
No. 519 22 541
% 74.5 34.9 71.2 postoperative arm measurement. At 24 months, the cumulative
ALND incidence of BCRL as defined by an RVC or WAC $ 10% was 7.72%
No. 125 34 159 (95% CI, 5.82% to 10.22%).
% 17.9 54 20.9
Neoadjuvant chemotherapy
No Nonprecautionary Behaviors
No. 606 50 656
% 86.9 79.4 86.3
In 8.5% (251 of 2,965) of responses, patients reported having
Yes one or more blood draw in their affected arm(s) since their last
No. 91 13 104 measurement, 2.1% (63 of 2,961) of responses reported having one
% 13.1 20.6 13.7 or more injection, 16.3% (482 of 2,961) of responses reported
Adjuvant chemotherapy
No
having one or more blood pressure measurements taken, and 1.0%
No. 416 25 441 (37 of 2,999) of responses reported trauma to the at-risk arm
% 59.7 39.7 58 (Table 2). Thirty percent (878 of 2,960) of patients had flown since
Yes their last measurement (Table 2).
No. 281 38 319
% 40.3 60.3 42
Hormonal therapy Univariate and Multivariate Analyses
No
No. 121 12 133 By univariate analysis, there was no significant association
% 17.4 19 17.5 between an increased RVC or WAC and undergoing one or more
Yes blood draws (P = .62), injections (P = .77), trauma to the at-risk
No. 576 51 627
arm (P = .08), number of flights (one or two [P = .77] and three or
% 82.6 81 82.5
Radiation therapy more [P = .91] v none), or duration of flights (1 to 12 hours [P =
None .43] and 12 hours or more [P = .54] v none) (Fig 1). Having a
No. 248 17 265 BMI $ 25 lb/in2 at time of diagnosis (P = .0064), undergoing
% 35.6 27 34.9
ALND (P = .0003), having blood pressure readings (P = .034),
(continued in next column)
RLNR (P , .001), and cellulitis (P , .001) were significantly
associated with arm volume increases (Table 3; Fig 1). When

www.jco.org © 2015 by American Society of Clinical Oncology 693


Ferguson et al

analyzed as continuous variables, age at diagnosis (P = .25), the


Table 2. Summary of Reported Risk Events
number of ipsilateral blood draws (P = .92), injections (P = .85),
No. of Measurements With Event blood pressure readings (P = .15), and number (P = .34) or duration
Lymphedema of flights (P = .98) were not associated with an increase in RVC or
Risk Factor No (n = 192) Yes (n = 2,849) Overall (N = 3,041) WAC by univariate analysis. In a subset of patients who underwent
Blood draws ALND, BMI $ 25 lb/in2 (P = .0051) and cellulitis (P , .001)
None remained significant, but blood draws (P = .26), injections (P = .35),
No. 2,538 176 2,714
% 89.1 91.7 89.2
blood pressures (P = .39), trauma (P = .23), and air travel (one or
1+ two [P = .96] and three or more [P = .88] v no flights) or duration of
No. 241 10 251 flights (1 to 12 hours [P = .85] and 12 hours or more [P = .69] v
% 8.5 5.2 8.3
Not available
none) were not associated with increased arm swelling.
No. 70 6 76 By multivariate analysis, factors significantly associated with
% 2.5 3.1 2.5 increases in arm volume included BMI $ 25 lb/in2 (P = .0236),
Injections
None
ALND (P ,.001), RLNR (P = .0364), and cellulitis (P ,.001) (Fig 2).
No. 2,726 183 2,909 None of the nonprecautionary behaviors were significantly
% 95.7 95.3 95.7 associated with arm volume change in the multivariate analysis.
1+
No. 60 3 63
Further investigation of the association between risk events
% 2.1 1.6 2.1 and subsequent cellulitis revealed that blood draws (hazard ratio
Not available [HR], 0.977; P = .91), injections (HR, 1.101; P = .5), and blood
No. 63 6 69
% 2.2 3.1 2.3
pressure readings (HR, 0.943; P = .1) on the ipsilateral arm were
Blood pressure not significantly associated with cellulitis.
None
No. 2,305 174 2,479
% 80.9 90.6 81.5
1+ DISCUSSION
No. 470 12 482
% 16.5 6.3 15.9
Not available
A large, prospective cohort of patients with breast cancer was
No. 74 6 80 analyzed to investigate the association of nonprecautionary
% 2.6 3.1 2.6 behaviors and the subsequent risk of lymphedema. No significant
Trauma
No association was found between undergoing ipsilateral blood draws,
No. 2,780 182 2,962 injections, and blood pressure readings, and trauma to the at-risk
% 97.6 94.8 97.4 arm, air travel, and arm swelling. Increase in arm swelling was
Yes
No. 29 8 37 associated with ALND, RLNR, BMI $ 25 lb/in2, and cellulitis.
% 1 4.2 1.2 Previous reports postulate that blood draws, injections, blood
Not available pressure readings, and trauma can cause infection and injury to the
No. 40 2 42
% 1.4 1 1.4 at-risk arm.3,6,37-39 The theory is that patients with surgically
Flights, No. removed lymph nodes have compromised lymphatic systems that
None cannot clear fluid resulting from an immune response, leading to
No. 1,949 133 2,082
% 68.4 69.3 68.5 build-up of lymphatic fluid in the arm and subsequent lymph-
1-2 edema.37 One of the first papers to recommend precautionary
No. 453 28 481 behaviors was published in 1992 as a case report of a woman who
% 15.9 14.6 15.8
3+ developed swelling 30 years after treatment in response to a finger
No. 389 8 397 pinprick.40 Smith37 advised avoidance of traumatic events to the at-
% 13.7 4.2 13.1
risk arm after examining 10 patients with BCRL, four of whom
Not available
No. 58 23 81 reported blood draws as triggering their arm swelling. These
% 2 12 2.7 studies are anecdotal and based on information from a small
Flights, hours
number of patients. Despite minimal data, the authors provided
None
No. 1,946 132 2,078 persistent recommendations to avoid blood draws, blood pressure
% 68.3 68.8 68.3 readings, and injections in the at-risk arm to reduce the risk of
1-12
lymphedema.6,31,37,41 The results of our prospective study provide
No. 429 23 452
% 15.1 12 14.9 evidence-based data that call these guidelines into question.
12+ Clark et al41 evaluated 188 patients with breast cancer who
No. 408 13 421
% 14.3 6.8 13.8
underwent ALND and correlated skin punctures with risk of
Not available developing lymphedema. With a median follow-up of 3 years,
No. 66 24 90 20.7% (39 of 188) of patients developed arm lymphedema, 9.6%
% 2.3 12.5 3
(18 of 188) of all patients had a skin puncture, and therefore 44%
(8 of 18) of patients with skin puncture developed lymphedema
during the follow-up period.41 This analysis performed a mea-
surement for lymphedema at 6 months and 3 years after surgery

694 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Risk Reduction Practices for Lymphedema

Risk Factor P 95% CI

No blood draws –0.22 to 0.49


1+ Blood draws .617 –0.58 to 0.59
No injections –0.22 to 0.48
1+ Injections .7697 –1.03 to1.01
No blood pressures –0.15 to 0.57
1+ Blood pressures .0338 –0.72 to 0.25
No trauma –0.24 to 0.47
Fig 1. Univariate analysis of association
Trauma .0811 –0.06 to 2.58 between risk events and arm volume
No cellulitis –0.36 to 0.34 increases. RVC, relative volume change;
WAC, weight-adjusted volume change.
Cellulitis < .001 1.99 to 4.13
No flights –0.28 to 0.45
1–2 Flights .7667 –0.33 to 0.61
3+ Flights .9133 –0.45 to 0.57
No flying hours –0.26 to 0.46
1–12 Flying hours .4261 –0.22 to 0.74
12+ Flying hours .5377 –0.54 to 0.46

–2.00% –1.00% 0.00% 1.00% 2.00% 3.00% 4.00%


Mean Difference in Arm Volume (RVC/WAC) in Subgroup

and did not specify the timing of skin puncture; therefore, it did In contrast, McLaughlin et al6 found no association in risk-
not take into account other events that could have contributed to reducing behaviors of patients with and without lymphedema
lymphedema.42 In our subset analysis of 159 patients undergoing when evaluated using tape measured circumferential arm mea-
ALND, nonprecautionary risk behaviors and specifically skin surements. Circumferential tape measurement is limited because
puncture were not correlated with increased arm swelling. By the measurements can be subject to both inter- and intrapersonal
performing arm measurements at regularly timed intervals for as variation.43 Unlike tape measurements, measurements by the
long as 5 years and recording the number of nonprecautionary Perometer and use of the RVC and WAC formulas account for
events since the previous measurement, we were able to reduce the preexisting arm volume differences and detect small volume
recall bias and more specifically evaluate effects of nonprecau- changes. In addition, obtaining reproducible, prospective arm
tionary behavior on arm swelling. measurements using the Perometer at the same time as collecting
individual incidence of risk-related events could have biased our
data to attribute small volume changes that may have not rep-
Table 3. Univariate Analysis of Categorical Treatment Factors resented swelling detectable via circumferential tape measurement
Patient- or Treatment- Mean RVC/WAC to risk events. Similarly, we found no association with non-
Related Risk Factor in Subgroup (%) 95% CI P precautionary behavior and increase in arm volume.
Surgical characteristics The Physical Activity and Lymphedema (PAL) trial assessed
Lumpectomy 0.04 20.42 to 0.51 —* the safety of progressive strength training in breast cancer sur-
Mastectomy v 0.26 20.28 to 0.79 .5542
lumpectomy vivors and investigated the association of 30 risk factors and arm
No nodal surgery 20.22 21.22 to 0.78 — swelling.44,45 The authors reported no significant association
SLNB v none 20.35 20.73 to 0.04 .8113 between ipsilateral blood draws, blood pressure readings, and air
ALND v none 1.89 1.22 to 2.55 ,.001
travel and incident arm swelling.44 The study included clinical
Radiation therapy
None 20.00 20.43 to 0.43 — evaluation of arm swelling using water displacement and patient-
Breast/chest wall 20.20 20.65 to 0.25 .42341 reported risk-reducing behaviors at 3, 6, and 12 months after
irradiation v none enrollment.44,45 Patients were given compression sleeves to wear
Breast/chest wall + 1.29 0.65 to 1.88 ,.001
RLNR v none during exercise and participated in a biweekly, year-long exercise
BMI, lb/in2 program. The incidence of lymphedema as defined as interlimb
, 25 20.43 20.98 to 0.11 — difference increase of $ 5% was low, which may be due to wearing
$ 25 0.57 0.10 to 1.03 .0064
a compression sleeve or the exercise program.44,45 In addition, the
Abbreviations: ALND, axillary lymph node dissection; BMI, body mass index; incidence of nonprecautionary behaviors was also low, which may be
RLNR, regional lymph node irradiation; RVC, relative volume change; WAC,
weight-adjusted volume change. due to the education about lymphedema provided to participants.
*Indicates the specified variable or comparison was not analyzed. Patients in our cohort do not routinely receive recommendations to
avoid blood draws, injections, or blood pressure readings or to wear a

www.jco.org © 2015 by American Society of Clinical Oncology 695


Ferguson et al

P 95% CI

BMI ≥ 25 .0236 0.06% to 1.38%

ALND < .001 0.94% to 2.55% Fig 2. Multivariate analysis of risk factors
associated with arm volume increases. ALND,
RLNR .0364 0.05% to 1.31% axillary lymph node dissection; BMI, body mass
index (in lb/in2); RNLR, regional lymph node
irradiation; RVC, relative volume change; WAC,
Cellulitis < .001 1.72% to 3.80%
weight-adjusted volume change.

–1.00% 0.00% 1.00% 2.00% 3.00% 4.00%


Mean Difference in Arm Volume (RVC/WAC) for Risk Factor

compressive sleeve while flying. However, our incidence of risk events therefore was not able to account for women who had physical
was low, and we enhanced our patient cohort to include patients with therapy, which might have placed them at lower risk for developing
both unilateral and bilateral breast surgery, increasing both the swelling, even after risk events.
number of risk events and the overall incidence of lymphedema. This study represents the largest cohort of patients with breast
It has been reported that flying incites the development cancer prospectively screened for lymphedema using preoperative
of lymphedema because of changes in cabin air pressure.6,46 Many arm measurements to determine the impact of risk-reducing
clinicians recommend using risk-reducing compression sleeves when practices on arm swelling. We evaluated the association between
flying based on few, mostly retrospective, studies.47 In these studies it risk events and changes in arm volume and found no significant
is unclear if the reported lymphedema was preexisting or influenced by associations between air travel, blood draws, injections, and blood
other factors. Kilbreath et al48 evaluated changes in arm volume after pressure readings in the at-risk arm with arm volume increase.
short- and long-distance flights and found no change in arm volume Patients who have a BMI $ 25 lb/in2, underwent ALND or RLNR,
as measured by bioimpedance. In our analysis, we prospectively col- and have had an episode of cellulitis should be more closely
lected information regarding air travel and quantified arm swelling monitored for changes in arm volume because of the significant
with short intervals of follow-up. This design enabled the investigation association with arm volume increases. The recommendations set
of the immediate effects of flying on arm volume and minimized the forth in the studies discussed still persist today and unfortunately are
risk of recall bias due to the shorter interval of follow up. supported with few evidence-based data. Although we cannot
In our study, BMI $ 25 lb/in2 at diagnosis, ALND, RLNR, and affirmatively state that risk-reduction practices have no effect on arm
cellulitis were significantly associated with arm swelling. High swelling, we hope to generate evidence that brings reasonable doubt
BMI, RLNR, and ALND are consistently cited in the literature as to burdensome guidelines and encourage further investigation into
risk factors for arm swelling and lymphedema, and our data further nonprecautionary behaviors and the risk of lymphedema.
support these claims.6,8,10,11,13-16,19,20,24,49,50 The increased risk for
developing lymphedema after infection including cellulitis is also
well documented in the literature,2,6,22,51 and our results support AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
these findings. Interestingly in our cohort, episodes of cellulitis OF INTEREST
were not associated with a risk event that occurred before the
cellulitis. This may be due to a lower incidence of risk events or to Disclosures provided by the authors are available with this article at
www.jco.org.
an overall lower risk of infection after risk events in a modern era of
attention to sterility.
Limitations of the study include a relatively short median AUTHOR CONTRIBUTIONS
follow-up time of 24 months, low incidence of risk events,
potential for recall bias, and lack of information about patients’ Conception and design: Chantal M. Ferguson, Meyha N. Swaroop, Melissa
receipt of physical therapy. As lymphedema has been shown to N. Skolny, Cynthia L. Miller, Lauren S. Jammallo, Cheryl Brunelle, Jean A.
develop an average of 14.4 months after surgery,8 longer follow-up O’Toole, Michelle C. Specht, Alphonse G. Taghian
time would strengthen the study. Although efforts were made to Collection and assembly of data: Chantal M. Ferguson, Meyha N.
ensure regular follow-up visits, future studies using repeat ques- Swaroop, Melissa N. Skolny, Cynthia L. Miller, Lauren S. Jammallo, Jean A.
tionnaires and sensitivity analyses might elucidate the effects of O’Toole, Laura Salama, Alphonse G. Taghian
Data analysis and interpretation: Chantal M. Ferguson, Meyha N.
recall bias. In addition, our analysis did not adjust for the number Swaroop, Nora Horick, Melissa N. Skolny, Cynthia L. Miller, Lauren S.
of positive lymph nodes because we have not found it to be sig- Jammallo, Cheryl Brunelle, Michelle C. Specht, Alphonse G. Taghian
nificant in a previous analysis; however, this is an important area Manuscript writing: All authors
for future investigation.52 Finally, the study did not capture and Final approval of manuscript: All authors

696 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Risk Reduction Practices for Lymphedema

16. Warren LE, Miller CL, Horick N, et al: The 34. Miller CL, Specht MC, Horick N, et al: A novel,
REFERENCES impact of radiation therapy on the risk of lymph- validated method to quantify breast cancer-related
edema after treatment for breast cancer: A pro- lymphedema (BCRL) following bilateral breast sur-
1. American Cancer Society: Lymphedema: What spective cohort study. Int J Radiat Oncol Biol Phys gery. Lymphology 46:64-74, 2013
every woman with breast cancer should know—For 88:565-571, 2014 35. O’Toole J, Russell T, Singer M, et al: A general
women at risk for lymphedema. http://www.cancer. 17. Coriddi M, Khansa I, Stephens J, et al: Analysis hospital’s multi-disciplinary approach to BCRL.
org/acs/groups/cid/documents/webcontent/002876- of factors contributing to severity of breast cancer- J Lymphoedema 5:81-85, 2010
pdf.pdf related lymphedema. Ann Plast Surg 74:22-25, 2015 36. Armer JM, Heckathorn PW: Post-breast can-
2. Indelicato DJ, Grobmyer SR, Newlin H, et al: 18. Tiwari P, Coriddi M, Lamp S: Lymphedema cer lymphedema in aging women: Self-management
Delayed breast cellulitis: An evolving complication of strategies for investigation and treatment: A review. and implications for nursing. J Gerontol Nurs 31:
breast conservation. Int J Radiat Oncol Biol Phys 66: Plast Surg Nurs 32:173-177, 2012; quiz 178-179 29-39, 2005
1339-1346, 2006 19. Hayes SB, Freedman GM, Li T, et al: Does 37. Smith J: The practice of venepuncture in
3. Loudon L, Petrek J: Lymphedema in women axillary boost increase lymphedema compared with lymphoedema. Eur J Cancer Care (Engl) 7:97-98,
treated for breast cancer. Cancer Pract 8:65-71, supraclavicular radiation alone after breast con- 1998
2000 servation? Int J Radiat Oncol Biol Phys 72:1449-1455, 38. Davis S: Lymphedema following breast cancer
4. National Lymphedema Network: Supplement 2008 treatment. Radiol Technol 70:42-56, 1998; quiz 57-60
to NLN position breast cancer screening: Screening 20. Jammallo LS, Miller CL, Singer M, et al: Impact 39. Drury AN, Jones NW: Observations upon the
and early detection of breast cancer-related lym- of body mass index and weight fluctuation on lym- rate at which edema forms when the veins of the
phedema—The imperative. http://www.lymphnet. phedema risk in patients treated for breast cancer. human limb are congested. Heart 2:580-581, 1927
org/pdfDocs/PP_Lymphedema_BC_Supplement.pdf Breast Cancer Res Treat 142:59-67, 2013 40. Brennan MJ, Weitz J: Lymphedema 30 years
5. National Lymphedema Network: Position 21. Ahmed RL, Schmitz KH, Prizment AE, Folsom after radical mastectomy. Am J Phys Med Rehabil
statement of the National Lymphedema Network: AR: Risk factors for lymphedema in breast cancer 71:12-14, 1992
Lymphedema risk reduction practices. http://www. survivors, the Iowa Women’s Health Study. Breast 41. Clark B, Sitzia J, Harlow W: Incidence and risk
lymphnet.org/pdfDocs/nlnriskreduction.pdf Cancer Res Treat 130:981-991, 2011 of arm oedema following treatment for breast cancer:
6. McLaughlin SA, Bagaria S, Gibson T, et al: 22. McLaughlin SA, Wright MJ, Morris KT, et al: A three-year follow-up study. QJM 98:343-348, 2005
Trends in risk reduction practices for the prevention Prevalence of lymphedema in women with breast 42. Cemal Y, Pusic A, Mehrara BJ: Preventative
of lymphedema in the first 12 months after breast cancer 5 years after sentinel lymph node biopsy or measures for lymphedema: Separating fact from
cancer surgery. J Am Coll Surg 216:380-389, 2013; axillary dissection: Objective measurements. J Clin fiction. J Am Coll Surg 213:543-551, 2011
quiz 511-513 Oncol 26:5213-5219, 2008 43. Deltombe T, Jamart J, Recloux S, et al: Reli-
7. DiSipio T, Rye S, Newman B, et al: Incidence 23. Wilke LG, McCall LM, Posther KE, et al: Sur- ability and limits of agreement of circumferential,
of unilateral arm lymphoedema after breast cancer: A gical complications associated with sentinel lymph water displacement, and optoelectronic volumetry in
systematic review and meta-analysis. Lancet Oncol node biopsy: Results from a prospective international the measurement of upper limb lymphedema.
14:500-515, 2013 cooperative group trial. Ann Surg Oncol 13:491-500, Lymphology 40:26-34, 2007
8. Specht MC, Miller CL, Russell TA, et al: 2006 44. Showalter SL, Brown JC, Cheville AL, et al:
Defining a threshold for intervention in breast cancer- 24. Soran A, Wu WC, Dirican A, et al: Estimating Lifestyle risk factors associated with arm swelling
related lymphedema: What level of arm volume the probability of lymphedema after breast cancer among women with breast cancer. Ann Surg Oncol
increase predicts progression? Breast Cancer Res surgery. Am J Clin Oncol 34:506-510, 2011 20:842-849, 2013
Treat 140:485-494, 2013 25. Swenson KK, Nissen MJ, et al: Case-control 45. Schmitz KH, Troxel AB, Cheville A, et al:
9. Shih YC, Xu Y, Cormier JN, et al: Incidence, study to evaluate predictors of lymphedema after Physical Activity and Lymphedema (the PAL trial):
treatment costs, and complications of lymphedema breast cancer surgery. Oncol Nurs Forum 36: Assessing the safety of progressive strength training
after breast cancer among women of working age: A 185-193, 2009 in breast cancer survivors. Contemp Clin Trials 30:
2-year follow-up study. J Clin Oncol 27:2007-2014, 26. Nesvold IL, Dahl AA, Løkkevik E, et al: Arm and 233-245, 2009
2009 shoulder morbidity in breast cancer patients after 46. Casley-Smith JR, Casley-Smith JR: Lymph-
10. Mansel RE, Fallowfield L, Kissin M, et al: breast-conserving therapy versus mastectomy. Acta edema initiated by aircraft flights. Aviat Space Environ
Randomized multicenter trial of sentinel node biopsy Oncol 47:835-842, 2008 Med 67:52-56, 1996
versus standard axillary treatment in operable breast 27. Hayes SC, Janda M, Cornish B, et al: Lym- 47. Graham PH: Compression prophylaxis may
cancer: The ALMANAC Trial. J Natl Cancer Inst 98: phedema after breast cancer: Incidence, risk factors, increase the potential for flight-associated lym-
599-609, 2006 and effect on upper body function. J Clin Oncol 26: phoedema after breast cancer treatment. Breast 11:
11. Norman SA, Localio AR, Kallan MJ, et al: Risk 3536-3542, 2008 66-71, 2002
factors for lymphedema after breast cancer treat- 28. Paskett ED, Naughton MJ, McCoy TP, et al: 48. Kilbreath SL, Ward LC, Lane K, et al: Effect of
ment. Cancer Epidemiol Biomarkers Prev 19: The epidemiology of arm and hand swelling in pre- air travel on lymphedema risk in women with history
2734-2746, 2010 menopausal breast cancer survivors. Cancer Epi- of breast cancer. Breast Cancer Res Treat 120:
12. Yang EJ, Park WB, Seo KS, et al: Longitudinal demiol Biomarkers Prev 16:775-782, 2007 649-654, 2010
change of treatment-related upper limb dysfunction 29. Crosby MA, Card A, Liu J, et al: Immediate 49. Shahpar H, Atieh A, Maryam A, et al: Risk
and its impact on late dysfunction in breast cancer breast reconstruction and lymphedema incidence. factors of lymph edema in breast cancer patients. Int
survivors: A prospective cohort study. J Surg Oncol Plast Reconstr Surg 129:789e-795e, 2012 J Breast Cancer 2013:641818, 2013
101:84-91, 2010 30. Ozaslan C, Kuru B: Lymphedema after treat- 50. Graham P, Jagavkar R, Browne L, et al:
13. Lucci A, McCall LM, Beitsch PD, et al: ment of breast cancer. Am J Surg 187:69-72, 2004 Supraclavicular radiotherapy must be limited laterally
American College of Surgeons Oncology Group: 31. Cole T: Risks and benefits of needle use in by the coracoid to avoid significant adjuvant breast
Surgical complications associated with sentinel patients after axillary node surgery. Br J Nurs 15: 969- nodal radiotherapy lymphoedema risk. Australas
lymph node dissection (SLND) plus axillary lymph 974, 976-979, 2006 Radiol 50:578-582, 2006
node dissection compared with SLND alone in the 32. Ancukiewicz M, Miller CL, Skolny MN, et al: 51. Mak SS, Yeo W, Lee YM, et al: Predictors of
American College of Surgeons Oncology Group Trial Comparison of relative versus absolute arm size lymphedema in patients with breast cancer under-
Z0011. J Clin Oncol 25:3657-3663, 2007 change as criteria for quantifying breast cancer- going axillary lymph node dissection in Hong Kong.
14. Park JH, Lee WH, Chung HS: Incidence and related lymphedema: The flaws in current studies Nurs Res 57:416-425, 2008
risk factors of breast cancer lymphoedema. J Clin and need for universal methodology. Breast Cancer 52. Miller CL, Skolny M, Ferguson C, et al: Does
Nurs 17:1450-1459, 2008 Res Treat 135:145-152, 2012 number of sentinel lymph nodes removed impact the
15. Tsai RJ, Dennis LK, Lynch CF, et al: The risk of 33. Ancukiewicz M, Russell TA, Otoole J, et al: risk of breast cancer- related lymphedema (BCRL)?
developing arm lymphedema among breast cancer Standardized method for quantification of developing The National Lymphedema Network Conference,
survivors: A meta-analysis of treatment factors. Ann lymphedema in patients treated for breast cancer. Int 11th International Conference, Washington DC , Sep-
Surg Oncol 16:1959-1972, 2009 J Radiat Oncol Biol Phys 79:1436-1443, 2011 tember 3-7, 2014

n n n

www.jco.org © 2015 by American Society of Clinical Oncology 697


Ferguson et al

GLOSSARY TERM

sentinel lymph node: the lymph node that is anatomically manner, this lymph node is said to stand guard or sentinel over the
located such that it is the first site of lymph drainage from the metastatic state of the tumor. For many cancers, the sentinel lymph
location of the primary tumor. It is suspected and assumed that if a node is biopsied as part of the staging process and presence of macro-
malignancy is going to disseminate via the lymphatic system, or micrometastases in the sentinel lymph node is a negative prog-
metastases will first be evident in the sentinel lymph node. In this nostic factor.

698 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Risk Reduction Practices for Lymphedema

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST


Impact of Ipsilateral Blood Draws, Injections, Blood Pressure Measurements, and Air Travel on the Risk of Lymphedema for Patients Treated for
Breast Cancer
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are
self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more
information about ASCO’s conflict of interest policy, please refer to www.asco.org/rwc or jco.ascopubs.org/site/ifc.
Chantal M. Ferguson Cheryl Brunelle
No relationship to disclose No relationship to disclose
Meyha N. Swaroop Jean A. O’Toole
No relationship to disclose No relationship to disclose
Nora Horick Laura Salama
No relationship to disclose No relationship to disclose
Melissa N. Skolny Michelle C. Specht
No relationship to disclose No relationship to disclose
Cynthia L. Miller Alphonse G. Taghian
No relationship to disclose Research Funding: ImpediMed L-Dex
Lauren S. Jammallo
No relationship to disclose

www.jco.org © 2015 by American Society of Clinical Oncology


Research

JAMA | Original Investigation

Effect of Acupuncture vs Sham Acupuncture or Waitlist Control


on Joint Pain Related to Aromatase Inhibitors Among Women
With Early-Stage Breast Cancer
A Randomized Clinical Trial
Dawn L. Hershman, MD, MS; Joseph M. Unger, PhD, MS; Heather Greenlee, ND, PhD; Jillian L. Capodice, MS, LAc;
Danika L. Lew, MA; Amy K. Darke, MS; Alice T. Kengla, MD; Marianne K. Melnik, MD; Carla W. Jorgensen, MD;
William H. Kreisle, MD; Lori M. Minasian, MD; Michael J. Fisch, MD; N. Lynn Henry, MD; Katherine D. Crew, MD, MS

Supplemental content
IMPORTANCE Musculoskeletal symptoms are the most common adverse effects of aromatase Related article at
inhibitors and often result in therapy discontinuation. Small studies suggest that acupuncture jamaoncology.com
may decrease aromatase inhibitor–related joint symptoms.

OBJECTIVE To determine the effect of acupuncture in reducing aromatase inhibitor–related


joint pain.

DESIGN, SETTING, AND PATIENTS Randomized clinical trial conducted at 11 academic centers
and clinical sites in the United States from March 2012 to February 2017 (final date of
follow-up, September 5, 2017). Eligible patients were postmenopausal women with
early-stage breast cancer who were taking an aromatase inhibitor and scored at least 3 on the
Brief Pain Inventory Worst Pain (BPI-WP) item (score range, 0-10; higher scores indicate
greater pain).

INTERVENTIONS Patients were randomized 2:1:1 to the true acupuncture (n = 110), sham
acupuncture (n = 59), or waitlist control (n = 57) group. True acupuncture and sham
acupuncture protocols consisted of 12 acupuncture sessions over 6 weeks (2 sessions per
week), followed by 1 session per week for 6 weeks. The waitlist control group did not receive
any intervention. All participants were offered 10 acupuncture sessions to be used between
weeks 24 and 52.

MAIN OUTCOMES AND MEASURES The primary end point was the 6-week BPI-WP score. Mean
6-week BPI-WP scores were compared by study group using linear regression, adjusted for
baseline pain and stratification factors (clinically meaningful difference specified as 2 points).

RESULTS Among 226 randomized patients (mean [SD] age, 60.7 [8.6] years; 88% white;
mean [SD] baseline BPI-WP score, 6.6 [1.5]), 206 (91.1%) completed the trial. From baseline
to 6 weeks, the mean observed BPI-WP score decreased by 2.05 points (reduced pain)
in the true acupuncture group, by 1.07 points in the sham acupuncture group, and by 0.99
points in the waitlist control group. The adjusted difference for true acupuncture vs sham
acupuncture was 0.92 points (95% CI, 0.20-1.65; P = .01) and for true acupuncture vs waitlist
control was 0.96 points (95% CI, 0.24-1.67; P = .01). Patients in the true acupuncture group
experienced more grade 1 bruising compared with patients in the sham acupuncture group
(47% vs 25%; P = .01).

CONCLUSIONS AND RELEVANCE Among postmenopausal women with early-stage breast


cancer and aromatase inhibitor–related arthralgias, true acupuncture compared with sham
acupuncture or with waitlist control resulted in a statistically significant reduction in joint pain Author Affiliations: Author
affiliations are listed at the end of this
at 6 weeks, although the observed improvement was of uncertain clinical importance.
article.
Corresponding Author: Dawn L.
TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01535066 Hershman, MD, MS, Columbia
University, 161 Ft Washington Ave,
10-1068, New York, NY 10032
JAMA. 2018;320(2):167-176. doi:10.1001/jama.2018.8907 (dlh23@columbia.edu).

(Reprinted) 167
© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Research Original Investigation Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer

A
romatase inhibitors have proven efficacy for the treat-
ment of hormone-sensitive breast cancer.1 However, Key Points
many patients experience adverse effects, including
Question Does acupuncture reduce joint pain related to
aromatase inhibitor–related arthralgias (pain and stiffness), aromatase inhibitors among postmenopausal women with
which occur in approximately 50% of patients2-4 and contrib- early-stage breast cancer?
ute to nonadherence with therapy.5,6 Data from clinical trials
Findings In this multicenter randomized clinical trial of 226
suggest that nonadherence to endocrine therapy is associ-
women with early-stage breast cancer, patients in the acupuncture
ated with reduced disease-free survival.7 group, compared with those in the sham acupuncture group or the
Acupuncture is a traditional Chinese therapy that in- waitlist control group, had significant reductions in changes in joint
volves insertion of fine, single-use, sterile needles in acu- pain scores from baseline to 6 weeks (between-group difference
points according to a system of channels and meridians that vs sham acupuncture, 0.92 points and 0.96 points vs the waitlist
were developed by early practitioners of Traditional Chinese control group [0- to 10-point scale]).
Medicine.8 The needles are stimulated by manual manipula- Meaning Acupuncture was associated with statistically significant
tion, electrical stimulation, or heat.8 Acupuncture is a popu- reductions in aromatase inhibitor–related joint pain at 6 weeks,
lar nonpharmacological modality used for treating a variety of although the magnitude of the improvement was of uncertain
conditions and may result in reduced opioid use.9 Several stud- clinical importance.
ies have evaluated acupuncture as a treatment for aromatase
inhibitor–related arthralgias.10-13 However, these studies pro-
vided uncertain interpretations because of small sample size, Exclusion criteria included prior acupuncture treatment
ineffective blinding, and implementation at single centers. Con- for joint symptoms at any time, although patients were al-
cerns were also raised that the sham control intervention may lowed to have received 2 or fewer acupuncture treatments
have had positive physiological effects and therefore could be within 12 months prior to registration for other reasons. Study
an active control. participants must not have received opioid analgesics, topi-
This multicenter randomized blinded sham- and waitlist- cal analgesics, oral corticosteroids, intramuscular corticoste-
controlled clinical trial was conducted to evaluate the effect roids, or intra-articular steroids, or any other medical therapy,
of acupuncture on joint pain related to aromatase inhibitors alternative therapy, or physical therapy for the treatment of
among women with early-stage breast cancer. joint pain or joint stiffness within 28 days prior to registra-
tion. Patients must not have had a history of bone fracture or
surgery of the affected knees, hands, or both within 6 months
prior to enrollment. Patients with a severe bleeding disorder
Methods or a latex allergy were excluded.
Study Oversight
The study was conducted at 11 academic and community sites Study Intervention
and was approved by appropriate individual institutional re- Study participants were randomized 2:1:1 to the true acupunc-
view boards. Sites were required to have 2 trained acupunc- ture group, the sham acupuncture group, or the waitlist con-
turists for the duration of the trial. Study participants were in- trol group, with randomization dynamically balanced by study
formed of the investigational nature of the study and provided site to account for potential differences in acupuncture
written informed consent. Race and ethnicity, which were self- administration.12 The acupuncture study interventions were
reported by patients and recorded by site investigators, were developed by a consensus of acupuncture experts based on pre-
reported to aid in the determination of the generalizability of vious studies of acupuncture for aromatase inhibitor–related
the findings.14 The trial protocol is reported in Supplement 1. arthralgias with adherence to the Standards for Reporting of
Controlled Trials in Acupuncture (STRICTA) recommendations.13
Patient Characteristics The details of the acupuncture point protocol (ie, body site of
Eligible study participants were women with histologically con- needle placement) and extensive training and standardization
firmed (stages I-III) primary invasive estrogen receptor– methods (in-person training, study manuals, monthly phone
positive carcinoma of the breast, progesterone receptor– calls, and remote quality assurance monitoring) have been pre-
positive carcinoma of the breast, or both. Patients were required viously described.17
to be postmenopausal or premenopausal with use of a Briefly, both true acupuncture and sham acupuncture con-
gonadotropin-releasing hormone agonist, to be taking a third- sisted of twelve 30- to 45- minute sessions administered over
generation aromatase inhibitor for more than 30 days prior to a period of 6 weeks (2 per week) followed by 1 session per week
registration with plans to continue for at least 1 additional year, for 6 weeks. For true acupuncture, stainless steel, single-use,
to have recovered from the effects of surgery or chemo- sterile and disposable needles were used and inserted at tra-
therapy, and to have a Zubrod performance status of 0 to 1.15 ditional depths and angles. The joint-specific protocol was tai-
Inclusion criteria included a score of greater than 3 (range, 0-10; lored to as many as 3 of the patient’s most painful joint areas.18
higher scores indicate greater pain) on the worst pain item of Needles were restimulated manually once during each ses-
the Brief Pain Inventory-Short Form (BPI-SF) that (by patient sion. The sham acupuncture consisted of a core standardized
report) started or increased since starting aromatase inhibi- prescription of minimally invasive, shallow needle insertion
tor therapy.16 using thin and short needles at nonacupuncture points. The

168 JAMA July 10, 2018 Volume 320, Number 2 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer Original Investigation Research

sham acupuncture protocol also included joint-specific treat- compared with both sham acupuncture or waitlist control at
ments and an auricular sham based on the application of ad- 6 weeks. For a 2-point difference between groups and an
hesives to nonacupuncture points on the ear. The waitlist con- assumed 3.0-point standard deviation at 6 weeks, 208 eli-
trol group received no acupuncture treatments and received gible patients were required for 82% power (using 2-sided
no other intervention for 24 weeks after randomization. At 24 tests; α = .025) for true acupuncture vs sham acupuncture
weeks, all patients received vouchers for 10 true acupuncture and true acupuncture vs waitlist control. The design further
bonus sessions to be used prior to the 52-week visit. specified an estimated 5% nonadherence and 10% dropout
rate at the primary end point evaluation time of 6 weeks.
Patient-Reported Outcome Measures In addition, the design incorporated a 10% contamination
Primary End Point rate. The primary outcome was analyzed using the intention-
There is no consensus on the best measure for aromatase to-treat principle (ie, as randomized), using a complete-case
inhibitor–induced arthralgias. The BPI-SF was used in several approach given limited missing data (<10%), without using
prior interventional trials of aromatase inhibitor arthralgias.10,19,20 imputation.
This 14-item questionnaire asks individuals to rate joint pain over The primary outcome at week 6 and the secondary out-
the prior week and the degree to which the pain interferes with comes at weeks 6 and 12 were examined using multivariable
activities using a 0- to 10-point scale (0 [no pain] to 10 [pain as linear regression and adjusting for the baseline score, indica-
bad as you can imagine]).16 For this study, the primary end point tor variables for study sites, and 2 indicator variables for in-
was the BPI Worst Pain Item (BPI-WP) score at 6 weeks of treat- tervention group. The baseline score was not included as a com-
ment. A reduction of 2 points on the BPI-WP has been identified ponent of the outcome because it was obtained prior to
as a clinically meaningful change for a patient.21 intervention administration.
Longitudinal measures through week 24 (12 weeks fol-
Prespecified Secondary End Points lowing completion of the intervention) were analyzed as sec-
Prespecified secondary end points were measured and evalu- ondary outcomes using linear mixed models with a random
ated using several instruments. The BPI-SF was used to mea- effect for patient and an a priori unstructured covariance
sure worst pain, worst stiffness, pain severity, and pain- matrix.26 Fixed-effect factors associated with the outcomes
related interference scores (range from no symptoms to of interest included intervention assignment using separate
worst, 0-10); administered at 6, 12, 16, 20, 24, and 52 weeks indicator variables (with true acupuncture as the reference
to assess the duration of effect following completion of the category), continuous time (both as a linear variable and qua-
intervention. dratic variable), an intervention × time interaction (both lin-
The Western Ontario and McMaster Universities Osteoar- ear and quadratic time), the baseline score, and indicator
thritis Index (WOMAC) version 3.1, a validated measure for variables for study sites.
assessing osteoarthritis of the knees or hips, evaluated In a post hoc analysis, we reported the proportion of pa-
patients using 24 questions related to 3 subscales (pain tients reporting an individual-level clinically meaningful re-
[0-500], stiffness [0-200], and physical function [0-1700])22; duction (2 points from baseline on a scale of 0-10 or [calcu-
administered at 6, 12, 24, and 52 weeks. The Modified Score lated separately] a relative reduction of 30% ), based on current
for the Assessment and Quantification of Chronic Rheuma- guidelines for clinical trials evaluating interventions to re-
toid Affections of the Hands (M-SACRAH) was used to assess duce pain.27,28 Risk differences (RDs) and relative risks (RRs)
hand pain, stiffness, and functional status (range, 0-100)23; were estimated using Poisson regression with robust stan-
administered at 6, 12, 24, and 52 weeks. For both instru- dard errors.29
ments, higher scores indicate greater symptom severity. Statistical analyses were conducted using SAS version 9.44
The Functional Assessment of Cancer Therapy-Endocrine (SAS Institute), including SAS proc mixed for linear mixed
Symptoms (FACT-ES) measured physical and functional well- models.30 For the primary end point, Bonferroni adjustment
being and endocrine symptoms (score range, 0-128); admin- was used for multiple comparisons (setting α = .025) for each
istered at 6, 12, 24, and 52 weeks. The PROMIS Pain Impact- independent comparison of true acupuncture vs sham acu-
Short Form (PROMIS PI-SF; score range, 6-30) 24,25 was puncture and true acupuncture vs waitlist control. No adjust-
administered at 6, 12, 24, and 52 weeks. Both insturments have ments for multiple comparisons were made for all secondary
5 response levels (low scores [not at all] to high scores [very or post hoc analyses, which are considered exploratory.
much]), and higher scores reflect better symptoms. Addi-
tional prespecified secondary end points were analgesic and
opioid use and safety and tolerability of acupuncture. Other
outcomes included an assessment of masking among pa-
Results
tients enrolled in the true acupuncture and sham acupunc- Patient Characteristics
ture study groups. From March 2012 to February 2017, a total of 226 patients were
randomly assigned to the true acupuncture (n = 110), sham acu-
Statistical Analyses puncture (n = 59), or waitlist control (n = 57) group (Figure 1).
The primary hypothesis was that true acupuncture would Patient baseline characteristics are shown in Table 1. Median
decrease joint pain associated with the use of aromatase age was lower in the sham acupuncture group (57.0 years) than
inhibitors (according to the BPI-SF worst pain item [BPI-WP]) in the waitlist control (60.6 years) or true acupuncture (60.8

jama.com (Reprinted) JAMA July 10, 2018 Volume 320, Number 2 169

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Research Original Investigation Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer

Figure 1. Flow of Randomized Patients for the Effect of True Acupuncture vs Sham Acupuncture and True Acupuncture vs Waitlist Control on Joint
Pain Using the 6-Week Brief Pain Inventory-Short Form Worst Pain Score

226 Patients randomizeda

110 Randomized to receive true acupuncture 59 Randomized to receive sham acupuncture 57 Randomized to the waitlist control group
108 Received true acupuncture 57 Received sham acupuncture 56 Were in the waitlist control group
as randomized as randomized as randomized
2 Did not receive true acupuncture 2 Did not receive sham acupuncture 1 Was not in the waitlist control group
as randomized as randomized as randomized

10 Not evaluable 5 Not evaluable (did not have a 6 Not evaluable


9 Did not have a 6-wk BPI measure baseline BPI measure) 5 Did not have a 6-wk BPI measure
1 Did not have a baseline BPI measure 1 Did not have a baseline BPI measure

100 Included in the 6-wk primary analysis 54 Included in the 6-wk primary analysis 51 Included in the 6-wk primary analysis
2 Received no treatmentb 2 Received no treatmentb 1 Did not wait for treatmentb
1 Received true acupuncture for 2 sessionsb
101 Included in the 12-wk analysisc 51 Included in the 12-wk analysis
54 Included in the 12-wk analysis

b
BPI indicates Brief Pain Inventory. Patients’ status as major deviations began immediately in the course of
a
Data were not collected to report the number of patients screened for follow-up and was applied to both the 6-week and 12-week analyses.
c
eligibility prior to randomization. One patient who was not available for 6-week evaluation was evaluated
at 12 weeks.

years) groups. Fewer Hispanic patients were randomized to the true acupuncture vs sham acupuncture of 0.92 points (95% CI,
waitlist control group, and more Asian patients were random- 0.20-1.65; P = .01) and between true acupuncture vs waitlist
ized to the true acupuncture group. The overall median time control of 0.96 points (95% CI, 0.24-1.67; P = .01) (Table 2).
for receiving aromatase inhibitor therapy prior to enrollment Thus, the study results rejected the null hypothesis that true
was 1.1 years. The patient characteristics were well balanced acupuncture generated the same outcomes as sham acupunc-
by study site and clinical factors. ture and waitlist control, although the magnitude of the ef-
Major protocol deviations were reported for 6 patients: 1 fect did not achieve the prespecified difference of 2 points.
patient randomized to the sham acupuncture group received
true acupuncture for 2 sessions, 4 patients randomized to Secondary Outcomes
acupuncture groups (true, 2; sham, 2) received no treatment, Patients randomized to the true acupuncture group had sta-
and 1 patient randomized to the waitlist control group was in tistically significant improved symptom scores compared with
too much pain and declined to participate (but did complete those randomized to the sham acupuncture and waitlist con-
the patient-reported questionnaires). Twenty-one patients trol groups at 6 weeks according to BPI average pain, pain se-
were not evaluable (Figure 1). No patients reported receiving verity, and worst stiffness (Table 2). Patients randomized to
acupuncture outside of the trial during the 12-week interven- true acupuncture had improved symptoms at 6 weeks com-
tion period. pared with those in the sham acupuncture group, but not
compared with the waitlist control group according to pain in-
Worst Pain Scores terference. At 12 weeks, patients randomized to the true acu-
In total, 91% (100/110) of patients in the true acupuncture puncture group compared with the sham acupuncture group
group, 92% (54/59) in the sham acupuncture group, and 89% had statistically significant improvements in average pain, but
(51/57) in the waitlist control group had both baseline and no significant improvement in worst pain, pain interference,
6-week BPI-WP scores for analysis (Figure 1). Reporting rates pain severity, or worst stiffness (Table 2). Compared with the
at weeks 6 and 12 were similar. There was no statistically sig- waitlist control group, patients randomized to the true acu-
nificant difference by study group in the amount of available puncture group had improved pain by all BPI measures at
data at follow-up (P = .93). Mean baseline BPI-WP score was week 12 (P≤.003).
6.84 in the true acupuncture group, 6.55 in the sham acupunc- Patients randomized to the true acupuncture group had
ture group, and 6.48 in the waitlist control group. improved symptoms compared with those in the sham acu-
puncture group at 6 weeks according to the M-SACRAH,
Primary Outcome WOMAC, and PROMIS PI-SF measures (eTable 3 in Supple-
Compared with baseline, the mean observed BPI-WP score was ment 2). Patients randomized to the true acupuncture
2.05 points lower (reduced pain) at 6 weeks in the true acu- group had improved symptoms compared with those in the
puncture group, 1.07 points lower in the sham acupuncture waitlist control group at 6 weeks according to the M-SACRAH
group, and 0.99 points lower for the waitlist control group, with and WOMAC measures. At week 12, patients randomized
differences in adjusted 6-week mean BPI-WP scores between to the true acupuncture group had improved symptoms

170 JAMA July 10, 2018 Volume 320, Number 2 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer Original Investigation Research

Table 1. Baseline Characteristics for Patients in the True Acupuncture, Sham Acupuncture,
or Waitlist Control Group

No. (%)a
True Acupuncture Sham Acupuncture Waitlist Control
(n = 110) (n = 59) (n = 57)
Age, median (range), yb 60.8 (34.1-80.6) 57.0 (40.6-77.5) 60.6 (27.1-76.0)
Hispanic
Yes 11 (10) 7 (12) 3 (5)
Unknown 0 1 0
Race
White 88 (83) 54 (93) 51 (91)
Black 6 (6) 2 (3) 2 (4)
Asian 11 (10) 2 (3) 2 (4)
Pacific Islander 0 0 1 (2)
American Indian 1 (1) 0 0
Unknown 4 1 1
Study sitec
Columbia University 14 (13) 8 (14) 7 (12)
Minority Underserved NCORP
Kaiser Permanente Medical Center 27 (25) 14 (24) 15 (26)
Spectrum Health Medical Group 34 (31) 17 (29) 17 (30)
Other (8) 35 (32) 20 (34) 18 (32)
Breast cancer stage
I 41 (39) 28 (47) 28 (49)
II 53 (50) 23 (39) 23 (40)
III 12 (11) 8 (14) 6 (11)
Prior chemotherapy 56 (51) 31 (53) 24 (42)
Prior tamoxifen 18 (16) 15 (25) 10 (18)
Current or prior aromatase inhibitor therapyd
Anastrozole 80 (73) 44 (75) 40 (70)
Currently using 64 (58) 37 (63) 37 (65)
Not currently using 15 (14) 7 (12) 3 (5)
Not reported 1 (1) 0 0
Letrozole 36 (33) 17 (29) 17 (30)
Currently using 29 (26) 13 (22) 11 (19)
Not currently using 7 (6) 4 (7) 6 (11) Abbreviations: NCORP, National
Cancer Institute Community
Exemestane 21 (19) 10 (17) 10 (18)
Oncology Research Program;
Currently using 15 (14) 9 (15) 8 (14) NSAID, nonsteroidal
Not currently using 6 (5) 1 (2) 2 (4) anti-inflammatory drug.
a
Time receiving aromatase inhibitors, 1.0 (0.1-8.0) 1.1 (0.1-9.0) 1.1 (0.1-3.1) Values are reported as No. (%)
median (range), y unless otherwise indicated.
b
Prior acupuncture 19 (17) 13 (22) 12 (21) Younger women were either
Pain medications (any use) d surgically or chemically
menopausal.
Acetaminophen/paracetamol 26 (24) 12 (20) 8 (14) c
See eTable 5 in Supplement 2 for
Ibuprofen 31 (28) 20 (34) 22 (39) specific site names and locations.
Other NSAID 15 (14) 8 (14) 8 (14) d
Indicates the number of patients
Other pain medication 6 (5) 3 (5) 4 (7) who answered yes. More than 1
subcategory type is allowed.
≥1 medication 66 (60) 36 (61) 33 (58)
Therefore, items may not sum.

compared with those in the sham acupuncutre group ac- over time (eTable 4 in Supplement 2). Through 24 weeks,
cording to the WOMAC and PROMIS PI-SF measures, and adjusted mean BPI-WP scores were 0.59 points lower (95%
they had improved symptoms compared with those in the CI, 0.34-1.14; P = .04) in the true acupuncture group com-
waitlist control group according to the M-SACRAH, WOMAC, pared with the sham acupuncture group and were 1.23 points
FACT-ES, and PROMIS PI-SF measures. lower (95% CI, 0.66-1.80; P<.001) in the true acupuncture
In linear mixed-model analysis, there was no significant group compared with the waitlist control group (Figure 2).
interaction between intervention assignment and time, and Similar findings for trends are shown for the secondary mea-
there was no significant quadratic relationship of BPI-WP sures (eFigure 1 in Supplement 2).

jama.com (Reprinted) JAMA July 10, 2018 Volume 320, Number 2 171

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
172
Table 2. Observed and Fitted Group Mean Results for Brief Pain Inventory-Short Form Scores at Weeks 6 and 12 in Each Group

Group Mean Differencea Proportion With >30% Improvement


Mean (95% CI) Fitted Difference (95% CI) Risk Difference, % (95% CI)
Analysis No. of True vs Sham True vs Sham
c
by Group Patients Baseline Follow-up True vs Waitlist P Value % True vs Waitlist P Value Relative Risk (95% CI)b P Value
Worst Paind
Week 6e
True 100 6.84 (6.55 to 7.13) 4.79 (4.36 to 5.22) 49.0 1 [Reference]
Research Original Investigation

Sham 54 6.55 (6.14 to 6.98) 5.48 (4.78 to 6.18) 0.92 (0.20 to 1.65) .01 24.1 24.9 (9.9 to 40.0) .001 1.95 (1.16 to 3.28) .01
Waitlist 51 6.48 (6.07 to 6.89) 5.49 (4.81 to 6.17) 0.96 (0.24 to 1.67) .01 23.5 25.5 (10.3 to 40.7) .001 2.04 (1.19 to 3.48) .009
Week 12
True 101 6.84 (6.55 to 7.13) 4.53 (4.05 to 5.01) 51.5 1 [Reference]
Sham 54 6.55 (6.14 to 6.98) 5.04 (4.33 to 5.74) 0.73 (−0.07 to 1.53) .08 46.3 5.2 (−11.3 to 21.7) .54 1.09 (0.77 to 1.54) .62
Waitlist 51 6.48 (6.07 to 6.89) 6.29 (5.73 to 6.86) 1.95 (1.19 to 2.70) <.001 15.7 35.8 (21.9 to 49.8) <.001 3.24 (1.66 to 6.34) <.001
Average Pain

JAMA July 10, 2018 Volume 320, Number 2 (Reprinted)


Week 6
True 100 4.95 (4.64 to 5.27) 3.50 (3.21 to 3.88) 43.0 1 [Reference]
Sham 54 4.69 (4.22 to 5.17) 3.93 (3.40 to 4.45) 0.60 (0.03 to 1.17) .04 25.9 17.1 (1.9 to 32.3) .03 1.60 (0.97 to 2.63) .07
Waitlist 51 4.95 (4.52 to 5.37) 4.14 (3.65 to 4.63) 0.71 (0.15 to 1.28) .01 17.7 25.4 (11.1 to 39.6) .0005 2.44 (1.28 to 4.62) .006
Week 12
True 101 4.95 (4.64 to 5.27) 3.00 (2.62 to 3.38) 60.4 1 [Reference]
Sham 53 4.69 (4.22 to 5.17) 3.62 (3.06 to 4.18) 0.79 (0.16 to 1.42) .02 45.3 15.1 (−1.3 to 31.6) .07 1.31 (0.94 to 1.83) .11

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Waitlist 51 4.95 (4.52 to 5.37) 4.33 (3.80 to 4.87) 1.38 (0.76 to 2.00) <.001 29.4 31.0 (15.3 to 46.7) .0001 2.04 (1.30 to 3.21) .002
Pain Interference
Week 6
True 100 4.24 (3.86 to 4.62) 2.55 (2.11 to 2.98) 59.0 1 [Reference]
Sham 54 3.85 (3.34 to 4.37) 3.03 (2.44 to 3.62) 0.73 (0.11 to 1.35) .02 44.4 14.6 (−1.8 to 30.9) .08 1.36 (0.98 to 1.89) .06
Waitlist 51 4.01 (3.54 to 4.48) 3.07 (2.47 to 3.67) 0.63 (−0.03 to 1.29) .06 35.3 23.7 (7.4 to 40.0) .004 1.68 (1.12 to 2.52) .01
Week 12
True 101 4.24 (3.86 to 4.62) 2.44 (2.00 to 2.88) 68.3 1 [Reference]
Sham 54 3.85 (3.34 to 4.37) 2.40 (1.84 to 2.96) 0.19 (−0.40 to 0.77) .53 64.8 3.5 (−12.1 to 19.1) .66 1.06 (0.84 to 1.33) .64

© 2018 American Medical Association. All rights reserved.


Waitlist 51 4.01 (3.54 to 4.48) 3.31 (2.69 to 3.94) 0.99 (0.36 to 1.63) .003 37.3 31.1 (15.0 to 47.1) <.001 1.85 (1.26 to 2.70) .002
Pain Severity
Week 6
True 100 4.71 (4.41 to 5.01) 3.21 (2.85 to 3.57) 50.0 1 [Reference]
Sham 54 4.75 (5.32 to 5.19) 3.75 (3.21 to 5.28) 0.56 (0.00 to 1.11) .05 33.3 16.7 (0.7 to 32.6) .04 1.52 (1.00 to 2.31) .05
Waitlist 51 4.64 (4.25 to 5.03) 3.82 (3.26 to 4.37) 0.71 (0.15 to 1.28) .01 33.3 16.7 (0.4 to 32.9) .04 1.51 (0.98 to 2.34) .06
Week 12
True 101 4.71 (4.41 to 5.01) 2.89 (2.52 to 3.26) 57.4 1 [Reference]
Sham 54 4.75 (5.32 to 5.19) 3.41 (2.86 to 3.95) 0.53 (−0.06 to 1.11) .08 44.4 13.0 (−3.4 to 29.4) .12 1.29 (0.92 to 1.81) .15
Waitlist 51 4.64 (4.25 to 5.03) 4.25 (3.72 to 4.78) 1.43 (0.84 to 2.02) <.001 21.6 35.9 (21.0 to 50.7) <.001 2.64 (1.53 to 4.55) <.001

(continued)

jama.com
Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer
Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer Original Investigation Research

Patients randomized to the true acupuncture group were

weeks (70% vs 43%, P = .009). However, there was no evidence that the intervention effect with respect to the
acupuncture group to believe they were receiving true acupuncture at 6 weeks (68% vs 36%, P<.001) and at 12
P Value
more likely to believe they were receiving true acupuncture

.002

primary outcome of worst pain differed between those believing vs not believing they were receiving true
.04
.02

.62
than those receiving sham acupuncture (68% vs 36%; P<.001).
However, the intervention effect for BPI-WP did not differ be-
tween those believing vs not believing they were receiving true
acupuncture when interaction tests were used (P for interac-

acupuncture at either 6 weeks (P for interaction = .16) or 12 weeks (P for interaction = .75).
Relative Risk (95% CI)b
tion = .16).

1.56 (1.03 to 2.37)


1.76 (1.11 to 2.78)

1.09 (0.78 to 1.53)


2.54 (1.42 to 4.54)
1 [Reference] Post Hoc Analysis

1 [Reference]
The proportion of patients experiencing at least a 2-point re-
duction in BPI-WP at 6 weeks was 58.0% (n = 58) for the true
acupuncture group, 33.3% (n = 18) for the sham acupuncture
group (RD, 24.7% [95% CI, 8.8%-40.5%]; RR, 1.64 [95% CI, 1.10-
2.44]; P = .02), and 31.4% (n = 16) for the waitlist control group
P Value

(difference for true acupuncture vs waitlist control, 26.6% [95%


.005

<.001
.02

.66

CI, [10.6%-42.6%]; RR, 1.75 [95% CI, 1.13-2.69]; P = .01; eFig-


Risk Difference, % (95% CI)

ure 2 and eTable 2 in Supplement 2) Similar results by treat-


Proportion With >30% Improvement

ment group were evident for the proportion of patients expe-


3.7 (−12.8 to 20.2)
30.4 (15.7 to 45.1)

Category indicates the primary end point.


18.7 (2.7 to 34.6)
22.6 (6.7 to 38.5)

riencing an improvement of greater than 30% (Figure 2; eTable


True vs Waitlist

2 and eTable 3 in Supplement 2).


True vs Sham
Table 2. Observed and Fitted Group Mean Results for Brief Pain Inventory-Short Form Scores at Weeks 6 and 12 in Each Group (continued)

Adverse Events
Bruising was the most common adverse event reported for
those receiving true acupuncture or sham acupuncture. More
52.0
33.3
29.4

50.0
46.3
19.6

patients in the true acupuncture group experienced grade 1


%

bruising (47%) than in the sham acupuncture group (25%;


P = .01) (eTable 1 in Supplement 2). There was 1 episode of grade
P Value

<.001

2 presyncope in the true acupuncture group and 1 episode in


.02
.01

.08

the sham acupuncture group. No other differences by study


e

group for selected adverse events were observed, and no grade


Fitted Difference (95% CI)

regression (adjusting for the baseline score and the stratification factor). Positive values favor true acupuncture.
Data for examination of relative risks were calculated using Poisson regression (adjusting for the baseline score

3 or higher adverse events were reported.


0.72 (−0.08 to 1.53)

Patients randomized to the true acupuncture group were more likely than patients randomized to the sham
1.00 (0.19 to 1.81)
1.09 (0.26 to 1.92)

1.80 (1.03 to 2.57)


Data for examination of group mean differences by study group were calculated using multivariable linear
True vs Waitlist
True vs Sham

Discussion
In this multicenter, sham- and waitlist-controlled clinical trial
of patients with early-stage breast cancer and aromatase
inhibitor–related joint pain, there were statistically signifi-
4.46 (3.94 to 4.98)
5.50 (4.86 to 6.14)
5.53 (4.91 to 6.14)

4.35 (3.82 to 4.88)


5.07 (4.47 to 5.68)
6.12 (5.59 to 6.64)

cant but modest improvements in pain scores with true acu-


puncture administered twice a week for 6 weeks compared
with both sham acupuncture and waitlist control. However,
Follow-up

and the stratification factor). Positive values favor true acupuncture.

the magnitude of the effect did not achieve the prespecified


between-group difference of 2 points in the primary end point
Data were calculated among patients with follow-up scores.

of BPI-WP, which had been considered to represent a clini-


Group Mean Differencea

cally meaningful difference by the design.


6.65 (6.26 to 7.04)
6.59 (6.03 to 7.16)
6.68 (6.26 to 7.10)

6.65 (6.26 to 7.04)


6.59 (6.03 to 7.16)
6.68 (6.26 to 7.10)

The adjusted mean between-group difference at 6 weeks


was 0.92 points for true acupuncture vs sham acupuncture
Mean (95% CI)

and 0.96 points for true acupuncture vs waitlist control.


Baselinec

Although there is some uncertainty about the clinical mean-


ing of this between-group difference,27,28 the observed differ-
ence in this study is consistent with other randomized stud-
ies of pain control interventions, which have reported
Patients
No. of

between-group mean differences ranging from 0.7 to


100
54
51

100
54
51

1.0 points.31,32 The pain-reporting guidelines describe the


Worst Stiffness

clinical meaning of an individual’s response, with studies


Waitlist

Waitlist

suggesting that a reduction of 2 points on an 11-point scale (or


by Group

Week 12
Analysis

Sham

Sham
Week 6
True

True

30%) represents a clinically important reduction (ie, much


improved).21,28 A post hoc hypothesis-generating, responder
b

d
a

jama.com (Reprinted) JAMA July 10, 2018 Volume 320, Number 2 173

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Research Original Investigation Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer

Figure 2. Linear Mixed-Model Results Through 24 Weeks for Selected Brief Pain Inventory-Short Form End Points

A Worst pain B Pain interference

8 8

6 Waitlist control 6
Mean BPI-SF Score

Mean BPI-SF Score


Sham acupuncture
True acupuncture
4 4
Waitlist control
Sham acupuncture
True acupuncture
2 2

0 0
Baseline 6 12 16 20 24 Baseline 6 12 16 20 24
Assessment Week Assessment Week
No. of patients No. of patients
Waitlist control 56 51 51 48 45 50 Waitlist control 56 51 51 48 45 50
Sham acupuncture 59 54 54 53 52 54 Sham acupuncture 59 54 54 53 52 54
True acupuncture 109 100 101 94 97 97 True acupuncture 109 100 101 94 97 97

C Pain severity D Worst stiffness

8 8

6 6 Waitlist control
Mean BPI-SF Score

Mean BPI-SF Score


Sham acupuncture
True acupuncture
4 Waitlist control 4
Sham acupuncture
True acupuncture

2 2

0 0
Baseline 6 12 16 20 24 Baseline 6 12 16 20 24
Assessment Week Assessment Week
No. of patients No. of patients
Waitlist control 56 51 51 48 45 50 Waitlist control 56 51 51 48 45 50
Sham acupuncture 59 54 54 53 52 54 Sham acupuncture 59 54 54 53 52 54
True acupuncture 109 100 101 94 97 97 True acupuncture 109 100 101 94 97 97

Horizontal bars at baseline indicate the observed baseline means. Vertical bars vs sham acupuncture and true acupuncture vs waitlist control for week 6, 12, 16,
indicate the interquartile range (IQR) for each study group at each assessment 20, and 24 assessment times.
time and are offset by a small margin to show the IQR for each group. Fitted A, P = .04 (vs sham acupuncture); P<.001 (vs waitlist control).
regression lines for each group are also shown, with P values for the effect of
B, P = .18 (vs sham acupuncture); P = .003 (vs waitlist control).
intervention on Brief Pain Inventory-Short Form outcomes derived from
multivariable linear mixed-model analyses with a random effect for patient, C, P = .08 (vs sham acupuncture); P = .003 (vs waitlist control).
adjusting for continuous time, the baseline score, and indicator variables for D, P = .02 (vs sham acupuncture); P<.001 (vs waitlist control).
study sites, and report values for comparisons between true acupuncture

analysis suggested that the proportion of patients with a Both pharmacological and nonpharmacological inter-
2-point improvement at 6 weeks in the true acupuncture ventions have been studied to treat aromatase inhibitor–
group (58%) was greater than that in the sham acupuncture associated arthralgias.19,20,33,34 Although some interventions
group (33%) and in the waitlist control group (31%). have shown a benefit from the intervention, most have been
In this study, the addition of maintenance true acu- difficult to interpret due to methodological limitations.
puncture once a week for an additional 6 weeks, compared Placebo-controlled trials of pharmacological interventions,
with sham acupuncture, was associated with statistically such as vitamin D, omega-3-fatty acids, and duloxetine, have
significant improvements in average pain, but no significant consistently shown placebo effects of as much as 50%;
improvement in worst pain, pain interference, pain severity, however, only duloxetine was found to show a statistically
or worst stiffness at 12 weeks. Compared with waitlist con- significant between-group difference. 1 9,3 3 ,3 4 Trials of
trol, additional maintenance true acupuncture was associ- unblinded, modality-based therapies, such as exercise, have
ated with significant improvements in worst pain, average shown benefit; however, no placebo effect was observed in
pain, pain interference, pain severity and worst stiffness at the control group,20 raising questions about response bias.35
12 weeks. The lack of a placebo effect in prior acupuncture trials raised

174 JAMA July 10, 2018 Volume 320, Number 2 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer Original Investigation Research

concerns about the integrity of the sham blinding, uninten- Limitations


tional crossover in the sham treatment groups, or a possibil- This study also has several limitations. First, blinding was not
ity of a physiological effect of the sham. The current trial possible for patients randomized to the waitlist control group.
was designed with a waitlist control group to address prior Second, a significantly higher proportion of patients in the true
concerns about the sham effects. acupuncture group compared with the sham acupuncture
The 4 prior studies of acupuncture for aromatase inhibi- group believed they were receiving true acupuncture, sug-
tor arthralgias did not evaluate maintenance or durability of gesting the possibility of lack of successful masking of this
effect.10,11,36,37 Most studies of acupuncture in patients with group. Third, primary outcomes were based on relatively short-
cancer have evaluated the benefits of true acupuncture term measurements, and long-term follow-up beyond 12
among women with hot flashes, and several evaluated persis- months was not available. Fourth, the study design did not in-
tent effects following completion of the intervention, sug- corporate the pain literature regarding a comparison be-
gesting that the effects may be long lasting. For example, in a tween groups of a clinically meaningful reduction for an in-
randomized unblinded trial of weekly acupuncture for 12 dividual, and therefore, these results are post hoc. Fifth, there
weeks, the intervention was associated with a significantly was no correction for multiple comparisons for the second-
lower hot flash score at 3 months and also at 6 months fol- ary analyses, and therefore, these findings should be consid-
lowing completion of the acupuncture intervention.38 Acu- ered exploratory. Sixth, accrual was slower than expected due
puncture is appealing to some patients because the adverse to acupuncture staff turnover and retraining, expansion of
effects are generally limited compared with medications study sites, scheduling difficulties, and the widespread avail-
used to control pain, such as opiates or duloxetine. However, ability of acupuncture, limiting patients’ enthusiasm for en-
despite the benefits and limited toxicities, acupuncture is not tering a randomized trial. Seventh, uptake in the community
covered by many insurance plans. may be limited by the expense of the intervention, logistical
This study has several strengths. The intervention was challenges, and uncertain experience of the acupuncturist with
conducted at multiple sites by acupuncturists who under- the study methodology.
went rigorous standardized training and administered the
repeat assessments throughout the time frame of the study. Conclusion
Acupuncturists had diverse training backgrounds, and Among postmenopausal women with early-stage breast can-
patients were treated at both academic and community sites cer and aromatase inhibitor–related arthralgias, true acu-
throughout the United States, both of which increase the puncture compared with sham acupuncture or with waitlist
generalizability of the findings. The retention and comple- control, resulted in a statistically significant reduction in
tion rates were similar across groups, reducing the concern joint pain at 6 weeks, although the observed improvement
about differential dropout. was of uncertain clinical importance.

ARTICLE INFORMATION Critical revision of the manuscript for important Disclaimer: The content is solely the responsibility
Accepted for Publication: June 6, 2018. intellectual content: Hershman, Unger, Greenlee, of the authors and does not necessarily represent
Lew, Darke, Kengla, Melnik, Jorgensen, Minasian, the official views of the NIH.
Author Affiliations: Columbia University Medical Fisch, Henry, Crew.
Center, New York, New York (Hershman); Fred Meeting Presentation: This research was
Statistical analysis: Hershman, Unger, Lew, Darke. presented at the San Antonio Breast Cancer
Hutchinson Cancer Research Center, Seattle, Obtained funding: Hershman, Crew.
Washington (Unger, Greenlee, Lew, Darke); SWOG Symposium, December 2017.
Administrative, technical, or material support:
Statistics and Data Management Center, Seattle, Hershman, Greenlee, Capodice, Jorgensen, Crew.
Washington (Unger, Lew, Darke); Mount Sinai REFERENCES
Supervision: Hershman, Unger, Greenlee, Capodice,
Hospital, New York, New York (Capodice); Kaiser Kengla, Crew. 1. Burstein HJ, Lacchetti C, Anderson H, et al.
Permanente Medical Center, Walnut Creek, Adjuvant endocrine therapy for women with
California (Kengla); Spectrum Health Medical Conflict of Interest Disclosures: All authors have hormone receptor-positive breast cancer: American
Group, Grand Rapids, Michigan (Melnik); NCORP of completed and submitted the ICMJE Form for Society of Clinical Oncology Clinical Practice
the Carolinas (Greenville Health System), Disclosure of Potential Conflicts of Interest. Ms Guideline Update on Ovarian Suppression. J Clin
Greenville, South Carolina (Jorgensen); St Luke’s Darke reports receipt of grants from the National Oncol. 2016;34(14):1689-1701.
Mountain States Tumor Institute (PCRC NCORP), Cancer Institute (NCI) during the conduct of the
study. No other disclosures were reported. 2. Crew KD, Greenlee H, Capodice J, et al.
Boise, Idaho (Kreisle); Division of Cancer Prevalence of joint symptoms in postmenopausal
Prevention, National Cancer Institute, Bethesda, Funding/Support: Funding was provided by the women taking aromatase inhibitors for early-stage
Maryland (Minasian); AIM Specialty Health, National Institutes of Health (NIH) National Center breast cancer. J Clin Oncol. 2007;25(25):3877-3883.
Chicago, Illinois (Fisch); University of Utah for Complementary and Integrative Health and
Huntsman Cancer Institute, Salt Lake City (Henry). the Office of Research on Women’s Health 3. Castel LD, Hartmann KE, Mayer IA, et al. Time
R01AT006376; NIH/NCI/DCP (Division of Cancer course of arthralgia among women initiating
Author Contributions: Drs Hershman and Unger aromatase inhibitor therapy and a postmenopausal
had full access to all of the data in the study and Prevention) grant UG1CA189974 and legacy grant
U10CA37429. comparison group in a prospective cohort. Cancer.
takes responsibility for the integrity of the data 2013;119(13):2375-2382.
and the accuracy of the data analysis. Role of the Funder: The funder had no role in the
Concept and design: Hershman, Unger, Greenlee, design and conduct of the study; collection, 4. Henry NL, Azzouz F, Desta Z, et al. Predictors of
Capodice, Minasian, Crew. management, analysis, and interpretation of the aromatase inhibitor discontinuation as a result of
Acquisition, analysis, or interpretation of data: data; preparation, review or approval of the treatment-emergent symptoms in early-stage
Hershman, Unger, Greenlee, Lew, Darke, Kengla, manuscript; and decision to submit the manuscript breast cancer. J Clin Oncol. 2012;30(9):936-942.
Melnik, Jorgensen, Kreisle, Fisch, Henry, Crew. for publication. 5. Hershman DL, Kushi LH, Shao T, et al. Early
Drafting of the manuscript: Hershman, Unger, discontinuation and nonadherence to adjuvant
Capodice, Kreisle, Crew.

jama.com (Reprinted) JAMA July 10, 2018 Volume 320, Number 2 175

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
Research Original Investigation Acupuncture vs Control for Joint Pain Related to Aromatase Inhibitor Treatment for Breast Cancer

hormonal therapy in a cohort of 8,769 early-stage 17. Greenlee H, Crew KD, Capodice J, et al. outcomes in chronic pain clinical trials: IMMPACT
breast cancer patients. J Clin Oncol. 2010;28(27): Methods to standardize a multicenter acupuncture recommendations. J Pain. 2008;9(2):105-121.
4120-4128. trial protocol to reduce aromatase inhibitor-related 29. Zou G. A modified poisson regression approach
6. Goss PE, Ingle JN, Pritchard KI, et al. Extending joint symptoms in breast cancer patients. to prospective studies with binary data. Am J
Aromatase-Inhibitor Adjuvant Therapy to 10 Years. J Acupunct Meridian Stud. 2015;8(3):152-158. Epidemiol. 2004;159(7):702-706.
N Engl J Med. 2016;375(3):209-219. 18. Hui KK, Nixon EE, Vangel MG, et al. 30. SAS Institute Inc. SAS 9.4 System Options. 5th
7. Chirgwin JH, Giobbie-Hurder A, Coates AS, et al. Characterization of the “deqi” response in ed. Cary, NC: SAS Institute Inc; 2016.
Treatment adherence and its impact on acupuncture. BMC Complement Altern Med.
2007;7:33. 31. Smith EM, Pang H, Cirrincione C, et al; Alliance
disease-free survival in the breast international for Clinical Trials in Oncology. Effect of duloxetine
group 1-98 trial of tamoxifen and letrozole, alone 19. Hershman DL, Unger JM, Crew KD, et al. on pain, function, and quality of life among patients
and in sequence. J Clin Oncol. 2016;34(21):2452- Randomized multicenter placebo-controlled trial of with chemotherapy-induced painful peripheral
2459. omega-3 fatty acids for the control of aromatase neuropathy. JAMA. 2013;309(13):1359-1367.
8. Helms J. Acupuncture Energetics: A Clinical inhibitor-induced musculoskeletal pain: SWOG
S0927. J Clin Oncol. 2015;33(17):1910-1917. 32. Kroenke K, Theobald D, Wu J, et al. Effect of
Approach for Physicians. Berkeley, CA: Medical telecare management on pain and depression in
Acupuncture Publishers; 1995. 20. Irwin ML, Cartmel B, Gross CP, et al. patients with cancer. JAMA. 2010;304(2):163-171.
9. Tedesco D, Gori D, Desai KR, et al. Drug-free Randomized exercise trial of aromatase
inhibitor-induced arthralgia in breast cancer 33. Shapiro AC, Adlis SA, Robien K, et al.
interventions to reduce pain or opioid consumption Randomized, blinded trial of vitamin D3 for treating
after total knee arthroplasty. JAMA Surg. 2017;152 survivors. J Clin Oncol. 2015;33(10):1104-1111.
aromatase inhibitor-associated musculoskeletal
(10):e172872. 21. Farrar JT, Young JP Jr, LaMoreaux L, Werth JL, symptoms (AIMSS). Breast Cancer Res Treat. 2016;
10. Crew KD, Capodice JL, Greenlee H, et al. Poole RM. Clinical importance of changes in chronic 155(3):501-512.
Randomized, blinded, sham-controlled trial of pain intensity measured on an 11-point numerical
pain rating scale. Pain. 2001;94(2):149-158. 34. Henry NL, Unger JM, Schott AF, et al.
acupuncture for the management of aromatase Randomized, multicenter, placebo-controlled
inhibitor-associated joint symptoms in women with 22. Bellamy N, Buchanan WW, Goldsmith CH, clinical trial of duloxetine versus placebo for
early-stage breast cancer. J Clin Oncol. 2010;28(7): Campbell J, Stitt LW. Validation study of WOMAC: aromatase inhibitor-associated arthralgias in
1154-1160. a health status instrument for measuring clinically early-stage breast cancer: SWOG S1202. J Clin Oncol.
11. Mao JJ, Xie SX, Farrar JT, et al. A randomised important patient relevant outcomes to 2018;36(4):326-332.
trial of electro-acupuncture for arthralgia related to antirheumatic drug therapy in patients with
osteoarthritis of the hip or knee. J Rheumatol. 1988; 35. Henry NL, Griggs JJ. The power of the placebo
aromatase inhibitor use. Eur J Cancer. 2014;50(2): in symptom management. J Clin Oncol. 2015;33(17):
267-276. 15(12):1833-1840.
1870-1872.
12. Pocock SJ, Simon R. Sequential treatment 23. Sautner J, Andel I, Rintelen B, Leeb BF.
Development of the M-SACRAH, a modified, 36. Bao T, Cai L, Giles JT, et al. A dual-center
assignment with balancing for prognostic factors in randomized controlled double blind trial assessing
the controlled clinical trial. Biometrics. 1975;31(1): shortened version of SACRAH (Score for the
Assessment and Quantification of Chronic the effect of acupuncture in reducing
103-115. musculoskeletal symptoms in breast cancer
Rheumatoid Affections of the Hands).
13. MacPherson H, Altman DG, Hammerschlag R, Rheumatology (Oxford). 2004;43(11):1409-1413. patients taking aromatase inhibitors. Breast Cancer
et al; STRICTA Revision Group. Revised standards Res Treat. 2013;138(1):167-174.
for reporting interventions in clinical trials of 24. Fallowfield LJ, Leaity SK, Howell A, Benson S,
Cella D. Assessment of quality of life in women 37. Oh B, Kimble B, Costa DS, et al. Acupuncture
acupuncture (STRICTA): extending the CONSORT for treatment of arthralgia secondary to aromatase
statement. J Evid Based Med. 2010;3(3):140-155. undergoing hormonal therapy for breast cancer.
Breast Cancer Res Treat. 1999;55(2):189-199. inhibitor therapy in women with early breast
14. National Institutes of Health. Racial and ethnic cancer. Acupunct Med. 2013;31(3):264-271.
categories and definitions for NIH diversity 25. Garcia SF, Cella D, Clauser SB, et al.
Standardizing patient-reported outcomes 38. Lesi G, Razzini G, Musti MA, et al. Acupuncture
programs and for other reporting purposes. Notice as an integrative approach for the treatment of hot
number NOT-OD-15-089. https://grants.nih.gov assessment in cancer clinical trials. J Clin Oncol.
2007;25(32):5106-5112. flashes in women with breast cancer. J Clin Oncol.
/grants/guide/notice-files/not-od-15-089.html. 2016;34(15):1795-1802.
April 8, 2015. Accessed June 21, 2018. 26. Laird NM, Ware JH. Random-effects models for
15. Oken MM, Creech RH, Tormey DC, et al. Toxicity longitudinal data. Biometrics. 1982;38(4):963-974.
and response criteria of the Eastern Cooperative 27. Dworkin RH, Turk DC, McDermott MP, et al.
Oncology Group. Am J Clin Oncol. 1982;5(6):649-655. Interpreting the clinical importance of group
16. Daut RL, Cleeland CS, Flanery RC. Development differences in chronic pain clinical trials: IMMPACT
of the Wisconsin Brief Pain Questionnaire to assess recommendations. Pain. 2009;146(3):238-244.
pain in cancer and other diseases. Pain. 1983;17(2): 28. Dworkin RH, Turk DC, Wyrwich KW, et al
197-210. Interpreting the clinical importance of treatment

176 JAMA July 10, 2018 Volume 320, Number 2 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: by a Tufts Univ. Hirsh Health Sciences Library User on 07/10/2018
original reports
Dietary Supplement Use During Chemotherapy
and Survival Outcomes of Patients With Breast
Cancer Enrolled in a Cooperative Group Clinical
Trial (SWOG S0221)
Christine B. Ambrosone, PhD1; Gary R. Zirpoli, PhD2; Alan D. Hutson, PhD1; William E. McCann1; Susan E. McCann, PhD, RD1;
William E. Barlow, PhD3; Kara M. Kelly, MD1; Rikki Cannioto, PhD, EdD1; Lara E. Sucheston-Campbell, PhD4; Dawn L. Hershman, MD5;
Joseph M. Unger, PhD3; Halle C.F. Moore, MD6; James A. Stewart, MD7; Claudine Isaacs, MD8; Timothy J. Hobday, MD9;
Muhammad Salim, MD10; Gabriel N. Hortobagyi, MD11; Julie R. Gralow, MD12; George T. Budd, MD6; and Kathy S. Albain, MD13
abstract

PURPOSE Despite reported widespread use of dietary supplements during cancer treatment, few empirical data
with regard to their safety or efficacy exist. Because of concerns that some supplements, particularly anti-
oxidants, could reduce the cytotoxicity of chemotherapy, we conducted a prospective study ancillary to
a therapeutic trial to evaluate associations between supplement use and breast cancer outcomes.
METHODS Patients with breast cancer randomly assigned to an intergroup metronomic trial of cyclophos-
phamide, doxorubicin, and paclitaxel were queried on their use of supplements at registration and during
treatment (n =1,134). Cox proportional hazards regression adjusting for clinical and lifestyle variables was used.
Recurrence and survival were indexed at 6 months after enrollment using a landmark approach.
RESULTS There were indications that use of any antioxidant supplement (vitamins A, C, and E; carotenoids;
coenzyme Q10) both before and during treatment was associated with an increased hazard of recurrence
(adjusted hazard ratio [adjHR], 1.41; 95% CI, 0.98 to 2.04; P = .06) and, to a lesser extent, death (adjHR, 1.40;
95% CI, 0.90 to 2.18; P = .14). Relationships with individual antioxidants were weaker perhaps because of small
numbers. For nonantioxidants, vitamin B12 use both before and during chemotherapy was significantly as-
sociated with poorer disease-free survival (adjHR, 1.83; 95% CI, 1.15 to 2.92; P , .01) and overall survival
(adjHR, 2.04; 95% CI, 1.22 to 3.40; P , .01). Use of iron during chemotherapy was significantly associated with
recurrence (adjHR, 1.79; 95% CI, 1.20 to 2.67; P , .01) as was use both before and during treatment (adjHR,
1.91; 95% CI, 0.98 to 3.70; P = .06). Results were similar for overall survival. Multivitamin use was not as-
sociated with survival outcomes.
CONCLUSION Associations between survival outcomes and use of antioxidant and other dietary supplements
both before and during chemotherapy are consistent with recommendations for caution among patients when
considering the use of supplements, other than a multivitamin, during chemotherapy.
J Clin Oncol 38:804-814. © 2019 by American Society of Clinical Oncology
ASSOCIATED
CONTENT
Appendix INTRODUCTION affecting treatment efficacy. To address this question
Data Supplement Use of dietary supplements after a cancer diagnosis is prospectively, we initiated the Diet, Exercise, Lifestyle
Author affiliations common1,2; however, few empirical data provide a basis and Cancer Prognosis (DELCaP) study,8 a correlative
and support
for clinical recommendations for use during chemo- to a phase III trial led by SWOG (S0221) that randomly
information (if assigned patients with breast cancer to different
applicable) appear therapy.3 Because one cytotoxic mechanism of cancer
therapeutics is through the generation of reactive oxygen treatment schedules with doxorubicin, cyclophos-
at the end of this
article. species (ROS), there has been concern that use of di- phamide, and paclitaxel9 and queried participants on
Accepted on October etary supplements during treatment, particularly anti- their use of supplements at randomization and at
9, 2019 and oxidants, could reduce treatment efficacy.4 In fact, there completion of chemotherapy. We previously reported
published at
have been clinical recommendations that patients not on the prevalence of supplement use in S0221, par-
ascopubs.org/journal/ ticularly in relation to recommendations from treating
jco on December 19, take antioxidant supplements during chemotherapy.2,5-7
oncologists8 and in relation to chemotherapy-induced
2019: DOI https://doi.
org/10.1200/JCO.19. The use of supplements, however, could possibly peripheral neuropathy (CIPN).10 No associations were
01203 reduce treatment-related adverse effects while not found between the use of antioxidants and CIPN, but

804 Volume 38, Issue 8


Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106
Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

there was reduced risk of CIPN with use of multivitamins Center.12 If surveys were not returned within 2 weeks, each
before diagnosis and, to a lesser extent, during chemo- patient was called with a reminder and an offer to complete
therapy. These associations, however, must be balanced the questionnaire over the telephone, followed by monthly
against potential adverse effects of supplement use on calls if no response. After 4 months of nonresponse, the
recurrence and mortality. Here, we address the primary patient was withdrawn as a passive refuser. For Q1 late
goal of the DELCaP study: To determine whether use of returns, 17% of surveys were received between 3 and
supplements during chemotherapy, particularly antioxi- 6 months after having been sent, and 2% were received
dants, has any effects on survival outcomes. after 6 months. Approximately 6 months after randomi-
zation, when chemotherapy was scheduled to be com-
pleted, a second questionnaire (Q2) was sent to patients
METHODS
who had completed Q1 (n = 1,340; 83%) that queried
Study Population about supplement use during chemotherapy; 1,134 (85%)
DELCaP began approximately 20 months after S0221 of these patients completed Q2. An example of questions
opened, with information embedded in the informed con- for Q1 and Q2 is shown in Data Supplement. Question-
sent for the trial as previously described.8 Of the 2,014 naires were reviewed for missing data, and patients with
patients who were eligible, 1,607 (80%) agreed to partic- missing responses were contacted to maximize complete
ipate in DELCaP (Fig 1). After initial contact, a baseline data. Reasons for not returning questionnaires are shown in
questionnaire (Q1) was sent for completion before begin- Figure 1, with passive refusal referring to patients who did
ning chemotherapy, which queried about regular use not return questionnaires but did not formally withdraw
(at least once per week) before diagnosis and between from the study. Patients who did not return Q2 were sig-
diagnosis and enrollment in the trial. The validated sur- nificantly more likely than those who did to be nonwhite, to
vey was adapted from the VITAL study11 and piloted be younger (48.35 v 51.65 years), and to have less grade 3
among patients with cancer enrolled in the DataBank and and 4 hematologic toxicity (5.16% v 11.73%). For a limited
BioRepository at Roswell Park Comprehensive Cancer number of institutions that did not allow contact with their

Randomly assigned to SWOG S0221 (N = 2,716) Enrolled in S0221 before DELCaP


Eligible for DELCaP (n = 2,014) (n = 702)

Did not consent to DELCaP


(n = 407)

FIG 1. Study schema for participants


included in the Diet, Exercise, Life-
Consented to DELCaP ancillary style and Cancer Prognosis (DELCaP)
(questionnaire component; n = 1,607) analysis, an observational study an-
Did not complete Q1 cillary to SWOG S0221, a randomized
(n = 267) treatment trial for high-risk breast
CRA Mail Total cancer. CRA, clinical research as-
Deceased 1 2 3 sociate; Q1, questionnaire 1 completed
Not eligible 5 25 30 at registration in the trial before initi-
Passive refusal 5 205 210 ation of chemotherapy; Q2, ques-
Active refusal 1 19 20 tionnaire 2 completed at completion
of active treatment (approximately
Completed Q1 (n = 1,340)
CRA administered (n = 119) 6 months after registration in S0221;
Did not complete Q2 ‘other’ patients were collapsed into
(n = 206) other categories). *Other: mental
CRA Mail Total disability, loss of vision, physician re-
Deceased 0 4 4 moved patient from study, dropped
Not eligible 0 1 1 on suspicion of metastasis.
Passive refusal 6 178 184
Active refusal 3 14 17
Total 9 197 206
Completed Q1 and Q2 (n = 1,134)
CRA administered (n = 43)

Journal of Clinical Oncology 805

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

patients (14 of 320), nurses or clinical research associates were included in the passive refusers category. All statistical
administered the questionnaires; this applied to 8% of tests were two-sided, and P , .05 was considered statis-
participants. This study was approved by the institutional tically significant. All analyses were performed using SAS
review boards at Roswell Park Comprehensive Cancer 9.4 statistical software (SAS Institute, Cary NC).
Center and at institutions participating in S0221.
Statistical Analyses RESULTS
A landmark survival analysis was performed that desig- Characteristics of the 1,340 patients who completed Q1 are
nated a time point that occurred during the follow-up period listed in Table 1. Patients who experienced recurrence
(landmark time) and analyzed only those patients who (n = 310) or died (n = 222) were more likely to be older, to
survived to that point13; here, the 6-month survey was the be postmenopausal, and to have a higher body mass index
landmark time point. Among patients alive without re- (all P , .05); to have completed grade school or some high
currence at 6 months, disease-free survival (DFS) was school (P = .03); to self-report as black (P = .10); and to
defined as the time from then until the first documentation have poor prognosis factors ($ 4 positive lymph nodes, ER
of breast cancer recurrence, new breast cancer, or death, negativity, progesterone receptor negativity, human epi-
whichever came first. Patients who had not experienced dermal growth factor receptor 2 negativity; P , .01).
recurrence were censored on the date of last clinical
Among the 1,134 patients who completed both Q1 and Q2,
contact. Overall survival (OS) was defined as the time from
there were 251 recurrences and 181 deaths. As previously
6 months after enrollment until death as a result of any
reported8 and listed in Appendix Table A1 (online only), the
cause, with patients who were still alive censored on their
prevalence of supplement use, particularly antioxidants,
last follow-up date. The median follow-up time for those
was low compared with reports in the literature of use by
who did not have an event was 8.1 years. Baseline char-
patients with cancer1,2 and tended to decrease during
acteristics in relation to DFS and OS were compared, and
treatment. For example, vitamin C was used by 20.5% of
Cox proportional hazards regressions were used to evaluate
patients before treatment but only 12.2% during therapy.
associations. Because use of supplements before enroll-
Vitamins E and A were taken during treatment by , 10% of
ment may influence relationships between use during
patients. Use of any antioxidant during treatment (vitamins
treatment and outcomes, we categorized use (yes [Y]/no
C, A, and E; carotenoids; or coenzyme Q10) was observed
[N]) at baseline and during chemotherapy combined as
among 17.5% of patients, whereas 44% of patients took
NN, NY, YN, and YY, with the NN group (did not use at
multivitamins during chemotherapy. Appendix Table A2
either time) set as the referent. Tests for proportionality
(online only) lists the prevalence of use at both time points
using time-varying covariates were carried out at a = .05 for
according to DFS and OS.
YY, NY, and YN versus NN in the univariable setting;
no violations were observed. Minimally adjusted models Table 2 lists the relationships between DFS and OS and
included dosing arm as a stratification factor that corre- antioxidant supplement use before and during chemo-
sponded to the original randomized treatment assign- therapy. In fully adjusted models, patients who used any
ment and age (model 1). Fully adjusted models also antioxidant both before and during treatment were at an
included tumor characteristics (estrogen receptor [ER], increased hazard of recurrence (adjusted hazard ratio
progesterone receptor, human epidermal growth factor [adjHR], 1.41; 95% CI, 0.98 to 2.04; P = .06; Fig 2A) and,
receptor 2, lymph node status, and tumor size [stage was to a lesser degree, death (HR, 1.40; 95% CI, 0.90 to 2.18;
not available]), grades 3 or 4 hematologic toxicity, body Fig 2B). There were no relationships with outcomes for use
mass index, physical activity, smoking, and alcohol con- of antioxidants only before treatment initiation or only
sumption (model 2). To account for additional sources during chemotherapy.
of antioxidants and the effects of use of multiple supple- Associations for the majority of single antioxidant supple-
ments, we further adjusted for multivitamin use (model 3). ments were not statistically significant. There was a sug-
In sensitivity analyses, patients who took five or more gestion that use of vitamins C, E, and coenzyme Q10 before
supplements (n = 189) were removed from analysis. In and during treatment was associated with an increased
exploratory analyses, we created a variable for number of hazard of recurrence (adjHR, 1.36 [95% CI, 0.87 to 2.13],
supplements, which was considered in models, and per- 1.47 [95% CI, 0.81 to 2.67], and 1.75 [95% CI, 0.77 to
formed further analyses stratified by ER status. 4.01], respectively), with similar HRs for OS. Although the
The landmark analysis at 6 months was based on the Q2 number of users was quite low (n = 5), use of vitamin A both
survey response among patients who also completed Q1. before and during chemotherapy was associated with
The small fraction of OS events in the first 6 months (n = 4) a significantly increased hazard of recurrence and death,
precluded those patients from completing the 6-month with similar relationships for use of carotenoid supple-
survey; thus, among patients who returned Q2, there ments. Inclusion of multivitamin use in fully adjusted
were no DFS events within the first 6 months. Patients who models resulted in slightly attenuated estimates for some
did not complete Q2 and died after the landmark time point variables (Table 2).

806 © 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

TABLE 1. Baseline Demographic and Clinical Characteristics of the Overall DELCaP Study Population in S0221 by DFS and OS Status
(n = 1,340)
DFS, No. (%) OS, No. (%)

Characteristic Progression Free Progression P Alive Deceased P


No. of patients 1,030 (76.87) 310 (23.13) 1,118 (83.44) 222 (16.56)
Demographic
Mean age, years (SD) 50.87 (9.64) 52.75 (10.56) , .01* 50.89 (9.72) 53.39 (10.47) , .01
Mean BMI, kg/m2 (SD) 29.14 (6.75) 30.16 (7.05) .02 29.15 (6.77) 30.49 (7.07) , .01
Menopausal status , .01 , .01
Pre 506 (80.32) 124 (19.68) 545 (86.51) 85 (13.49)
Post 515 (73.99) 181 (26.01) 563 (80.89) 133 (19.11)
Self-reported race .38 .10
White 864 (77.28) 254 (22.72) 937 (83.81) 181 (16.19)
Black 64 (68.09) 30 (31.91) 69 (73.40) 25 (26.60)
Multiracial 35 (77.78) 10 (22.22) 38 (84.44) 7 (15.56)
American Indian 10 (76.92) 3 (23.08) 11 (84.62) 2 (15.38)
Asian/Pacific Islander 40 (83.33) 8 (16.67) 44 (91.17) 4 (0.08)
Other 14 (77.78) 4 (22.22) 15 (83.33) 3 (16.67)
Self-reported ethnicity .52 .73
Non-Hispanic 978 (76.95) 293 (23.05) 1,059 (83.32) 212 (16.68)
Hispanic 44 (73.33) 16 (26.67) 51 (85.00) 9 (15.00)
Education .06 .03
Grade school or some high school 61 (65.59) 32 (34.41) 69 (74.19) 24 (25.81)
High school graduate or GED 219 (76.31) 68 (23.69) 234 (81.53) 53 (18.47)
Some college or technical school 380 (78.51) 104 (21.49) 412 (85.12) 72 (14.88)
College graduate 227 (79.37) 59 (20.63) 248 (86.71) 38 (13.29)
Advanced degree 137 (70.46) 47 (25.54) 149 (80.98) 35 (19.02)
Clinical
Nodal status , .01 , .01
Negative 290 (83.09) 59 (16.91) 309 (88.54) 40 (11.46)
1-3 positive nodes 406 (80.88) 96 (19.12) 433 (86.25) 69 (13.75)
$ 4 positive nodes 332 (68.17) 155 (31.83) 374 (76.80) 113 (23.20)
ER/PR status .01 , .01
Positive (either or both positive) 690 (79.04) 183 (20.96) 751 (86.03) 122 (13.97)
Negative (both negative) 338 (72.84) 126 (27.16) 365 (78.66) 99 (21.34)
HER2 status , .01 .02
Negative 793 (75.17) 262 (24.83) 867 (82.18) 188 (17.82)
Positive 231 (82.80) 48 (17.20) 245 (87.81) 34 (12.19)

NOTE. P values reflect pooled t test for age and BMI.


Abbreviations: BMI, body mass index; DELCaP, Diet, Exercise, Lifestyle and Cancer Prognosis; DFS, disease-free survival; ER, estrogen
receptor; GED, general educational development; HER2, human epidermal growth factor receptor 2; OS, overall survival; PR, progesterone
receptor; SD, standard deviation.
*Satterthwaite approximation for age and progression-free survival (rejecting equal variance at P # .05); otherwise, x2 test.

Table 3 lists associations with nonantioxidant supplements. treatment outcomes and use of vitamin B12 and iron
Multivitamin use before, during, or at both time points was supplements (Figs 3A and 3B). Use of vitamin B12 both
not associated with survival outcomes (Figs 2C and 2D), nor before and during treatment was associated with poorer DFS
was vitamin D use. Striking associations were found between (HR, 1.83; 95% CI, 1.15 to 2.92) and OS (HR, 2.04; 95% CI,

Journal of Clinical Oncology 807

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

TABLE 2. Adjusted Models for Associations Between Use of Antioxidant Supplements Before and During Chemotherapy and DFS and OS in S0221: DELCaP
Study Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134)
Reported DFS, HR (95% CI) OS, HR (95% CI)
Antioxidant Use
Before and
During Fully Adjusted With Fully Adjusted With
Chemotherapy Minimally Adjusted* Fully Adjusted† MV Use‡ Minimally Adjusted* Fully Adjusted† MV Use‡
Any
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 0.79 (0.43 to 1.43) 0.93 (0.52 to 1.64) 0.92 (0.52 to 1.64) 0.97 (0.50 to 1.87) 1.03 (0.53 to 1.98) 1.01 (0.52 to 1.97)
YN 1.02 (0.74 to 1.43) 1.10 (0.79 to 1.53) 1.04 (0.74 to 1.47) 1.08 (0.27 to 1.58) 1.19 (0.81 to 1.76) 1.11 (0.75 to 1.66)
YY 1.42 (1.00 to 2.03) 1.41 (0.98 to 2.04) 1.38 (0.94 to 2.02) 1.37 (0.90 to 2.09) 1.40 (0.90 to 2.18) 1.34 (0.85 to 2.13)
Vitamin C
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.01 (0.56 to 1.83) 1.14 (0.65 to 2.01) 1.14 (0.64 to 2.03) 1.11 (0.56 to 2.20) 1.14 (0.58 to 2.27) 1.15 (0.58 to 2.31)
YN 1.05 (0.73 to 1.50) 1.10 (0.76 to 1.59) 1.04 (0.72 to 1.52) 1.19 (0.79 to 1.79) 1.35 (0.89 to 2.04) 1.27 (0.83 to 1.93)
YY 1.37 (0.89 to 2.11) 1.36 (0.87 to 2.13) 1.31 (0.83 to 2.08) 1.42 (0.86 to 2.34) 1.42 (0.83 to 2.40) 1.37 (0.80 to 2.34)
Vitamin A
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.61 (0.78 to 3.32) 1.51 (0.71 to 3.23) 1.51 (0.70 to 3.29) 1.27 (0.46 to 3.49) 1.20 (0.44 to 3.28) 1.25 (0.45 to 3.49)
YN 0.85 (0.39 to 1.87) 0.75 (0.33 to 1.71) 0.71 (0.31 to 1.63) 0.77 (0.29 to 2.05) 0.71 (0.26 to 1.94) 0.66 (0.24 to 1.83)
YY 4.05 (1.46 to 11.25) 4.23 (1.32 to 13.57) 4.06 (1.26 to 13.16) 4.14 (1.81 to 9.51) 3.53 (1.05 to 11.93) 3.20 (0.93 to 10.99)
Vitamin E
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.16 (0.60 to 2.24) 1.15 (0.60 to 2.18) 1.13 (0.59 to 2.16) 1.13 (0.52 to 2.49) 1.22 (0.56 to 2.62) 1.19 (0.55 to 2.58)
YN 0.99 (0.68 to 1.43) 1.03 (0.71 to 1.50) 0.98 (0.67 to 1.44) 1.0 (0.66 to 1.53) 1.10 (0.71 to 1.71) 1.04 (0.66 to 1.62)
YY 1.62 (0.92 to 2.88) 1.47 (0.81 to 2.67) 1.38 (0.75 to 2.54) 1.65 (0.86 to 3.17) 1.53 (0.77 to 3.04) 1.39 (0.68 to 2.82)
Coenzyme Q10
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.16 (0.53 to 2.56) 1.34 (0.59 to 3.02) 1.35 (0.59 to 3.06) 1.11 (0.42 to 2.92) 1.28 (0.47 to 3.48) 1.34 (0.49 to 3.67)
YN 1.33 (0.70 to 2.52) 1.30 (0.66 to 2.56) 1.28 (0.65 to 2.51) 1.16 (0.54 to 2.47) 1.13 (0.50 to 2.58) 1.08 (0.47 to 2.48)
YY 2.02 (0.92 to 4.47) 1.75 (0.77 to 4.01) 1.68 (0.73 to 3.89) 2.20 (0.92 to 5.26) 2.04 (0.82 to 5.09) 1.88 (0.75 to 4.76)
Carotenoid
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 2.59 (0.78 to 8.57) 3.15 (1.14 to 8.65) 3.20 (1.16 to 8.87) 3.59 (0.93 to 13.84) 2.98 (0.91 to 9.73) 3.21 (0.97 to 10.61)
YN 1.46 (0.62 to 3.44) 1.04 (0.38 to 2.83) 0.99 (0.36 to 2.70) 0.83 (0.20 to 3.44) 0.80 (0.20 to 3.28) 0.74 (0.18 to 3.04)
YY 4.03 (1.12 to 14.50) 2.46 (0.75 to 8.03) 2.24 (0.68 to 7.37) 2.68 (0.87 to 8.27) 1.74 (0.41 to 7.42) 1.50 (0.35 to 6.55)

NOTE. Use of vitamin C, vitamin A, vitamin E, carotenoids, or coenzyme Q10.


Abbreviations: DELCaP, Diet, Exercise, Lifestyle and Cancer Prognosis; DFS, disease-free survival; HR, hazard ratio; MV, multivitamin; NN, neither before
nor during; NY, not before but during; OS, overall survival; YN, before but not during; YY, both before and during.
*Stratified on treatment assignment and adjusted for age.
†Stratified on treatment assignment and adjusted for age, tumor characteristics (estrogen receptor, progesterone receptor, human epidermal growth factor
receptor 2, lymph node status, and tumor size), grades 3 or 4 hematologic toxicity, body mass index, physical activity, smoking, and alcohol consumption.
‡Stratified on treatment assignment and adjusted for age, tumor characteristics (estrogen receptor, progesterone receptor, human epidermal growth factor
receptor 2, lymph node status, and tumor size), grades 3 or 4 hematologic toxicity, body mass index, physical activity, smoking, alcohol consumption, and
MV use.

1.22 to 3.40), as was iron supplementation, with greater fatty acids both before and during treatment was associated
recurrence with use during treatment (HR, 1.79; 95% CI, with DFS (HR, 1.67; 95% CI, 1.12 to 2.49) but not OS. Other
1.20 to 2.67) and with use both before and during che- supplements commonly taken were not associated with
motherapy (HR, 1.91; 95% CI, 0.98 to 3.70). There were poorer outcomes in adjusted models. The addition of pol-
similar relationships with death (Fig 3D). Use of omega-3 ysupplement use to models was nonsignificant and not

808 © 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

A B
1.0 1.0
Log-rank P = .1579 Log-rank P = .3323
DFS (probability)

OS (probability)
0.8 0.8

0.6 0.6

0.4 NN 0.4 NN
NY NY
0.2 0.2
YN YN
0.2 YY 0.2 YY

0 24 48 72 96 120 144 0 24 48 72 96 120 144


Time (months) Time (months)
No. at risk: No. at risk:
NN 739 721 693 657 626 583 451 388 316 232 107 22 0 NN 739 736 717 695 669 624 487 421 347 254 117 25 0
NY 75 72 70 68 63 62 45 40 32 25 10 1 0 NY 75 75 73 72 68 63 45 41 33 26 11 1 0
YN 207 200 190 183 178 162 133 118 93 58 30 4 0 YN 207 206 200 194 190 177 147 129 102 65 33 6 0
YY 126 125 113 108 103 89 75 57 43 30 14 4 0 YY 126 126 122 116 111 101 85 65 48 34 16 4 0

C D
1.0 1.0
Log-rank P = .6834 Log-rank P = .4326
DFS (probability)

OS (probablity)
0.8 0.8

0.6 0.6

0.4 0.4
NN NN
0.2 NY 0.2 NY
YN YN
0.2 YY
0.2 YY

0 24 48 72 96 120 144 0 24 48 72 96 120 144


Time (months) Time (months)
No. at risk: No. at risk:
NN 460 451 431 410 396 365 277 233 184 129 63 12 0 NN 460 458 447 434 421 388 297 250 200 142 67 13 0
NY 128 124 122 118 110 105 87 79 67 53 28 8 0 NY 128 128 123 122 119 113 93 85 73 58 32 8 0
YN 178 172 163 153 146 134 114 99 81 57 19 5 0 YN 178 176 172 166 162 147 126 108 88 63 22 7 0
YY 373 363 342 327 310 284 224 191 151 106 51 6 0 YY 373 373 362 347 328 308 246 212 168 116 56 8 0

FIG 2. Product limit estimates for use of antioxidant supplements and (A) disease-free survival (DFS) and (B) overall survival (B) and for use of
multivitamins and (C) DFS and (D) OS. NN, neither before nor during; NY, not before but during; YN, before but not during; YY, both before and during.

retained in any of the stepwise models (data not shown). weaker association with mortality. Although there were
Analyses stratified by ER status are listed in Appendix Table suggestions of increased risk with individual antioxidants,
A3 (online only). The resultant small sample sizes for the including vitamins A, E, and C and carotenoids and co-
majority of supplements precluded us from drawing con- enzyme Q, the numbers of users and events were small, and
clusions. However, for use of any antioxidant, while re- risk estimates were unstable. Nonetheless, these findings of
lationships for DFS were similar by ER status, the association increased risk of poor outcomes with use of antioxidant
with OS was driven largely by ER-positive breast cancer (HR, supplements are congruent with concerns that use of an-
1.70; 95% CI, 0.99 to 2.92), with no association for ER- tioxidants during chemotherapy could reduce the cytotoxic
negative disease (HR, 0.93; 95% CI, 0.46 to 1.88). effects of ROS generated by numerous chemotherapy
agents4,8,14 and seem to support the recommendations by
DISCUSSION some that antioxidant supplements not be consumed during
cancer therapy.2,5,15 However, a review by Ladas and Kelly16
In this observational study ancillary to an intergroup clinical
in 2010 concluded that insufficient evidence existed with
trial for high-risk breast cancer, we found some support for
regard to safety of dietary supplements to make recom-
the notion that use of dietary supplements during chemo-
mendations, and that may still be the case.
therapy could have a negative impact on recurrence and OS.
When we considered use of any antioxidant (vitamins C, A, No associations were found with regard to use of multivi-
and E; carotenoids; and coenzyme Q10), there was a 41% tamins either before or during chemotherapy, consistent
increase in hazard of recurrence with use both before and with the only other study of lifestyle factors embedded in
during treatment of borderline significance, with a similar but a clinical trial for colon cancer CALGB89803.17 However,

Journal of Clinical Oncology 809

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

TABLE 3. Adjusted Models for Associations Between Use of Dietary Supplements Other Than Antioxidants Before and During Chemotherapy and DFS and
OS in S0221: DELCaP Study Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134)
DFS, HR (95% CI) OS, HR (95% CI)
Reported Supplement Use
Before and During Fully Adjusted With Fully Adjusted With
Chemotherapy Minimally Adjusted* Fully Adjusted† MV Use‡ Minimally Adjusted* Fully Adjusted† MV Use‡
MVs
NN 1.0 1.0 — 1.0 1.0 —
NY 1.02 (0.67 to 1.55) 1.02 (0.67 to 1.56) — 0.92 (0.55 to 1.56) 0.91 (0.54 to 1.55) —
YN 1.20 (0.98 to 1.71) 1.27 (0.88 to 1.84) — 1.20 (0.78 to 1.83) 1.35 (0.87 to 2.09) —
YY 1.11 (0.83 to 1.49) 1.21 (0.90 to 1.64) — 1.20 (0.85 to 1.68) 1.31 (0.92 to 1.88) —
Vitamin D
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.25 (0.86 to 1.81) 1.20 (0.83 to 1.74) 1.19 (0.81 to 1.74) 1.13 (0.73 to 1.75) 1.06 (0.68 to 1.66) 1.05 (0.66 to 1.65)
YN 1.01 (0.67 to 1.51) 1.02 (0.66 to 1.56) 0.96 (0.62 to 1.48) 1.07 (0.67 to 1.71) 1.16 (0.71 to 1.90) 1.07 (0.65 to 1.77)
YY 1.17 (0.81 to 1.70) 1.25 (0.84 to 1.86) 1.22 (0.81 to 1.84) 1.10 (0.71 to 1.70) 1.18 (0.73 to 1.91) 1.11 (0.67 to 1.82)
Vitamin B6
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 0.86 (0.61 to 1.23) 0.90 (0.64 to 1.28) 0.89 (0.63 to 1.27) 0.92 (0.61 to 1.39) 0.98 (0.64 to 1.48) 0.97 (0.64 to 1.47)
YN 0.79 (0.43 to 1.47) 0.70 (0.38 to 1.29) 0.65 (0.35 to 1.22) 0.96 (0.48 to 1.90) 0.96 (0.43 to 1.73) 0.79 (0.39 to 1.60)
YY 1.09 (0.61 to 1.97) 1.11 (0.61 to 2.03) 1.07 (0.58 to 1.96) 1.13 (0.57 to 2.24) 1.20 (0.60 to 2.41) 1.13 (0.56 to 2.29)
Vitamin B12
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.08 (0.66 to 1.75) 1.08 (0.66 to 1.76) 1.08 (0.66 to 1.77) 0.81 (0.42 to 1.58) 0.85 (0.44 to 1.63) 0.85 (0.44 to 1.64)
YN 0.91 (0.55 to 1.51) 0.86 (0.51 to 1.44) 0.80 (0.47 to 1.36) 0.80 (0.42 to 1.52) 0.76 (0.39 to 1.47) 0.70 (0.36 to 1.36)
YY 1.87 (1.20 to 2.98) 1.83 (1.15 to 2.92) 1.77 (1.10 to 2.84) 2.07 (1.37 to 3.38) 2.04 (1.22 to 3.40) 1.91 (1.13 to 3.22)
Iron
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.63 (1.09 to 2.44) 1.79 (1.20 to 2.67) 1.79 (1.18 to 2.70) 1.62 (1.02 to 2.58) 1.71 (1.06 to 2.76) 1.67 (1.02 to 2.72)
YN 0.66 (0.34 to 1.31) 0.60 (0.29 to 1.24) 0.58 (0.28 to 1.19) 0.59 (0.25 to 1.41) 0.54 (0.22 to 1.35) 0.50 (0.20 to 1.26)
YY 2.54 (1.36 to 4.78) 1.91 (0.98 to 3.70) 1.88 (0.96 to 3.67) 2.67 (1.36 to 5.27) 1.90 (0.90 to 4.02) 1.80 (0.85 to 3.84)
Folic acid
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.21 (0.74 to 1.99) 1.22 (0.73 to 2.04) 1.21 (0.72 to 2.04) 1.05 (0.55 to 2.02) 1.11 (0.58 to 2.14) 1.11 (0.58 to 2.16)
YN 0.82 (0.49 to 1.36) 0.76 to 0.45 to 1.28) 0.72 (0.42 to 1.21) 0.75 (0.40 to 1.41) 0.69 (0.40 to 1.33) 0.63 (0.32 to 1.22)
YY 1.32 (0.69 to 2.52) 1.35 (0.71 to 2.60) 1.32 (0.68 to 2.54) 1.54 (0.78 to 3.06) 1.79 (0.89 to 3.59) 1.70 (0.84 to 3.43)
Calcium
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.23 (0.81 to 1.87) 1.18 (0.77 to 1.81) 1.17 (0.76 to 1.80) 1.05 (0.62 to 1.77) 0.99 (0.58 to 1.70) 0.96 (0.55 to 1.66)
YN 1.28 (0.91 to 1.79) 1.31 (0.93 to 1.85) 1.24 (0.87 to 1.78) 1.44 (0.98 to 2.11) 1.60 (1.08 to 2.36) 1.49 (0.99 to 2.24)
YY 1.22 (0.88 to 1.70) 1.26 (0.89 to 1.76) 1.20 (0.84 to 1.74) 1.19 (0.81 to 1.75) 1.31 (0.87 to 1.96) 1.19 (0.77 to 1.84)
Omega-3 fatty acids
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.23 (0.66 to 2.26) 1.15 (0.62 to 2.13) 1.14 (0.61 to 2.12) 1.07 (0.50 to 2.29) 1.03 (0.48 to 2.23) 1.00 (0.46 to 2.18)
YN 0.77 (0.51 to 1.16) 0.82 (0.54 to 1.25) 0.77 (0.50 to 1.18) 0.74 (0.45 to 1.20) 0.88 (0.54 to 1.45) 0.81 (0.49 to 1.34)
YY 1.54 (1.04 to 2.29) 1.67 (1.12 to 2.49) 1.63 (1.07 to 2.48) 1.23 (0.75 to 2.04) 1.35 (0.81 to 2.25) 1.23 (0.72 to 2.10)
Glucosamine
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 0.21 (0.03 to 1.49) 0.20 (0.03 to 1.40) 0.20 (0.03 to 1.44) — — —
YN 0.83 (0.51 to 1.35) 0.86 (0.52 to 1.40) 0.82 (0.50 to 1.35) 0.80 (0.45 to 1.43) 0.84 (0.47 to 1.52) 0.79 (0.44 to 1.44)
YY 1.40 (0.84 to 2.32) 1.39 (0.84 to 2.31) 1.32 (0.78 to 2.22) 1.61 (0.96 to 2.70) 1.52 (0.87 to 2.65) 1.41 (0.80 to 2.50)
(continued on following page)

810 © 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

TABLE 3. Adjusted Models for Associations Between Use of Dietary Supplements Other Than Antioxidants Before and During Chemotherapy and DFS and
OS in S0221: DELCaP Study Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134) (continued)
DFS, HR (95% CI) OS, HR (95% CI)
Reported Supplement Use
Before and During Fully Adjusted With Fully Adjusted With
Chemotherapy Minimally Adjusted* Fully Adjusted† MV Use‡ Minimally Adjusted* Fully Adjusted† MV Use‡
Melatonin
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 0.69 (0.16 to 2.92) 0.68 (0.17 to 2.76) 0.70 (0.17 to 2.83) 1.05 (0.25 to 4.50) 1.12 (0.27 to 4.57) 1.16 (0.28 to 4.75)
YN 0.69 (0.21 to 2.24) 0.79 (0.25 to 2.50) 0.78 (0.25 to 2.47) 0.97 (0.30 to 3.08) 1.01 (0.31 to 3.24) 1.00 (0.31 to 3.21)
YY 1.51 (0.47 to 4.92) 0.98 (0.31 to 3.18) 0.98 (0.31 to 3.16) 1.38 (0.36 to 5.36) 0.87 (0.21 to 3.65) 0.85 (0.20 to 3.54)
Acidophilus
NN 1.0 1.0 1.0 1.0 1.0 1.0
NY 1.36 (0.63 to 2.96) 1.29 (0.60 to 2.78) 1.27 (0.59 to 2.74) 1.73 (0.74 to 4.05) 1.70 (0.74 to 3.91) 1.67 (0.73 to 3.84)
YN 1.34 (0.57 to 3.10) 1.34 (0.59 to 3.06) 1.33 (0.58 to 3.02) 1.19 (0.42 to 3.37) 1.15 (0.42 to 3.17) 1.11 (0.40 to 3.04)
YY 1.90 (0.76 to 4.71) 1.43 (0.58 to 3.51) 1.44 (0.58 to 3.55) 1.93 (0.74 to 5.02) 1.52 (0.55 to 4.20) 1.54 (0.56 to 4.24)

NOTE. Use of vitamin C, vitamin A, vitamin E, carotenoids, and coenzyme Q10.


Abbreviations: DELCaP, Diet, Exercise, Lifestyle and Cancer Prognosis; DFS, disease-free survival; HR, hazard ratio; MV, multivitamin; NN, neither before
nor during; NY, not before but during; OS, overall survival; YN, before but not during; YY, both before and during.
*Stratified on treatment assignment and adjusted for age.
†Stratified on treatment assignment and adjusted for age, tumor characteristics (estrogen receptor, progesterone receptor, human epidermal growth factor
receptor 2, lymph node status, and tumor size), grades 3 or 4 hematologic toxicity, body mass index, physical activity, smoking, and alcohol consumption.
‡Stratified on treatment assignment and adjusted for age, tumor characteristics (estrogen receptor, progesterone receptor, human epidermal growth factor
receptor 2, lymph node status and tumor size), grades 3 or 4 hematologic toxicity, body mass index, physical activity, smoking, alcohol consumption, and
multivitamin use.

use of omega-3 fatty acid supplements, iron, and vitamin proliferation. Iron may also impair antitumor immunity.
B12 both before and during chemotherapy was associated Thus, iron supplementation could be associated with DFS
with DFS and OS. In DELCaP, omega-3 fatty acid intake was through these mechanisms.
derived from the question, “Did you take fish oil, EPA
How the use of vitamin B12 both before and during che-
[eicosapentaenoic acid], omega-3, flaxseed, or cod liver
motherapy could be associated with poorer outcomes remains
oil?” Thus, it is unclear what supplement was most com-
to be understood. Studies that examined circulating levels of
monly used and how that may drive associations with in-
vitamin B12 (cobalamin) in relation to cancer and cancer
creased risk. In CALGB89803, omega-3 polyunsaturated
outcomes had mixed results.23 In a cohort study of . 25,000
fatty acid was associated with a decreased risk of re-
patients with measured cobalamin levels before diagnosis,
currence, but levels were derived from fish intake18 and not
elevated levels were associated with higher mortality,23 but it is
from supplements.
unknown whether the higher levels were indicators of un-
derlying pathologic conditions. Earlier randomized controlled
Increased risk with vitamin B12 and iron use both before
trials of B vitamins or placebo in patients with ischemic heart
and during chemotherapy, as well as any antioxidant, infers
disease found higher cancer incidence and all-cause mortality
that habitual use over time might be associated with greater
in patients who were randomly assigned to folic acid plus
recurrence and mortality. Habitual use could be due to
vitamin B12 but not with vitamin B6.24
indication, with patients using these supplements to treat
an existing condition such as anemia, which could, of itself, Unlike the majority of observational studies that evaluated
be related to breast cancer recurrence and death.19 supplements and cancer outcomes,8 the strength of
However, the addition of hematologic toxicities to models DELCaP is that it was conducted in the context of a ther-
did not diminish observed associations. On the other hand, apeutic clinical trial, with surveys before beginning che-
relationships between iron and poorer outcomes were also motherapy and at completion of treatment. This allowed us
observed for those who used iron supplements only during to directly assess the potential interaction of dietary sup-
treatment as well as during both time periods. Iron sup- plements with chemotherapy. Furthermore, all patients in
plementation may independently play a role in poorer DFS the trial received the same drugs, a common chemotherapy
and OS because iron plays unique roles in tumor initiation regimen, but with varying dosing schedules. This relative
and progression directly and through effects on the tumor homogeneity in treatments allowed for more direct in-
microenvironment.20-22 Iron enables the production of ROS, ferences about supplement use in relation to treatment
which can contribute to malignant transformation, and outcomes, unlike population-based studies where treat-
once established, tumors require high amounts of iron for ments may be extremely heterogeneous.

Journal of Clinical Oncology 811

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

A B
1.0 1.0 Log-rank P = .0023
Log-rank P = .0196
DFS (probability)

OS (probability)
0.8 0.8

0.6 0.6

0.4 0.4
NN NN
0.2 NY 0.2 NY
YN YN
0.2 YY 0.2 YY

0 24 48 72 96 120 144 0 24 48 72 96 120 144


Time (months) Time (months)
No. at risk: No. at risk:
NN 918 897 856 815 778 724 571 497 403 296 141 29 0 NN 918 914 891 866 835 776 618 535 438 323 153 33 0
NY 78 74 72 70 68 63 54 41 33 21 8 1 0 NY 78 78 75 72 70 66 58 46 38 24 11 2 0
YN 86 83 83 79 75 65 48 40 28 16 7 0 YN 86 86 85 81 80 72 54 46 31 18 7 0
YY 60 59 50 47 44 39 30 24 20 12 5 1 0 YY 60 60 56 53 48 46 33 28 23 14 6 1 0

C D
1.0 1.0 Log-rank P = .0044
Log-rank P = .0012
DFS (probability)

OS (probability)
0.8 0.8

0.6 0.6

0.4 NN
0.4 NN
0.2 NY 0.2 NY
YN YN
0.2 YY 0.2 YY

0 24 48 72 96 120 144 0 24 48 72 96 120 144


Time (months) Time (months)
No. at risk: No. at risk:
NN 979 957 912 867 831 766 603 520 418 305 138 25 0 NN 979 976 950 917 886 822 650 561 454 331 150 29 0
NY 88 84 80 76 69 64 49 39 31 20 9 2 0 NY 88 88 86 84 79 71 57 47 38 25 11 2 0
YN 53 52 52 52 51 49 41 34 27 17 12 3 0 YN 53 53 52 52 51 50 43 37 29 19 13 4 0
YY 22 21 18 17 15 14 11 10 8 3 2 1 0 YY 22 21 19 19 17 17 14 11 9 4 3 1 0

FIG 3. Product limit estimates for use of vitamin B12 supplements and (A) disease-free survival (DFS) and (B) overall survival (OS) and for use of iron
supplements and (C) DFS and (D) OS. NN, neither before nor during; NY, not before but during; YN, before but not during; YY, both before and during.

Despite the strengths of the study design, we were still were perhaps more compliant because of their enrollment
hampered by the limits of the data, and as in many ob- in a clinical trial for high-risk breast cancer. Furthermore,
servational studies, there was the potential for a number of although we collected detailed data about supplement use,
biases, including selection, recall, and confounding biases. the relatively low numbers of users precluded our evalu-
Although there are reports in the literature of up to 60% of ation of higher dosages of antioxidants. However, the
patients taking antioxidant supplements during adjuvant majority of users also took a multivitamin, so it might be
treatment,1,8,10 this was not the case in S0221. The fairly assumed that use of additional single supplements could
small numbers of patients taking antioxidants and other represent somewhat higher doses than that usually found
supplements thus reduced our statistical power to be able in a multivitamin, and we further controlled for multivitamin
to identify moderate or weak associations or potentially use in fully adjusted models.
resulted in spurious associations. Although it is possible
For the small proportion of patients who did not return
that patients under-reported use, we took every effort to
questionnaires within the desired time frame, responses
obtain accurate information by asking patients to check
could be affected by recall bias. This may be relevant for Q1
supplement bottles for details. Nonetheless, under-
with regard to supplement use in the past, but we expect
reporting may have been a possibility, particularly for the
that late responses about use during treatment may be less
small number of patients for whom the questionnaire was
subject to recall bias because of the relative recency of
administered by the clinical research associate. Use may
treatment.
also have been low because patients were adhering to
recommendations from their clinicians not to take sup- Patients who took supplements during treatment could
plements during treatment, as previously reported,8 and have differed from those who did not, which would result in

812 © 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

uncontrolled confounding. Indeed, patients who took any trial of a-tocopherol and b-carotene during radiation
antioxidant, as well as vitamin B12 and iron, tended to be treatment of patients with head and neck cancer, the
older than those who did not. However, adjustment for age, intervention was associated with higher mortality.25 It is
additional lifestyle factors, and tumor characteristics only possible that relationships with the use of antioxidants
somewhat attenuated risk estimates and widened the CIs. during chemotherapy could be primarily among patients
An additional concern could be that patients may be taking who also received adjuvant radiation subsequent to this
a number of supplements, which could be the true drivers trial. Finally, although a landmark analysis approach
of associations with antioxidants. Adjustment for use of appropriately separates predictor (supplement use) from
multivitamins did not greatly affect associations, however, survival outcomes, it does not rule out the possibility of
and in sensitivity analyses where the small number of selection bias between groups.
patients taking more than five supplements were removed In summary, our findings from this prospective study in
(n = 129), associations remained the same. the context of a cooperative group clinical trial indicate
The constraints of the intergroup trial design also limited that use of antioxidant supplements during chemother-
availability of data. A possibility exists that patients who apy, as well as iron and vitamin B12, may increase the
chose to take supplements were less likely to adhere to risk of breast cancer recurrence and mortality. Short of
adjuvant hormonal therapy, which could be associated a randomized trial of supplements in patients with cancer,
with poorer survival outcomes, but information on ad- the findings provide some empirical data for consideration
herence was not available for this trial. We also lack data when discussing with patients the use of dietary sup-
on possible adjuvant radiation therapy. In a randomized plements during chemotherapy.

AFFILIATIONS CA180863, CA180858, CA180828, CA180801, CA68183, CA04919,


1
Roswell Park Comprehensive Cancer Center, Buffalo, NY CA13612, CA46282; and, in part, by Amgen.
2
Boston University, Boston, MA
3
Fred Hutchinson Cancer Research Center, Seattle, WA AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF
4
The Ohio State University, Columbus, OH INTEREST AND DATA AVAILABILITY STATEMENT
5
Columbia University, New York, NY
6
Disclosures provided by the authors and data availability statement (if
Cleveland Clinic, Cleveland, OH
7
applicable) are available with this article at DOI https://doi.org/10.1200/
Baystate Medical Center, Springfield, MA
8
JCO.19.01203.
Georgetown University, Washington, DC
9
Mayo Clinic, Rochester, MN
10
Allan Blair Cancer Centre, Regina, Saskatchewan, Canada AUTHOR CONTRIBUTIONS
11
The University of Texas MD Anderson Cancer Center, Houston, TX Conception and design: Christine B. Ambrosone, Gary R. Zirpoli, Susan E.
12
Seattle Cancer Care Alliance, Seattle, WA McCann, Dawn L. Hershman, Joseph M. Unger, James A. Stewart, George
13
Loyola University Chicago Stritch School of Medicine, Chicago, IL T. Budd, Kathy S. Albain
Financial support: Christine B. Ambrosone
Administrative support: Christine B. Ambrosone, Gabriel N. Hortobagyi
CORRESPONDING AUTHOR
Provision of study material or patients: Christine B. Ambrosone, Halle C.F.
Christine B. Ambrosone, PhD, Roswell Park Comprehensive Cancer
Moore, James A. Stewart, Gabriel N. Hortobagyi, George T. Budd, Kathy
Center, Elm and Carlton Sts, Buffalo, NY 14263; e-mail:
S. Albain
christine.ambrosone@roswellpark.org.
Collection and assembly of data: Christine B. Ambrosone, Gary R. Zirpoli,
William E. McCann, William E. Barlow, Timothy J. Hobday, Muhammad
EQUAL CONTRIBUTION Salim, Gabriel N. Hortobagyi, George T. Budd
G.T.B. and K.S.A. are joint senior authors. Data analysis and interpretation: Christine B. Ambrosone, Gary R. Zirpoli,
Alan D. Hutson, William E. McCann, Susan E. McCann, William E.
Barlow, Kara M. Kelly, Rikki Cannioto, Lara E. Sucheston-Campbell,
SUPPORT
Dawn L. Hershman, Joseph M. Unger, Halle C.F. Moore, Claudine Isaacs,
Supported by grants R01 CA116395 (C.B.A.) and R01 CA139426
Timothy J. Hobday, Gabriel N. Hortobagyi, Julie R. Gralow, George T.
(C.B.A.), the Breast Cancer Research Foundation (C.B.A.), and Roswell
Budd, Kathy S. Albain
Park Comprehensive Cancer Center Cancer Center Support Grant P30
Manuscript writing: All authors
CA016056. S0221 was supported, in part, by National Cancer Institute
Final approval of manuscript: All authors
(NCI), Division of Cancer Prevention, SWOG NCI Community Oncology
Accountable for all aspects of the work: All authors
Research Program Research Base Grant No. 5UG1 CA189974-02; by
NCI, National Clinical Trials Network, Grant No. CA180888, CA180819,

REFERENCES
1. Velicer CM, Ulrich CM: Vitamin and mineral supplement use among US adults after cancer diagnosis: A systematic review. J Clin Oncol 26:665-673, 2008
2. Cassileth BR, Vickers AJ: High prevalence of complementary and alternative medicine use among cancer patients: Implications for research and clinical care.
J Clin Oncol 23:2590-2592, 2005
3. Harvie M: Nutritional supplements and cancer: Potential benefits and proven harms. Am Soc Clin Oncol Educ Book 34:e478-e486, 2014

Journal of Clinical Oncology 813

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

4. Lawenda BD, Kelly KM, Ladas EJ, et al: Should supplemental antioxidant administration be avoided during chemotherapy and radiation therapy? J Natl Cancer
Inst 100:773-783, 2008
5. Norman HA, Butrum RR, Feldman E, et al: The role of dietary supplements during cancer therapy. J Nutr 133:3794S-3799S, 2003
6. Rock CL, Doyle C, Demark-Wahnefried W, et al: Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin 62:242-274, 2012
7. World Cancer Research Fund and American Institute for Cancer Research: Cancer survivors: Evidence on survivors of breast and other cancers. https://
www.wcrf.org/dietandcancer/cancer-survivors
8. Zirpoli GR, Brennan PM, Hong CC, et al: Supplement use during an intergroup clinical trial for breast cancer (S0221). Breast Cancer Res Treat 137:903-913,
2013
9. Budd GT, Barlow WE, Moore HC, et al: SWOG S0221: A phase III trial comparing chemotherapy schedules in high-risk early-stage breast cancer. J Clin Oncol
33:58-64, 2015
10. Zirpoli GR, McCann SE, Sucheston-Campbell LE, et al: Supplement use and chemotherapy-induced peripheral neuropathy in a cooperative group trial (S0221):
The DELCaP study. J Natl Cancer Inst 109:djx098, 2017
11. Satia-Abouta J, Patterson RE, King IB, et al: Reliability and validity of self-report of vitamin and mineral supplement use in the vitamins and lifestyle study. Am
J Epidemiol 157:944-954, 2003
12. Luc L, Baumgart C, Weiss E, et al: Dietary supplement use among participants of a databank and biorepository at a comprehensive cancer centre. Public Health
Nutr 18:916-926, 2015
13. Anderson JR, Cain KC, Gelber RD: Analysis of survival by tumor response. J Clin Oncol 1:710-719, 1983
14. Ambrosone C, Ahn J, Schoenenberger V: Antioxidant supplements, genetics and chemotherapy outcomes. Curr Cancer Ther Rev 1:251-258, 2005
15. American Institute for Cancer Research: Nutrition of the Cancer Patient. Washington, DC, American Institute for Cancer Research, 2000. https://www.aicr.org/
patients-survivors/healthy-or-harmful/supplements.html
16. Ladas E, Kelly KM: The antioxidant debate. Explore (NY) 6:75-85, 2010
17. Ng K, Meyerhardt JA, Chan JA, et al: Multivitamin use is not associated with cancer recurrence or survival in patients with stage III colon cancer: Findings from
CALGB 89803. J Clin Oncol 28:4354-4363, 2010
18. Van Blarigan EL, Fuchs CS, Niedzwiecki D, et al: Marine v-3 Polyunsaturated fatty acid and fish intake after colon cancer diagnosis and survival: CALGB 89803
(Alliance). Cancer Epidemiol Biomarkers Prev 27:438-445, 2018
19. Caro JJ, Salas M, Ward A, et al: Anemia as an independent prognostic factor for survival in patients with cancer: A systemic, quantitative review. Cancer 91:
2214-2221, 2001
20. Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, et al: Iron in the tumor microenvironment-connecting the dots. Front Oncol 8:549, 2018
21. Ludwig H, Evstatiev R, Kornek G, et al: Iron metabolism and iron supplementation in cancer patients. Wien Klin Wochenschr 127:907-919, 2015 [Erratum:
Wien Klin Wochenschr 127:920-921, 2015]
22. Torti SV, Torti FM: Iron and cancer: More ore to be mined. Nat Rev Cancer 13:342-355, 2013
23. Arendt JF, Farkas DK, Pedersen L, et al: Elevated plasma vitamin B12 levels and cancer prognosis: A population-based cohort study. Cancer Epidemiol 40:
158-165, 2016
24. Ebbing M, Bønaa KH, Nygård O, et al: Cancer incidence and mortality after treatment with folic acid and vitamin B12. JAMA 302:2119-2126, 2009
25. Bairati I, Meyer F, Jobin E, et al: Antioxidant vitamins supplementation and mortality: A randomized trial in head and neck cancer patients. Int J Cancer 119:
2221-2224, 2006

n n n

814 © 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST


Dietary Supplement Use During Chemotherapy and Survival Outcomes of Patients With Breast Cancer Enrolled in a Cooperative Group Clinical Trial (SWOG
S0221)
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted.
Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript.
For more information about ASCO’s conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/journal/jco/site/ifc.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

William E. Barlow Gabriel N. Hortobagyi


Research Funding: AbbVie (Inst), Merck (Inst), AstraZeneca (Inst) Consulting or Advisory Role: Novartis, Peregrine Pharmaceuticals, Agendia
Research Funding: Novartis (Inst)
Kara M. Kelly
Travel, Accommodations, Expenses: Novartis
Research Funding: Merck (Inst)
Travel, Accommodations, Expenses: Bristol-Myers Squibb Julie R. Gralow
Consulting or Advisory Role: Novartis, Genentech, Pfizer, Merck, Puma
Dawn L. Hershman
Biotechnology, Sandoz, AstraZeneca, Immunomedics, Genomic Health
Consulting or Advisory Role: AIM Specialty Health
George T. Budd
Halle C.F. Moore
Research Funding: Genentech (Inst), Roche (Inst), TRACON Pharma (Inst),
Research Funding: Puma Biotechnology (Inst), AbbVie (Inst)
Daiichi Sankyo (Inst), Eli Lilly (Inst), Macrogenics (Inst), Ambrx (Inst), Deciphera
Claudine Isaacs (Inst)
Honoraria: Genentech, Roche, AstraZeneca, Pfizer
Kathy S. Albain
Consulting or Advisory Role: Pfizer, Genentech, Roche, Novartis, AstraZeneca,
Consulting or Advisory Role: Novartis, Pfizer, Myriad Genetics, Genomic Health,
Medivation, NanoString Technologies, Syndax, Puma Biotechnology, Context
Agendia, Genentech, Roche
Therapeutics
Research Funding: Seattle Genetics (Inst)
Speakers’ Bureau: Genentech, Pfizer, AstraZeneca
Other Relationship: Puma Biotechnology
Research Funding: Novartis (Inst), Pfizer (Inst), Genentech (Inst), Tesaro (Inst)
Patents, Royalties, Other Intellectual Property: McGraw Hill Publishing, No other potential conflicts of interest were reported.
UpToDate, Wolters Kluwer, Elsevier
Timothy J. Hobday
Consulting or Advisory Role: AbbVie
Research Funding: Novartis (Inst)

Journal of Clinical Oncology

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

APPENDIX

TABLE A1. Supplement Use Before and During Breast Cancer TABLE A1. Supplement Use Before and During Breast Cancer
Treatment Among Patients Who Completed Questionnaires 1 and 2 in Treatment Among Patients Who Completed Questionnaires 1 and 2 in
the DELCaP Study (n = 1,134) the DELCaP Study (n = 1,134) (continued)
Supplement Use During Supplement Use During
Treatment, No. (%) Treatment, No. (%)
Supplement Use Supplement Use
Before Diagnosis No Yes Total, No. (%) Before Diagnosis No Yes Total, No. (%)
Vitamin C Yes 86 (7.6) 60 (5.3) 146 (12.9)
No 844 (74.4) 57 (5.0) 901 (79.5) Total 997 (87.9) 137 (12.1) 1,134 (100)
Yes 152 (13.4) 81 (7.1) 233 (20.5) Iron
Total 996 (87.8) 138 (12.2) 1,134 (100) No 973 (85.8) 86 (7.6) 1,059 (93.4)
Vitamin A Yes 53 (4.7) 22 (1.9) 75 (6.6)
No 1,077 (94.9) 21 (1.8) 1,098 (96.8) Total 1,028 (90.1) 109 (9.6) 1,134 (100)
Yes 31 (2.7) 5 (0.1) 36 (3.2) Folic acid
Total 1,108 (97.7) 26 (2.3) 1,134 (100) No 962 (84.8) 58 (5.1) 1,020 (90.0)
Vitamin E Yes 80 (7.1) 34 (2.9) 114 (10.0)
No 911 (80.3) 41 (3.6) 952 (84.0) Total 1,042 (91.9) 92 (8.1) 1,134 (100)
Yes 147 (12.9) 35 (3.1) 182 (16.0) Calcium
Total 1,058 (93.3) 76 (6.7) 1,134 (100) No 634 (55.9) 110 (9.7) 744 (65.6)
Coenzyme Q10 Yes 181 (15.9) 209 (18.4) 390 (34.4)
No 1,057 (93.4) 26 (2.3) 1,083 (95.7) Total 815 (71.9) 319 (28.1) 1,134 (100)
Yes 33 (2.9) 15 (1.3) 48 (4.3) Omega-3 fatty acids
Total 1,090 (96.4) 41 (3.6) 1,131 (100) No 831 (73.3) 46 (4.1) 877 (77.3)
Carotenoid Yes 160 (14.1) 97 (8.6) 257 (22.7)
No 1,106 (97.5) 6 (0.6) 1,112 (98.1) Total 991 (87.4) 143 (12.6) 1,134 (100)
Yes 17 (1.5) 5 (0.5) 22 (1.9) Glucosamine
Total 1,123 (99.0) 11 (1.0) 1,134 (100) No 967 (85.3) 18 (1.6) 985 (87.0)
Any antioxidant* Yes 88 (7.8) 60 (5.3) 148 (13.0)
No 736 (65.0) 74 (6.5) 810 (71.6) Total 1,055 (93.1) 78 (6.9) 1,133 (100)
Yes 206 (18.2) 126 (11.1) 332 (29.3) Melatonin
Total 932 (82.3) 200 (17.6) 1,132 (100) No 1,089 (96.3) 14 (1.2) 1,108 (97.4)
Multivitamins Yes 19 (1.7) 10 (0.9) 29 (2.6)
No 459 (40.4) 127 (11.2) 588 (51.6) Total 1,108 (97.9) 24 (2.1) 1,132 (100)
Yes 178 (15.7) 370 (32.6) 551 (48.4) Acidophilus
Total 637 (56.1) 497 (43.8) 1,134 (100) No 1,067 (94.4) 27 (2.4) 1,094 (96.8)
Vitamin D Yes 23 (2.0) 13 (1.2) 36 (3.2)
No 736 (64.9) 140 (12.3) 876 (77.2) Total 1,090 (96.4) 40 (3.5) 1,130 (100)
Yes 119 (10.5) 139 (12.3) 258 (22.7)
NOTE. Total numbers for each supplement may vary because of
Total 855 (75.4) 279 (24.6) 1,134 (100) missing data.
Vitamin B6 Abbreviation: DELCaP, Diet, Exercise, Lifestyle and Cancer
Prognosis.
No 846 (74.5) 178 (15.8) 1,022 (90.3)
*Use of vitamin C, vitamin A, vitamin E, carotenoids, and coenzyme
Yes 62 (5.5) 48 (4.2) 110 (9.7) Q10.
Total 908 (80.0) 226 (20.0) 1,134 (100)
Vitamin B12
No 911 (80.3) 77 (6.8) 988 (87.1)
(continued in next column)

© 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

TABLE A2. Prevalence of Use of Dietary Supplements Before and During Chemotherapy for High-Risk Breast Cancer in S0221: DELCaP Study
Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134)
Progression, No. (%) Death, No. (%)
Reported Supplement Use Before
and During Chemotherapy No Yes No Yes

Any antioxidant*
NN 580 (79) 156 (21) 629 (85) 110 (15)
NY 62 (84) 12 (16) 65 (87) 10 (13)
YN 160 (78) 46 (22) 172 (83) 35 (17)
YY 89 (71) 37 (29) 100 (79) 26 (21)
Vitamin C
NN 663 (79) 181 (21) 721 (85) 127 (15)
NY 45 (79) 12 (21) 49 (84) 9 (16)
YN 117 (77) 35 (23) 124 (82) 28 (18)
YY 58 (72) 23 (28) 64 (79) 17 (21)
Vitamin A
NN 844 (78) 234 (22) 914 (84) 169 (16)
NY 14 (67) 7 (33) 17 (81) 4 (19)
YN 25 (81) 6 (19) 27 (87) 4 (13)
YY 2 (40) 3 (60) 2 (40) 3 (60)
Vitamin E
NN 715 (78) 196 (22) 776 (85) 140 (15)
NY 31 (76) 10 (24) 34 (83) 7 (17)
YN 114 (78) 33 (22) 122 (83) 25 (17)
YY 23 (66) 12 (34) 26 (74) 9 (26)
Coenzyme Q10
NN 829 (78) 228 (21) 897 (85) 164 (15)
NY 20 (77) 6 (23) 22 (85) 4 (15)
YN 24 (73) 9 (27) 28 (82) 6 (18)
YY 9 (60) 6 (40) 10 (67) 5 (33)
Carotenoid
NN 868 (78) 240 (22) 938 (84) 174 (16)
NY 3 (50) 3 (50) 4 (57) 3 (43)
YN 12 (70) 5 (29) 15 (88) 2 (12)
YY 2 (40) 3 (60) 3 (60) 2 (40)
Multivitamins
NN 365 (80) 94 (20) 394 (86) 66 (14)
NY 99 (78) 28 (22) 110 (86) 18 (14)
YN 135 (76) 43 (24) 147 (83) 31 (17)
YY 284 (77) 86 (23) 306 (82) 67 (18)
Vitamin D
NN 582 (79) 155 (21) 628 (85) 113 (15)
NY 105 (75) 35 (25) 116 (83) 24 (17)
YN 93 (78) 26 (22) 99 (83) 20 (17)
YY 105 (76) 34 (24) 116 (83) 24 (17)
Calcium
NN 513 (58) 123 (19) 554 (87) 86 (13)
NY 84 (76) 27 (24) 94 (85) 17 (15)
YN 133 (73) 48 (27) 141 (78) 40 (22)
YY 157 (75) 52 (25) 172 (82) 38 (18)
Vitamin B6
(continued on following page)

Journal of Clinical Oncology

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

TABLE A2. Prevalence of Use of Dietary Supplements Before and During Chemotherapy for High-Risk Breast Cancer in S0221: DELCaP Study
Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134) (continued)
Progression, No. (%) Death, No. (%)
Reported Supplement Use Before
and During Chemotherapy No Yes No Yes

NN 656 (78) 190 (22) 715 (84) 134 (16)


NY 141 (79) 37 (21) 152 (84) 28 (16)
YN 51 (82) 11 (18) 53 (85) 9 (15)
YY 36 (75) 12 (25) 39 (81) 9 (19)
Vitamin B12
NN 718 (79) 195 (21) 776 (85) 142 (15)
NY 60 (77) 18 (23) 68 (87) 10 (13)
YN 70 (81) 16 (19) 76 (88) 10 (12)
YY 38 (63) 22 (37) 41 (68) 19 (32)
Iron
NN 771 (79) 204 (21) 831 (85) 148 (15)
NY 60 (70) 27 (31) 68 (77) 20 (23)
YN 45 (85) 8 (15) 48 (91) 5 (9)
YY 12 (55) 10 (45) 14 (63) 8 (36)
Folic acid
NN 754 (78) 210 (22) 816 (84) 152 (16)
NY 43 (73) 16 (27) 49 (83) 10 (17)
YN 65 (81) 15 (19) 71 (88) 10 (12)
YY 24 (71) 10 (29) 25 (74) 9 (26)
Omega-3 fatty acids
NN 647 (78) 182 (22) 698 (84) 135 (16)
NY 34 (76) 11 (24) 38 (84) 7 (16)
YN 133 (83) 27 (17) 14 (88) 19 (12)
YY 67 (69) 30 (31) 79 (81) 19 (19)
Glucosamine
NN 753 (78) 214 (22) 820 (84) 152 (16)
NY 17 (94) 1 (6) 18 (100) 0
YN 70 (80) 18 (20) 75 (85) 13 (15)
YY 42 (70) 18 (30) 44 (73) 16 (27)
Melatonin
NN 845 (78) 244 (22) 919 (84) 175 (16)
NY 12 (86) 2 (14) 12 (86) 2 (14)
YN 16 (84) 3 (16) 16 (84) 3 (16)
YY 7 (70) 3 (30) 8 (80) 2 (20)
Acidophilus
NN 837 (78) 230 (22) 906 (78) 166 (15)
NY 20 (74) 7 (26) 21 (74) 6 (22)
YN 17 (74) 6 (26) 19 (83) 4 (17)
YY 8 (62) 5 (38) 9 (69) 4 (31)

NOTE. Total numbers for each supplement may vary because of missing data.
Abbreviations: DELCaP, Diet, Exercise, Lifestyle and Cancer Prognosis; NN, neither before nor during; NY, not before but during; YN, before
but not during; YY, both before and during.
*Use of vitamin C, vitamin A, vitamin E, carotenoids, or coenzyme Q10.

© 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Supplement Use During Chemotherapy and Breast Cancer Survival

TABLE A3. Associations Between Supplements Before and During Chemotherapy for High-Risk Breast Cancer and DFS and OS in S0221
Stratified by ER Status: DELCaP Study Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134)
Progression,
No. (%) Death, No. (%)

Reported Supplement Use No Yes DFS, HR (95% CI) No Yes OS, HR (95% CI)
Antioxidants
Vitamin C
YY 58 (72) 23 (28) 1.37 (0.89 to 2.11) 64 (79) 17 (21) 1.41 (0.86 to 2.34)
ER positive 38 (76) 12 (24) 1.28 (0.70 to 2.34) 41 (82) 9 (18) 1.55 (0.78 to 3.10)
ER negative 20 (67) 10 (33) 1.25 (0.66 to 2.37) 23 (77) 7 (23) 1.04 (0.48 to 2.24)
Vitamin A
YY 2 (40) 3 (60) 4.05 (1.46 to 11.25) 2 (40) 3 (60) 4.14 (1.81 to 9.50)
ER positive 2 (40) 3 (60) 5.34 (1.76 to 16.17) 2 (67) 3 (33) 5.21 (2.20 to 12.34)
ER negative 0 (0) 0 (0) — 0 (0) 0 (0) —
Vitamin E
YY 23 (66) 12 (34) 1.62 (0.92 to 2.88) 26 (74) 9 (26) 1.65 (0.86 to 3.17)
ER positive 13 (62) 8 (38) 1.98 (0.96 to 4.08) 16 (76) 5 (24) 1.76 (0.74 to 4.15)
ER negative 10 (71) 4 (29) 1.05 (0.39 to 2.84) 10 (71) 4 (29) 1.36 (0.50 to 3.74)
Coenzyme Q10
YY 9 (60) 6 (40) 2.02 (0.92 to 4.47) 10 (67) 5 (33) 2.20 (0.92 to 5.26)
ER positive 6 (67) 3 (33) 1.76 (0.59 to 5.25) 7 (78) 2 (22) 1.60 (0.41 to 6.27)
ER negative 3 (50) 3 (50) 2.22 (0.71 to 6.97) 3 (50) 3 (50) 2.79 (0.90 to 8.64)
Carotenoid
YY 2 (40) 3 (60) 4.03 (1.12 to 14.50) 3 (60) 2 (40) 2.68 (0.87 to 8.27)
ER positive 0 (0) 0 (0) — 0 (0) 0 (0) —
ER negative 2 (40) 3 (60) 3.15 (1.07 to 9.32) 3 (60 2 (40) 1.88 (1.64 to 5.55)
Any antioxidant*
YY 89 (71) 37 (29) 1.42 (1.00 to 2.03) 100 (79) 26 (21) 1.37 (0.90 to 2.08)
ER positive 61 (73) 22 (27) 1.42 (0.89 to 2.24) 67 (81) 16 (19) 1.70 (0.99 to 2.92)
ER negative 28 (67) 14 (33) 1.32 (0.74 to 2.36) 33 (79) 9 (21) 0.93 (0.46 to 1.88)
Nonantioxidants
Multivitamins
YY 284 (77) 86 (23) 1.11 (0.83 to 1.48) 306 (82) 67 (18) 1.20 (0.85 to 1.68)
ER positive 204 (79) 55 (21) 1.17 (0.81 to 1.69) 218 (84) 41 (16) 1.40 (0.89 to 2.19)
ER negative 79 (71) 33 (29) 1.06 (0.65 to 1.73) 87 (78) 25 (22) 1.01 (0.59 to 1.73)
Vitamin D
YY 105 (76) 34 (24) 1.17 (0.81 to 1.70) 116 (83) 24 (17) 1.09 (0.71 to 1.70)
ER positive 71 (76) 22 (24) 1.38 (0.87 to 2.20) 78 (84) 15 (16) 1.50 (0.85 to 2.63)
ER negative 34 (74) 12 (26) 0.83 (0.44 to 1.57) 38 (83) 8 (17) 0.65 (0.31 to 1.36)
Vitamin B6
YY 36 (75) 12 (25) 1.09 (0.61 to 1.97) 39 (81) 9 (19) 1.13 (0.56 to 2.24)
ER positive 21 (70) 9 (30) 1.62 (0.81 to 3.24) 24 (80) 6 (20) 1.51 (0.64 to 3.54)
ER negative 15 (83) 3 (17) 0.54 (0.17 to 1.72) 15 (83) 3 (17) 0.75 (0.23 to 2.44)
Vitamin B12
YY 38 (63) 22 (37) 1.87 (1.20 to 2.98) 41 (68) 19 (32) 2.07 (1.37 to 3.38)
ER positive 25 (64) 14 (36) 2.15 (1.21 to 3.82) 27 (69) 12 (31) 2.53 (1.35 to 4.73)
ER negative 13 (65) 7 (35) 1.32 (0.58 to 2.99) 14 (70) 6 (30) 1.31 (0.60 to 3.04)
(continued on following page)

Journal of Clinical Oncology

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ambrosone et al

TABLE A3. Associations Between Supplements Before and During Chemotherapy for High-Risk Breast Cancer and DFS and OS in S0221
Stratified by ER Status: DELCaP Study Participants Who Completed Both Questionnaires 1 and 2 (n = 1,134) (continued)
Progression,
No. (%) Death, No. (%)

Reported Supplement Use No Yes DFS, HR (95% CI) No Yes OS, HR (95% CI)
Iron
YY 12 (55) 10 (45) 2.54 (1.36 to 4.78) 14 (63) 8 (36) 2.67 (1.36 to 5.27)
ER positive 8 (50) 8 (50) 3.28 (1.60 to 6.70) 10 (63) 6 (37) 3.44 (1.62 to 7.32)
ER negative 4 (67) 2 (33) 1.52 (0.31 to 7.42) 4 (67) 2 (33) 1.90 (0.39 to 9.36)
Folic acid
YY 24 (71) 10 (29) 1.32 (0.69 to 2.52) 25 (74) 9 (26) 1.54 (0.78 to 3.086)
ER positive 17 (68) 8 (32) 1.77 (0.82 to 3.79) 18 (72) 7 (28) 2.13 (0.95 to 4.79)
ER negative 7 (78) 2 (22) 0.70 (0.18 to 2.70) 7 (78) 2 (22) 0.92 (0.23 to 3.79)
Calcium
YY 157 (75) 52 (25) 1.22 (0.88 to 1.70) 172 (82) 38 (18) 1.54 (0.78 to 3.06)
ER positive 108 (77) 32 (23) 1.12 (0.74 to 1.70) 117 (84) 23 (16) 1.30 (0.79 to 2.14)
ER negative 48 (71) 20 (29) 1.37 (0.78 to 2.41) 54 (79) 14 (21) 1.03 (0.55 to 1.93)
Omega-3 fatty acids
YY 67 (69) 30 (31) 1.54 (1.03 to 2.29) 79 (81) 19 (19) 1.23 (0.75 to 2.04)
ER positive 43 (73) 16 (27) 1.36 (0.82 to 2.35) 51 (86) 8 (14) 0.92 (0.44 to 1.96)
ER negative 24 (60) 14 (40) 1.60 (0.86 to 2.98) 28 (74) 10 (26) 1.37 (0.69 to 2.73)
Glucosamine
YY 42 (70) 18 (30) 1.40 (0.84 to 2.32) 44 (73) 16 (27) 1.61 (0.96 to 2.70)
ER positive 30 (70) 13 (30) 1.69 (0.91 to 3.14) 31 (72) 12 (28) 2.19 (1.16 to 4.10)
ER negative 12 (75) 4 (25) 0.79 (0.30 to 2.11) 13 (81) 3 (19) 0.70 (0.23 to 2.12)
Melatonin
YY 7 (70) 3 (30) 1.51 (0.47 to 4.92) 8 (80) 2 (20) 1.38 (0.36 to 5.36)
ER positive 4 (67) 2 (33) 1.79 (0.42 to 7.56) 4 (67) 2 (33) 2.77 (0.66 to 11.70)
ER negative 3 (75) 1 (25) 1.10 (0.12 to 9.95) 4 (100) 0 (0) 0.0
Acidophilus
YY 8 (62) 5 (38) 1.90 (0.76 to 4.71) 8 (62) 4 (31) 1.93 (0.74 to 5.02)
ER positive 8 (80) 2 (20) 0.93 (0.22 to 4.03) 8 (80) 2 (20) 1.40 (0.35 to 5.64)
ER negative 0 (0) 3 (100) 5.81 (3.91 to 8.63) 1 (33) 2 (77) 3.66 (1.12 to 11.97)

NOTE. Total numbers for each supplement may vary because of missing data.
Abbreviations: DELCaP, Diet, Exercise, Lifestyle and Cancer Prognosis; ER, estrogen receptor; HR, hazard ratio; YY, both before and during.
*Use of vitamin C, vitamin A, vitamin E, carotenoids, or coenzyme Q10.

© 2019 by American Society of Clinical Oncology Volume 38, Issue 8

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Printed by Salvador Macias on 8/24/2022 5:23:20 AM. For personal use only. Not approved for distribution. Copyright © 2022 National Comprehensive Cancer Network, Inc., All Rights Reserved.

NCCN Guidelines Version 3.2022 NCCN Guidelines Index


Table of Contents
Non-Small Cell Lung Cancer Discussion

FINDINGS FOLLOW-UPc,d,g,h
<6 mm No routine follow-up
Consider CT
Low riske 6–8 mm CT at 6–12 mo Stable
at 18–24 mo
Consider CT at 3 mo,
>8 mm
PET/CT,i or biopsyj
Incidental
finding: solid
nodule(s) on
chest CT CT at 12 mo
<6 mm Stable No routine follow-up
(optional)
Repeat CT at
High riskf 6–8 mm CT at 6–12 mo Stable
18–24 mo

Consider CT at 3 mo,
>8 mm
PET/CT,i or biopsyj

c Principles of Diagnostic Evaluation (DIAG-A 1 of 3). i PET/CT performed skull base to knees or whole body. A positive PET result is defined
d The most important radiologic factor is change or stability compared with a previous as a standardized uptake value (SUV) in the lung nodule greater than the baseline
imaging study. mediastinal blood pool. A positive PET scan finding can be caused by infection or
e Low risk = minimal or absent history of smoking or other known risk factors. inflammation, including absence of lung cancer with localized infection, presence
f High risk = history of smoking or other known risk factors. Known risk factors include of lung cancer with associated (eg, postobstructive) infection, and presence of lung
history of lung cancer in a first-degree relative; exposure to asbestos, radon, or cancer with related inflammation (eg, nodal, parenchymal, pleural). A false-negative
uranium. PET scan can be caused by a small nodule, low cellular density (nonsolid nodule or
g Non-solid (ground-glass) nodules may require longer follow-up to exclude indolent ground-glass opacity [GGO]), or low tumor avidity for FDG (eg, adenocarcinoma in
adenocarcinoma. situ [previously known as bronchoalveolar carcinoma], carcinoid tumor).
h Adapted from Fleischner Society Guidelines: MacMahon H, Naidich DP, Goo j If empiric therapy is contemplated without tissue confirmation, multidisciplinary
JM, et al. Guidelines for management of incidental pulmonary nodules detected evaluation that at least includes interventional radiology, thoracic surgery, and
on CT images: From the Fleischner Society 2017. Radiology 2017;284:228-243. interventional pulmonology is required to determine the safest and most efficient
©
Radiological Society of North America. Fleischner Society Guidelines do not direct approach for biopsy, or to provide consensus that a biopsy is too risky or difficult and
whether or not contrast is necessary or if an LDCT is appropriate. LDCT is preferred that the patient can proceed with therapy without tissue confirmation. (IJsseldijk MA,
unless there is a reason for contrast enhancement for better diagnostic resolution. et al. J Thorac Oncol 2019;14:583-595.)

Note: All recommendations are category 2A unless otherwise indicated.


Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.

Version 3.2022, 03/16/22 © 2022 National Comprehensive Cancer Network® (NCCN®), All rights reserved. NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN.
DIAG-2
Printed by Salvador Macias on 8/24/2022 5:23:20 AM. For personal use only. Not approved for distribution. Copyright © 2022 National Comprehensive Cancer Network, Inc., All Rights Reserved.

NCCN Guidelines Version 3.2022 NCCN Guidelines Index


Table of Contents
Non-Small Cell Lung Cancer Discussion

FINDINGS FOLLOW-UPc,d,g,h
Solitary pure <6 mm No routine follow-up
ground-glass CT at 6–12 mo to confirm no growth or
nodules ≥6 mm development of a solid component, then
CT every 2 y until 5 y
Incidental
finding: Solitary <6 mm No routine follow-up
subsolid part-solid • CT at 3–6 mo to confirm no growth or change in solid
nodule(s) nodules ≥6 mm component, then annual CT for 5 y
on chest CT • If solid component ≥6 mm, consider PET/CTi or biopsyj

• CT at 3–6 mo
<6 mm
If stable, consider CT at 2 and 4 y
Multiple
subsolid
• CT at 3–6 mo
nodules
• Subsequent management
≥6 mm
based on most
suspicious nodule(s)

i PET/CT performed skull base to knees or whole body. A positive PET result is defined
as a SUV in the lung nodule greater than the baseline mediastinal blood pool. A
positive PET scan finding can be caused by infection or inflammation, including
absence of lung cancer with localized infection, presence of lung cancer with
c Principles of Diagnostic Evaluation (DIAG-A 1 of 3). associated (eg, postobstructive) infection, and presence of lung cancer with related
d The most important radiologic factor is change or stability
compared with a previous inflammation (eg, nodal, parenchymal, pleural). A false-negative PET scan can be
imaging study. caused by a small nodule, low cellular density (nonsolid nodule or GGO), or low tumor
g Non-solid (ground-glass) nodules may require longer follow-up to exclude indolent avidity for FDG (eg, adenocarcinoma in situ [previously known as bronchoalveolar
adenocarcinoma. carcinoma], carcinoid tumor).
h Adapted from Fleischner Society Guidelines: MacMahon H, Naidich DP, Goo j If empiric therapy is contemplated without tissue confirmation, multidisciplinary
JM, et al. Guidelines for management of incidental pulmonary nodules detected evaluation that at least includes interventional radiology, thoracic surgery, and
on CT images: From the Fleischner Society 2017. Radiology 2017;284:228-243. interventional pulmonology is required to determine the safest and most efficient
©
Radiological Society of North America. Fleischner Society Guidelines do not direct approach for biopsy, or to provide consensus that a biopsy is too risky or difficult and
whether or not contrast is necessary or if an LDCT is appropriate. LDCT is preferred that the patient can proceed with therapy without tissue confirmation. (IJsseldijk MA,
unless there is a reason for contrast enhancement for better diagnostic resolution. et al. J Thorac Oncol 2019;14:583-595.)

Note: All recommendations are category 2A unless otherwise indicated.


Clinical Trials: NCCN believes that the best management of any patient with cancer is in a clinical trial. Participation in clinical trials is especially encouraged.

Version 3.2022, 03/16/22 © 2022 National Comprehensive Cancer Network® (NCCN®), All rights reserved. NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN.
DIAG-3
17
CAPÍTULO 

Nódulo pulmonar solitario


E. Guzmán de Alba

El cáncer de pulmón se ha convertido en nuestro país, y En estudios grandes se ha encontrado que la probabili-
en todo el mundo, en uno de los más frecuentes y más dad de malignidad de un nódulo es muy baja, del 0 al 1.5%
mortales, motivos por los que, al igual que en cualquier otra en nódulos < 5 mm, riesgo que aumenta desde el 33 hasta
neoplasia, el diagnóstico temprano y el tratamiento ade- el 64% en nódulos de 11 a 20 mm de diámetro y que pue-
cuado y bien dirigido permiten alcanzar los mejores resul- de ser tan elevada como del 82% en nódulos > 20 mm7. La
tados y el mejor pronóstico para el paciente. El nódulo pul- historia de tabaquismo es preponderante en el riesgo de
monar solitario se define como un nódulo de 3 cm de desarrollar un cáncer de pulmón, por lo que es uno de los
diámetro como máximo, completamente rodeado de pa- factores que se debe evaluar de manera más profunda en
rénquima pulmonar aparentemente sano y que no se aso- el momento de determinar la posibilidad de que un nódu-
cia a ganglios linfáticos aumentados de tamaño ni a derra- lo pulmonar sea de origen neoplásico8. La intensidad en el
me pleural1. La detección de un nódulo pulmonar solitario consumo del tabaco (índice tabáquico) se puede medir
en general se produce en pacientes asintomáticos a quie- mediante la determinación del número de paquetes de
nes se les realizó un estudio radiológico de tórax por encon- cigarrillos fumados al año por una persona utilizando la
trarse dentro de un programa de detección temprana de siguiente fórmula:
cáncer de pulmón o por alguna otra razón, como el segui-
miento por alguna neoplasia previa, el estudio preoperato- n.º de cigarrillos
rio previo a alguna cirugía o en el contexto de alguna otra fumados al día
enfermedad pulmonar o cardíaca que requiere de estudios x n.º de años de fumador
radiológicos de tórax. La radiografía de tórax simple en ge- = n.º de paquetes/año
20
neral nos da poca información sobre las características pro-
pias del nódulo, y el estudio de elección para determinar si
De esta manera se puede determinar que una persona
el nódulo tiene características de un posible cáncer pulmo-
que ha fumado una cajetilla de cigarrillos al día por 20 años
nar es la tomografía computarizada (TC) de tórax2.
tiene un índice tabáquico de 20, lo que la coloca en un área
de alto riesgo para padecer cáncer de pulmón9.
FACTORES DE RIESGO PARA CÁNCER
DE PULMÓN EN PACIENTE CON UN
CARACTERIZACIÓN RADIOLÓGICA
NÓDULO PULMONAR
Como ya se había mencionado anteriormente, la radio-
Parte fundamental del estudio de un nódulo pulmonar
grafía simple de tórax no es un estudio suficiente para eva-
solitario es determinar si el paciente cuenta con factores de
luar las características radiológicas de un nódulo pulmonar;
riesgo para ser portador de un nódulo pulmonar maligno.
sin embargo, es el estudio en el que generalmente se rea-
En múltiples estudios se ha demostrado que los factores de
liza el hallazgo del nódulo (Fig. 1).
riesgo para que un nódulo pulmonar encontrado en estu-
En el momento en el que se encuentra un nódulo pul-
dios radiológicos sea maligno son la edad del paciente, la
monar, se debe hacer todo lo posible para obtener la his-
historia de malignidad previa en el paciente, la historia de
toria radiológica del paciente. En el caso de que éste cuen-
tabaquismo del paciente y el tiempo posterior al cese del
te con estudios radiológicos previos, se deberá realizar una
tabaquismo3-5. En la tabla 1 se pude observar cómo catego-
búsqueda intencionada del nódulo que estamos viendo en
rizar a los pacientes con un nódulo pulmonar en alto, me-
el estudio actual: si la lesión se encuentra presente en es-
dio o bajo riesgo por sus características clínicas6.
tudios de más de dos años previos al actual y sus

106
Nódulo pulmonar solitario

Tabla 1. Características de riesgo del paciente para tener un nódulo pulmonar maligno
Característica Riesgo
Bajo Medio Alto

Tamaño del nódulo < 8 mm 8-20 mm > 20 mm


Edad < 45 años 45-60 años > 60 años
Antecedentes neoplásicos Sin antecedentes Con antecedentes
Tabaquismo Sin tabaquismo < 20 paquetes/año > 20 paquetes/año
Cese del tabaquismo > 7 años < 7 años Activo
EPOC No Sí
Características del nódulo Aspecto benigno Indeterminado Aspecto maligno
en la TC

relaciona directamente con las posibilidades de ser malig-


no: los nódulos < 5 mm son muy poco probables de ser
malignos, mientras que los nódulos > 11 mm aumentan
sus posibilidades de ser malignos hasta un 33% y los nódu-
los > 20 mm llegan hasta un 80% de malignidad10. En rela-
ción con los bordes del nódulo, generalmente los nódulos
con un contorno bien definido, de bordes lisos y bien re-
dondeados se asocian a mayor posibilidad de ser benignos,
mientras que los bordes irregulares, espiculados y mal de-
finidos se asocian con mayor frecuencia a malignidad11. La
presencia de las espiculaciones en los bordes de los nódu-
los se encuentra en relación con el crecimiento de células
neoplásicas a lo largo del intersticio pulmonar, mientras
que los bordes irregulares y mal definidos están asociados
al crecimiento irregular de diferentes zonas dentro de una
neoplasia maligna12, por esto son de gran ayuda para de-
Figura 1. Radiografía simple de tórax con presencia de nódulo en región
terminar el riesgo de que un nódulo sea de origen maligno.
parahiliar izquierda.
La presencia de bordes espiculados, llamados en ocasiones
imágenes de corona radiada, tiene un alto valor predictivo
de malignidad, que puede llegar a ser del 90%, motivo por
características se mantienen sin cambios, el paciente no el que los nódulos que presentan esta característica en la
requerirá mayor estudio de ese nódulo; si, por el contrario, mayoría de las ocasiones deben ser estudiados a través de
la lesión actual no está presente en estudios previos o sus métodos de citología o histología13 (Fig. 2).
características han cambiado, como el aumento de tamaño La presencia de calcificaciones dentro del nódulo gene-
o el que se asocie a retracción o engrosamiento pleural, se ralmente aumenta la posibilidad de benignidad. Las calci-
deberá continuar con el estudio del paciente mediante la ficaciones que normalmente se asocian a benignidad son
obtención de una TC de tórax. las difusas, centrales, laminares y lobuladas o en «palomita
La TC de tórax es el estudio de elección para poder de maíz». Las calcificaciones difusas, centrales o laminares
determinar las características radiológicas del tumor y con en general se asocian con la presencia de un granuloma,
ello proponer alguna otra medida de diagnóstico radioló- mientras que las calcificaciones lobuladas se asocian ma-
gico o histológico. Las características que normalmente se yormente a tumores benignos de tipo del hamartoma14
estudian en un nódulo pulmonar en la TC son el tamaño, (Fig. 3).
los bordes, las características internas (densidad, atenua- Es importante resaltar que se puede observar algún
ción, calcificaciones, perfusión) y las características del te- tipo de calcificación en carcinomas de pulmón; sin embar-
jido que lo circunda, como la presencia de retracción pleu- go, estas calcificaciones en general son puntiformes, irre-
ral, broncograma aéreo y alteración de la estructura normal gulares o espiculadas dentro del nódulo y no siguen el
del parénquima pulmonar que lo rodea. Como se había mismo patrón de las calcificaciones benignas mencionadas
mencionado anteriormente, el tamaño del nódulo se anteriormente15.

107
Oncología general para profesionales de la salud de primer contacto

A B

Figura 2. Característica de los bordes de un nódulo en la TC. A: nódulo pulmonar con características benignas, de aspecto redondeado, bordes lisos
y regulares que no alteran la anatomía normal del parénquima pulmonar. B: nódulo pulmonar con características de malignidad, con bordes irregulares,
espiculados y áreas de retracción pleural.

Durante la realización de la TC para el estudio de un


nódulo pulmonar se puede solicitar la realización de un
estudio de perfusión del nódulo, durante el cual se realizan
mediciones de las UH del nódulo antes de la administra-
ción intravenosa de medio de contraste iónico y a los 2, 3
y 4 min. El aumento de la densidad medida en el nódulo
está relacionada con la vascularidad y riego sanguíneo del
mismo, el cual es mayor en lesiones malignas, por lo que
un aumento de 20 unidades Houdsfield (HD) o más en el
nódulo es altamente sugestivo de una neoplasia maligna17.
En caso de que hayamos decidido mantener en obser-
vación y dar seguimiento a un paciente con un nódulo
Figura 3. Nódulo pulmonar en el que se observan calcificaciones periféricas pulmonar solitario, uno de los factores determinantes para
sugestivas de granuloma.
decidir realizar alguna intervención en dicho paciente es el
crecimiento del nódulo. En la TC el crecimiento del nódulo
se mide calculando el tiempo que tarda en duplicar su
La evolución de la tecnología en la TC ha permitido que volumen. Recordemos que, a pesar de que en estudios ra-
cada vez seamos más capaces de ver características finas diológicos vemos un nódulo como una circunferencia, en
de los nódulos, lo que ha facilitado localizar nódulos que realidad es una esfera; por tanto, duplicar el volumen de un
no están completamente sólidos dentro del parénquima nódulo implica el aumento de alrededor del 30% de su
pulmonar; a estos nódulos se les llama subsólidos y se ca- circunferencia, es decir, que para que un nódulo de 10 mm
racterizan ser nódulos con áreas denominadas de «vidrio duplique su volumen, en el siguiente estudio deberá medir
despulido». Estos nódulos en su mayoría se deben a proce- 13 mm aproximadamente. La velocidad de duplicación se
sos infecciosos o inflamatorios; sin embargo, hasta el 50% mide en días: un nódulo maligno tiene un promedio de
de los adenocarcinomas pulmonares inician con patrones velocidad de duplicación de 20 a 400 días, con una media
de nódulo subsólido, por lo que es importante mantener- de 100 días, por lo que se recomienda realizar los estudios
los bajo una vigilancia estrecha con el fin de detectar cam- de seguimiento con una diferencia de cuatro meses entre
bios en sus características que puedan sugerir una neopla- cada uno para poder definir si el nódulo está creciendo. En
sia maligna, como aumento del tamaño, confluencia de las caso de que un nódulo sólido se mantenga sin cambios
áreas subsólidas que forman un nódulo sólido, retracción durante un periodo de dos años, podemos afirmar que es
de las estructuras normales del pulmón produciendo espi- un nódulo benigno; mientras que un nódulo que aumenta
culaciones o retracción de la pleura, lo que en su caso ame- su tamaño en un 30% en cualquiera de nuestros estudios
ritaría el estudio histológico de ese pulmón con el fin de de control deberá ser considerado un nódulo altamente
descartar la presencia de un tumor maligno que se esté sugestivo de malignidad y se deberán tomar medidas para
desarrollando en ese sitio16 (Fig. 4). su tratamiento18. Los nódulos subsólidos presentan un cre-
cimiento mucho más lento que los nódulos sólidos, por lo

108
Nódulo pulmonar solitario

A B

Figura 4. Tomografía computarizada con nódulos subsólidos. A: se observa claramente la presencia de la imagen nodular de aspecto en «vidrio despulido».
B: nódulo subsólido de aspecto maligno que ya presenta un área sólida central (flecha).

A B

Figura 5. A: TC con nódulo pulmonar de aspecto maligno. B: mismo paciente en estudio de PET-CT con gran captación del radiotrazador, lo que sugiere
malignidad.

que el seguimiento se deberá mantener por al menos cinco la presencia de un nódulo maligno en una PET-CT, como
años, y en algunas publicaciones se refieren hasta siete las enfermedades granulomatosas infecciosas o inflamato-
años19. rias, como la tuberculosis pulmonar o las micosis pulmona-
El uso de la tomografía por emisión de positrones-to- res20,21 (Fig. 5).
mografía computarizada (PET-CT) en pacientes con nódulo
pulmonar solitario está reservado para aquellos pacientes
con una muy alta sospecha de malignidad y en los que se DIAGNÓSTICO HISTOLÓGICO DEL
desee descartar la presencia de enfermedad metastásica. NÓDULO PULMONAR SOLITARIO
Como la PET-CT es un estudio caro, se deberá utilizar de Una vez determinado que el nódulo pulmonar que pre-
manera juiciosa y con indicaciones claras. El uso de la PET- senta el paciente es de alto riesgo para ser una neoplasia,
CT en nódulos < 10 mm tiene baja sensibilidad, por lo que ya sea porque es un nódulo sugestivo de malignidad en
no se requiere su uso en estos casos. En casos de pacientes estudios radiológicos o porque el paciente es un paciente
con nódulos > 10 mm de diámetro en su parte sólida, se de alto riesgo (fumador mayor de 60 años de edad) con un
dará como positivos a la posibilidad de malignidad cuando nódulo indeterminado, el siguiente paso es obtener un
la captación en el nódulo sea ≥ 2.4 Standard Uptake Value diagnóstico citológico o histológico de certeza de benigni-
(SUV). En el caso de que se cuente con un estudio de PET- dad o malignidad. Los métodos diagnósticos utilizados
CT positivo en el nódulo pulmonar, se deberá efectuar una para este fin son: punción transtorácica con guía radiológi-
corroboración citológica o histológica de malignidad del ca o sin ella, broncoscopia o procedimientos quirúrgicos
mismo, ya que existen otras etiologías que pueden emular como resección por toracoscopia o toracotomía. Lo ideal

109
Oncología general para profesionales de la salud de primer contacto

Figura 6.

siempre será que el estudio histológico de un nódulo pul- nódulo < 30 mm, periférico y que en general se encuentra
monar se realice en un centro especializado en este tipo de lejos del alcance del broncoscopio tradicional. Mediante el
padecimientos. uso de técnicas convencionales de broncoscopia se ha en-
Actualmente las recomendaciones para el uso de méto- contrado una certeza diagnóstica tan baja como del 30%
dos diagnósticos menos invasivos a la cirugía son para en pacientes con nódulos < 2 cm, que aumenta al 60%
aquellos nódulos con probabilidad baja-intermedia de ma- cuando el nódulo es > 2 cm, y más central aún en las ma-
lignidad, ya que en pacientes con nódulos con alta proba- nos más expertas en el uso de esta técnica, mientras que
bilidad de ser malignos la recomendación es la resección el uso de fluoroscopia como guía para la toma de biopsias
quirúrgica con estudio histológico transoperatorio22. no ha demostrado aumentar la tasa de éxito24.
El uso de la punción transtorácica guiada por TC ha
demostrado una buena sensibilidad; sin embargo, siempre
se deberán tomar en cuenta el tamaño del nódulo, la proxi- TRATAMIENTO QUIRÚRGICO
midad a la pared torácica, el calibre de la aguja utilizada y DEL NÓDULO PULMONAR
la posibilidad de contar con un citopatólogo en el sitio El tratamiento quirúrgico del nódulo pulmonar es el
para que determine que la muestra es adecuada, pues método de elección para manejar este tipo de problemas.
todo ello aumenta las posibilidades de tener un buen diag- La resección quirúrgica de un nódulo pulmonar se encuen-
nóstico. Por ello, nódulos ≥ 10 mm y que se encuentran a tra indicada en tres instancias25,26:
una distancia promedio de 30 mm de la pleura parietal al- • El nódulo se diagnosticó como maligno, en cuyo caso
canzan un diagnóstico en un 66% de los casos. Sin embar- la resección quirúrgica es el tratamiento de elección.
go, este procedimiento no está libre de complicaciones, y • El nódulo es sugestivo de malignidad a pesar de tener
se llega a presentar un neumotórax en el 52% de los pa- biopsias u otros estudios que no han demostrado ma-
cientes, con necesidad de colocación de un tubo de drena- lignidad en sus resultados.
je pleural en el 9% de éstos23. • El nódulo es altamente sugestivo de malignidad en
El uso de la broncoscopia para nódulos pulmonares so- un paciente de alto riesgo, en cuyo caso no se reco-
litarios es cada vez más aceptado con el uso de técnicas miendan otros estudios y es preferible realizar la re-
como el ultrasonido radial o la electronavegación; sin em- sección quirúrgica como primer paso de diagnóstico
bargo, aun así, el diagnóstico certero es muy variable, ya y terapéutico.
que por definición un nódulo pulmonar solitario es un

110
Nódulo pulmonar solitario

La resección quirúrgica del nódulo permite tener un 7. Wahidi MM, Govert JA, Goudar RK, Gould MK, McCrory DC, American College
of Chest Physicians. Evidence for the Treatment of Patients With Pulmonary
diagnóstico preciso de la etiología del mismo y, al mismo Nodules: When Is It Lung Cancer? Chest. 2007;132(3):94S-107S.
tiempo, realizar un procedimiento de tratamiento definiti- 8. Henschke CI, McCauley DI, Yankelevitz DF, et al. Early Lung Cancer Action
Project: overall design and findings from baseline screening. Lancet.
vo en caso de que en el estudio transoperatorio el resulta-
1999;354(9173):99-105.
do sea de un nódulo maligno. En caso de que el nódulo sea 9. Torre Bouscoulet L, De la Garza Salazar J, García Sancho Figueroa C, Palomar
secundario a un cáncer pulmonar, el procedimiento quirúr- Lever A. Generalidades del cáncer de pulmón de células no pequeñas. En:
Arriete Rodríguez O, Guzmán de Alba E, eds. Cáncer de pulmón de células no
gico estándar para su tratamiento es la lobectomía con la pequeñas. México; 2016. p. 1-14.
resección de los nodos ganglios linfáticos regionales, inclu- 10. Naidich DP, Bankier AA, MacMahon H, et al. Recommendations for the Manage-
ment of Subsolid Pulmonary Nodules Detected at CT: A Statement from the
yendo los ganglios mediastinales. Existen otros procedi-
Fleischner Society. Radiology. 2013;266(1):304-17.
mientos aceptados como la segmentectomía anatómica y 11. Gurney JW, Lyddon DM, McKay JA. Determining the likelihood of malignancy
la resección en cuña acompañados de la resección de los in solitary pulmonary nodules with Bayesian analysis. Part II. Application. Ra-
diology. 1993;186(2):415-22.
ganglios regionales y mediastinales; sin embargo, la deci- 12. Heitzman ER, Markarian B, Raasch BN, Carsky EW, Lane EJ, Berlow ME. Pathways
sión de alguno de estos procedimientos depende de las of tumor spread through the lung: radiologic correlations with anatomy and
pathology. Radiology. 1982;144(1):3-14.
características del nódulo y del paciente, y deberá ser rea-
13. Winer-Muram HT. The Solitary Pulmonary Nodule. Radiology. 2006;239(1):34-49.
lizada exclusivamente por cirujanos altamente capacitados 14. Truong MT, Ko JP, Rossi SE, et al. Update in the Evaluation of the Solitary Pul-
en tratar pacientes con cáncer de pulmón27. monary Nodule. RadioGraphics. 2014;34(6):1658-79.
15. Grewal RG, Austin JH. CT demonstration of calcification in carcinoma of the
Actualmente existen dos modalidades de tratamiento lung. J Comput Assist Tomogr. 1994;18(6):867-71.
quirúrgico para un nódulo pulmonar: la cirugía de mínima 16. Lee HY, Choi YL, Lee KS, et al. Pure Ground-Glass Opacity Neoplastic Lung
Nodules: Histopathology, Imaging, and Management. Am J Roentgenol.
invasión o videotoracoscopia, la cual actualmente es el mé-
2014;202(3):W224-33.
todo de elección, ya que representa una técnica con menor 17. Swensen SJ, Viggiano RW, Midthun DE, et al. Lung Nodule Enhancement at CT:
molestia para el paciente, menor tiempo de recuperación Multicenter Study. Radiology. 2000;214(1):73-80.
18. Nathan MH, Collins VP, Adams RA. Differentiation of Benign and Malignant
e inicio de sus actividades cotidianas, con baja morbimor- Pulmonary Nodules by Growth Rate. Radiology. 1962;79(2):221-32.
talidad y con los mismos resultados desde el punto de 19. Aoki T, Nakata H, Watanabe H, et al. Evolution of Peripheral Lung Adenocarci-
nomas. Am J Roentgenol. 2000;174(3):763-8.
vista oncológicos, y la resección del nódulo mediante tora-
20. Cronin P, Dwamena BA, Kelly AM, Bernstein SJ, Carlos RC. Solitary pulmonary
cotomía28 (Fig. 5). nodules and masses: a meta-analysis of the diagnostic utility of alternative
En la figura 6 se muestra en flujograma para el buen imaging tests. Eur Radiol. 2008;18(9):1840-56.
21. Yilmaz F, Tastekin G. F-FDG PET in evaluation of solitary pulmonary nodules. Int
diagnóstico de un nódulo pulmonar solitario. J Clin Exp Med. 2015;8(1):45-51.
22. Gould MK, Donington J, Lynch WR, et al. Evaluation of Individuals With Pulmo-
nary Nodules: When Is It Lung Cancer? Chest. 2013;143(5):e93S-e120S.
BIBLIOGRAFÍA 23. Ng YL, Patsios D, Roberts H, et al. CT-guided percutaneous fine-needle aspira-
tion biopsy of pulmonary nodules measuring 10 mm or less. Clin Radiol.
1. Ost DE, Gould MK. Decision making in patients with pulmonary nodules. Am J 2008;63(3):272-7.
Respir Crit Care Med. 2012;185(4):363-72. 24. Rivera MP, Mehta AC, American College of Chest Physicians. Initial Diagnosis of
2. Ost D, Fein AM, Feinsilver SH. The Solitary Pulmonary Nodule. N Engl J Med. Lung Cancer. Chest. 2007;132(3):131S-48.
2003;348(25):2535-42. 25. Baldwin DR. Management of pulmonary nodules according to the 2015 British
3. Al-Ameri A, Malhotra P, Thygesen H, et al. Risk of malignancy in pulmonary Thoracic Society guidelines. Key messages for clinical practice. Polish Arch In-
nodules: A validation study of four prediction models. Lung Cancer. tern Med. 2016;126(4):262-74.
2015;89(1):27-30. 26. Sihoe ADL, Hiranandani R, Wong H, Yeung ESL. Operating on a suspicious lung
4. Swensen SJ, Silverstein MD, Ilstrup DM, Schleck CD, Edell ES. The probability of mass without a preoperative tissue diagnosis: pros and cons. Eur J Cardiothorac
malignancy in solitary pulmonary nodules. Application to small radiologically Surg. 2013;44(2):231-7; discussion 237.
indeterminate nodules. Arch Intern Med. 1997;157(8):849-55. 27. Liu Y, Huang C, Liu H, Chen Y, Li S. Sublobectomy versus Lobectomy for stage
5. Gould MK, Ananth L, Barnett PG, Veterans Affairs SNAP Cooperative Study IA (T1a) non-small-cell lung cancer: a meta-analysis study. World J Surg Oncol.
Group. A clinical model to estimate the pretest probability of lung cancer in 2014;12(1):138.
patients with solitary pulmonary nodules. Chest. 2007;131(2):383-8. 28. Cai Y, Fu X, Xu Q, Sun W, Zhang N. Thoracoscopic Lobectomy versus Open
6. Zhan P, Xie H, Xu C, Hao K, Hou Z, Song Y. Management strategy of solitary Lobectomy in Stage I Non-Small Cell Lung Cancer: A Meta-Analysis. PLoS One.
pulmonary nodules. J Thorac Dis. 2013;5(6):824-9. 2013;8(12):e82366.

111
Clinical Review & Education

JAMA | US Preventive Services Task Force | RECOMMENDATION STATEMENT

Screening for Lung Cancer


US Preventive Services Task Force Recommendation Statement
US Preventive Services Task Force

Viewpoint page 933 and


IMPORTANCE Lung cancer is the second most common cancer and the leading cause of Editorial page 939
cancer death in the US. In 2020, an estimated 228 820 persons were diagnosed with lung Multimedia
cancer, and 135 720 persons died of the disease. The most important risk factor for lung
Related articles pages 971 and
cancer is smoking. Increasing age is also a risk factor for lung cancer. Lung cancer has a
988 and JAMA Patient Page
generally poor prognosis, with an overall 5-year survival rate of 20.5%. However, early-stage page 1016
lung cancer has a better prognosis and is more amenable to treatment.
Supplemental content
OBJECTIVE To update its 2013 recommendation, the US Preventive Services Task Force
CME Quiz at
(USPSTF) commissioned a systematic review on the accuracy of screening for lung cancer
jamacmelookup.com and CME
with low-dose computed tomography (LDCT) and on the benefits and harms of screening for Questions page 1000
lung cancer and commissioned a collaborative modeling study to provide information about
the optimum age at which to begin and end screening, the optimal screening interval, and the Related articles at
jamaoncology.com
relative benefits and harms of different screening strategies compared with modified
jamanetworkopen.com
versions of multivariate risk prediction models. jamasurgery.com
POPULATION This recommendation statement applies to adults aged 50 to 80 years who have
a 20 pack-year smoking history and currently smoke or have quit within the past 15 years.

EVIDENCE ASSESSMENT The USPSTF concludes with moderate certainty that annual screening
for lung cancer with LDCT has a moderate net benefit in persons at high risk of lung cancer
based on age, total cumulative exposure to tobacco smoke, and years since quitting smoking.

RECOMMENDATION The USPSTF recommends annual screening for lung cancer with LDCT
in adults aged 50 to 80 years who have a 20 pack-year smoking history and currently smoke
or have quit within the past 15 years. Screening should be discontinued once a person
has not smoked for 15 years or develops a health problem that substantially limits life Author/Group Information: The US
Preventive Services Task Force
expectancy or the ability or willingness to have curative lung surgery. (B recommendation) (USPSTF) members are listed at the
This recommendation replaces the 2013 USPSTF statement that recommended annual end of this article.
screening for lung cancer with LDCT in adults aged 55 to 80 years who have a 30 pack-year Corresponding Author: Alex H.
smoking history and currently smoke or have quit within the past 15 years. Krist, MD, MPH, Virginia
Commonwealth University, 830 E
Main St, One Capitol Square,
JAMA. 2021;325(10):962-970. doi:10.1001/jama.2021.1117 Sixth Floor, Richmond, VA 23219
(chair@uspstf.net).

Summary of Recommendation

Adults aged 50 to 80 years who have a 20 pack-year The USPSTF recommends annual screening for lung cancer with low-dose computed
smoking history and currently smoke or have quit tomography (LDCT) in adults aged 50 to 80 years who have a 20 pack-year smoking
within the past 15 years history and currently smoke or have quit within the past 15 years. Screening should
B
be discontinued once a person has not smoked for 15 years or develops a health
problem that substantially limits life expectancy or the ability or willingness to have
curative lung surgery.

See the Figure for a more detailed summary of the recommendation for clinicians. USPSTF indicates US Preventive Services Task Force.

L
ung cancer is the second most common cancer and the lead- a relative risk of lung cancer approximately 20-fold higher in smokers
ing cause of cancer death in the US. In 2020, an estimated than in nonsmokers.3 Increasing age is also a risk factor for lung cancer.
228 820 persons were diagnosed with lung cancer, and The median age of diagnosis of lung cancer is 70 years.4,5
135 720 persons died of the disease.1 Lung cancer has a generally poor prognosis, with an overall
Themostimportantriskfactorforlungcancerissmoking.2,3 Smok- 5-year survival rate of 20.5%.1 However, early-stage lung cancer has
ingisestimatedtoaccountforabout90%ofalllungcancercases,2 with a better prognosis and is more amenable to treatment.

962 JAMA March 9, 2021 Volume 325, Number 10 (Reprinted) jama.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
USPSTF Evidence Report: Screening for Lung Cancer US Preventive Services Task Force Clinical Review & Education

Figure. Clinician Summary: Screening for Lung Cancer

What does the USPSTF Adults aged 50 to 80 years who have a 20 pack-year smoking history and currently smoke or have quit within the past 15 years:
recommend? • Screen for lung cancer with low-dose computed tomography (CT) every year.
• Stop screening once a person has not smoked for 15 years or has a health problem that limits life expectancy or the ability
to have lung surgery.
Grade: B

To whom does this Adults aged 50 to 80 years who have a 20 pack-year smoking history and currently smoke or have quit within the past 15 years.
recommendation apply? (See below for definition of pack-year.)

What’s new? The USPSTF has revised the recommended ages and pack-years for lung cancer screening. It expanded the age range to 50 to
80 years (previously 55 to 80 years) and reduced the pack-year history to 20 pack-years of smoking (previously 30 pack-years).

How to implement this 1. Assess risk based on age and pack-year smoking history: Is the person aged 50 to 80 years and have they accumulated 20
recommendation? pack-years or more of smoking?
a. A pack-year is a way of calculating how much a person has smoked in their lifetime. One pack-year is the equivalent of
smoking an average of 20 cigarettes—1 pack—per day for a year.
2. Screen: If the person is aged 50 to 80 years and has a 20 pack-year or more smoking history, engage in shared decision-making
about screening.
a. The decision to undertake screening should involve a discussion of its potential benefits, limitations, and harms.
b. If a person decides to be screened, refer them for lung cancer screening with low-dose CT, ideally to a center with experience
and expertise in lung cancer screening.
c. If the person currently smokes, they should receive smoking cessation interventions.

How often? • Screen every year with low-dose CT.


• Stop screening once a person has not smoked for 15 years or has a health problem that limits life expectancy or the
ability to have lung surgery.

What are other The USPSTF has made recommendations on interventions to prevent the initiation of tobacco use in children and adolescents,
relevant USPSTF and on behavioral and pharmacotherapy interventions for tobacco smoking cessation in adults, including pregnant women.
recommendations? These recommendations are available at https://www.uspreventiveservicestaskforce.org

Where to read the full Visit the USPSTF website (https://www.uspreventiveservicestaskforce.org) to read the full recommendation statement.
recommendation This includes more details on the rationale of the recommendation, including benefits and harms; supporting evidence;
statement? and recommendations of others.

The USPSTF recognizes that clinical decisions involve more considerations than evidence alone. Clinicians should understand the evidence but individualize
decision-making to the specific patient or situation.

USPSTF indicates US Preventive Services Task Force.

Table. Summary of USPSTF Rationale


USPSTF Assessment of Magnitude of Net Benefit
Rationale Assessment
Detection The USPSTF found adequate evidence that LDCT has
The US Preventive Services Task Force (USPSTF) concludes with sufficient sensitivity and specificity to detect early–stage
moderate certainty that annual screening for lung cancer with LDCT lung cancer
has a moderate net benefit in persons at high risk of lung cancer Benefits of early The USPSTF found adequate evidence that annual screening
detection and for lung cancer with LDCT in a defined population of
based on age, total cumulative exposure to tobacco smoke, and years intervention and high-risk persons can prevent a substantial number of lung
since quitting smoking. The moderate net benefit of screening de- treatment cancer–related deaths
pends on limiting screening to persons at high risk, the accuracy of Harms of early • The harms associated with LDCT screening include
detection and false-positive results leading to unnecessary tests and
image interpretation being similar to or better than that found in clini- intervention and invasive procedures, incidental findings, short-term
cal trials, and the resolution of most false-positive results with se- treatment increases in distress due to indeterminate results,
overdiagnosis, and radiation exposure
rial imaging rather than invasive procedures. • The USPSTF found adequate evidence that the harms of
screening for lung cancer with LDCT are moderate in
See the Figure, Table, and eFigure in the Supplement for more magnitude
information on the USPSTF recommendation rationale and assess- USPSTF The USPSTF concludes with moderate certainty that annual
ment. For more details on the methods the USPSTF uses to deter- assessment screening for lung cancer with LDCT is of moderate net
benefit for persons at high risk of lung cancer based on age,
mine the net benefit, see the USPSTF Procedure Manual.6 total cumulative exposure to tobacco smoke, and years
since quitting smoking
Abbreviations: LDCT, low-dose computed tomography; USPSTF, US Preventive
Services Task Force.
Practice Considerations
Patient Population Under Consideration Assessment of Risk
This recommendation applies to adults aged 50 to 80 years who Smoking and older age are the 2 most important risk factors for
have a 20 pack-year smoking history and currently smoke or have lung cancer. 3-5 The risk of lung cancer in persons who smoke
quit within the past 15 years. increases with cumulative quantity and duration of smoking and

jama.com (Reprinted) JAMA March 9, 2021 Volume 325, Number 10 963

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
Clinical Review & Education US Preventive Services Task Force USPSTF Evidence Report: Screening for Lung Cancer

with age but decreases with increasing time since quitting for Screening Eligibility, Screening Intervals, and Starting
persons who formerly smoked.3 The USPSTF considers adults aged and Stopping Ages
50 to 80 years who have a 20 pack-year smoking history and cur- As noted above, the USPSTF recommends annual screening for lung
rently smoke or have quit within the past 15 years to be at high risk cancer with LDCT in adults aged 50 to 80 years who have at least a
and recommends screening for lung cancer with annual LDCT in 20 pack-year smoking history. Screening should be discontinued
this population. once a person has not smoked for 15 years.
African American/Black (Black) men have a higher incidence of The NLST9 and the NELSON trial11 enrolled generally healthy per-
lung cancer than White men, and Black women have a lower inci- sons, so those study findings may not accurately reflect the bal-
dence than White women.1 These differences are likely related to ance of benefits and harms in persons with comorbid conditions. The
differences in smoking exposure (ie, prevalence of smoking) and re- USPSTF recommends discontinuing screening if a person develops
lated exposure to carcinogens in cigarettes.7,8 The differences may a health problem that substantially limits life expectancy or the abil-
also be related to other social risk factors. ity or willingness to have curative lung surgery.
Other risk factors for lung cancer include environmental
exposures, prior radiation therapy, other (noncancer) lung dis- Smoking Cessation Counseling
eases, and family history. Lower level of education is also associ- All persons enrolled in a screening program who are current smok-
ated with a higher risk of lung cancer.7 The task force recommends ers should receive smoking cessation interventions. To be consis-
using age and smoking history to determine screening eligibility tent with the USPSTF recommendation on counseling and inter-
rather than more elaborate risk prediction models because there is ventions to prevent tobacco use and tobacco-caused disease,19
insufficient evidence to assess whether risk prediction model– persons referred for lung cancer screening through primary
based screening would improve outcomes relative to using the care should receive these interventions concurrent with referral.
risk factors of age and smoking history for broad implementation Because many persons may enter screening through pathways
in primary care. besides referral from primary care, the USPSTF encourages incor-
porating such interventions into all screening programs.
Screening Tests
Low-dose computed tomography has high sensitivity and reason- Shared Decision-making
able specificity for the detection of lung cancer, with demon- Shared decision-making is important when clinicians and patients
strated benefit in screening persons at high risk.9-11 Other potential discuss screening for lung cancer. The benefit of screening varies
screening modalities that are not recommended because they have with risk because persons at higher risk are more likely to benefit.
not been found to be beneficial include sputum cytology, chest ra- Screening does not prevent most lung cancer deaths; thus,
diography, and measurement of biomarker levels.12,13 smoking cessation remains essential. Lung cancer screening
has the potential to cause harm, including false-positive results
Screening Intervals and incidental findings that can lead to subsequent testing and
The 2 lung cancer screening trials that showed a benefit of lung treatment, including the anxiety of living with a lung lesion that
cancer screening used different screening intervals. The National may be cancer. Overdiagnosis of lung cancer and the risks of
Lung Screening Trial (NLST) screened annually for 3 years.9 The radiation exposure are harms, although their exact magnitude is
Nederlands-Leuvens Longkanker Screenings Onderzoek (NELSON) uncertain. The decision to undertake screening should involve a
trial screened at intervals of 1 year, then 2 years, then 2.5 years.11 thorough discussion of the potential benefits, limitations, and
Modeling studies from the Cancer Intervention and Surveillance harms of screening.
Modeling Network (CISNET)14,15 suggest that annual screening for
lung cancer leads to greater benefit than does biennial screening. Standardization of LDCT Screening and Follow-up
Based on the available evidence and these models, the USPSTF of Abnormal Findings
recommends annual screening. The randomized clinical trials (RCTs) that provide evidence for
the benefit of screening for lung cancer with LDCT were pri-
Treatment and Interventions marily conducted in academic centers with expertise in the per-
Lung cancer can be treated with surgery, chemotherapy, radiation formance and interpretation of LDCT and the management of
therapy, targeted therapies, immunotherapy, or combinations of lung lesions seen on LDCT. Clinical settings that have similar expe-
these treatments.16 Surgical resection is generally considered the rience and expertise are more likely to duplicate the beneficial
current treatment of choice for patients with stage I or II non–small results found in trials.
cell lung cancer (NSCLC).17 In an effort to minimize the uncertainty and variation about
the evaluation and management of lung nodules and to standard-
Implementation of Lung Cancer Screening ize the reporting of LDCT screening results, the American College
Available data indicate that uptake of lung cancer screening is low. of Radiology developed the Lung Imaging Reporting and Data Sys-
One recent study using data for 10 states found that 14.4% of per- tem (Lung-RADS) classification system and endorses its use in lung
sons eligible for lung cancer screening (based on 2013 USPSTF cri- cancer screening.20 Lung-RADS provides guidance to clinicians on
teria) had been screened in the prior 12 months.18 Increasing lung which findings are suspicious for cancer and the suggested man-
cancer screening discussions and offering screening to eligible per- agement of lung nodules detected on LDCT. Data suggest that the
sons who express a preference for it is a key step to realizing the po- use of Lung-RADS may decrease the rate of false-positive results in
tential benefit of lung cancer screening. lung cancer screening.21

964 JAMA March 9, 2021 Volume 325, Number 10 (Reprinted) jama.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
USPSTF Evidence Report: Screening for Lung Cancer US Preventive Services Task Force Clinical Review & Education

Additional Tools and Resources


The Centers for Disease Control and Prevention has several web- Box. US Preventive Services Task Force Low-Dose Computed
sites with many resources to help patients stop smoking: Tomographic Screening Recommendations for Lung Cancer
• “How to Quit” resources (https://www.cdc.gov/quit)
• Smoking Cessation: Fast Facts (https://www.cdc.gov/tobacco/ A-55-80-30-15
In 2013, The US Preventive Services Task Force (USPSTF)
data_statistics/fact_sheets/cessation/smoking-cessation-fast-
recommended annual screening for lung cancer with low-dose
facts/) computed tomography (LDCT) for adults aged 55 to 80 years who
• Tips From Former Smokers (https://www.cdc.gov/tobacco/ have a 30 pack-year smoking history and currently smoke or have
campaign/tips/) quit within the past 15 years (abbreviated as A-55-80-30-15).23
The National Cancer Institute has developed resources to help
A-50-80-20-15
patients stop smoking (https://www.smokefree.gov). It has also For this updated recommendation, the USPSTF has changed the
developed patient and clinician guides on screening for lung cancer: age range and pack-year eligibility criteria and recommends annual
• Lung Cancer Screening (PDQ)–Patient Version (https://www.cancer. screening for lung cancer with LDCT for adults aged 50 to 80 years
gov/types/lung/patient/lung-screening-pdq) who have a 20 pack-year smoking history and currently smoke or
• Lung Cancer Screening (PDQ)–Health Professional Version (https:// have quit within the past 15 years (abbreviated as A-50-80-20-15).
www.cancer.gov/types/lung/hp/lung-screening-pdq)

Other Related USPSTF Recommendations proaches (eg, to reduce false-positive results). In addition, the re-
Prevention of initiation of smoking and smoking cessation for view assessed whether the use of risk prediction models for
those who smoke are the most important interventions to prevent identifying adults at higher risk of lung cancer mortality improves
lung cancer. The USPSTF has made recommendations on interven- the balance of benefits and harms of screening compared with the
tions to prevent the initiation of tobacco use in children and use of trial eligibility criteria or variants of the prior USPSTF recom-
adolescents22 and on the use of pharmacotherapy and counseling mendation criteria.
for tobacco cessation.19 In addition to the systematic evidence review, the USPSTF com-
missioned collaborative modeling studies from CISNET14,15 to pro-
vide information about the optimal age at which to begin and end
screening, the optimal screening interval, and the relative benefits
Update of Previous USPSTF Recommendation
and harms of different screening strategies, including risk factor–
This recommendation replaces the 2013 USPSTF recommendation based strategies using age, pack-year smoking history, and years since
on screening for lung cancer. In 2013 the USPSTF recommended quitting smoking for former smokers, compared with modified ver-
annual screening for lung cancer with LDCT in adults aged 55 to sions of multivariate risk prediction models. The modeling studies
80 years who have a 30 pack-year smoking history and currently complement the evidence that the systematic review provides.
smoke or have quit within the past 15 years (abbreviated as
A-55-80-30-15).23 For this updated recommendation, the USPSTF Accuracy of Screening Tests
has changed the age range and pack-year eligibility criteria and The USPSTF reviewed several RCTs and cohort studies that re-
recommends annual screening for lung cancer with LDCT in adults ported on the sensitivity, specificity, or predictive value of LDCT, using
aged 50 to 80 years who have a 20 pack-year smoking history and eventual diagnosis of lung cancer as the reference standard.24,25 Not
currently smoke or have quit within the past 15 years (A-50-80- all of the reviewed studies reported all test accuracy data. In the stud-
20-15). Abbreviations for screening recommendations are ies that reported it, sensitivity ranged from 59% to 100%, specific-
expanded in the Box. ity ranged from 26.4% to 99.7%, positive predictive value ranged
As in the 2013 recommendation, the USPSTF recommends from 3.3% to 43.5%, and negative predictive value ranged from
that screening should be discontinued once a person has not 97.7% to 100%.
smoked for 15 years or develops a health problem that substantially In the NLST26 and NELSON trial,11 the reported sensitivities were
limits life expectancy or the ability or willingness to have curative 93.1% and 59%, respectively, and reported specificities were 76.5%
lung surgery. and 95.8%, respectively. Although the negative predictive values
were similar for the NLST and NELSON trial (99.9% and 97.7%, re-
spectively), the positive predictive values were very different (3.3%
and 43.5%, respectively). This discrepancy is largely accounted for
Supporting Evidence
by the trials’ differing definitions of a positive finding and screening
Scope of Review protocols—the NELSON trial used a volumetric approach and added
To update its 2013 recommendation, the USPSTF commissioned a an indeterminate nodule result category (ie, an indeterminate find-
systematic review24,25 on the accuracy of screening for lung cancer ing was not considered a positive result even if it led to additional
with LDCT and the benefits and harms of screening for lung cancer. testing). The NLST used an approach of maximum diameter with-
The review also assessed whether the benefits of screening vary by out an indeterminate category (ie, any nodule meeting the diam-
subgroup (eg, by race or sex) or by the number or frequency of LDCT eter criteria was considered a positive result).
scans and whether the harms associated with screening and the Three retrospective studies compared how various ap-
evaluation of lung nodules differ with the use of Lung-RADS, Inter- proaches for nodule classification would alter the accuracy of
national Early Lung Cancer Action Program (I-ELCAP), or similar ap- LDCT.21,27,28 The first study demonstrated that using Lung-RADS in

jama.com (Reprinted) JAMA March 9, 2021 Volume 325, Number 10 965

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
Clinical Review & Education US Preventive Services Task Force USPSTF Evidence Report: Screening for Lung Cancer

the NLST would have increased specificity while decreasing grams that provide this level of mortality benefit and also maxi-
sensitivity.21 The other 2 studies found that use of I-ELCAP criteria mize, or come close to maximizing, both lung cancer deaths averted
(increase in nodule size threshold to an average diameter of 5 mm, and life-years gained for any given level of LDCT screening, in at least
6 mm, or larger) would increase positive predictive value.27,28 3 of the 4 CISNET models (ie, “consensus-efficient” programs), the
majority (52%) have a minimum pack-year eligibility criterion of 20
Benefits of Early Detection and Treatment pack-years. Almost all have a starting age of 50 or 55 years, and all
The USPSTF reviewed 7 RCTs that evaluated lung cancer screening have a stopping age of 80 years.14,15
with LDCT.24,25 The NLST9 and the NELSON trial11 were the only trials Relative to the 2013 USPSTF screening program (A-55-80-30-
adequately powered to detect a lung cancer mortality benefit. 15), CISNET modeling analyses suggest that annually screening per-
The NLST, the largest RCT to date (n = 53 454), enrolled par- sons aged 50 to 80 years who have at least a 20 pack-year smoking
ticipants aged 55 to 74 years at the time of randomization who had history and currently smoke or have quit within the past 15 years
a tobacco use history of at least 30 pack-years and were current (A-50-80-20-15) would be associated with lung cancer mortality
smokers or had quit within the past 15 years. The mean pack-year reduced by 13.0% vs 9.8%, with avoiding 503 vs 381 lung cancer
smoking history in NLST participants was 56 pack-years.9 The deaths, and with 6918 life-years gained vs 4882 life-years gained
NELSON trial (n = 15 792) enrolled participants aged 50 to 74 years per 100 000 persons in the population aged 45 to 90 years over a
who had a tobacco use history of at least 15 cigarettes a day lifetime of screening.14 Thus, this screening program would be
(three-fourths of a pack per day) for more than 25 years or 10 ciga- associated with important reductions in lung cancer deaths and
rettes a day (one-half of a pack per day) for more than 30 years and increases in life-years gained compared with the previous recom-
were current smokers or had quit within the past 10 years. The mendation and is supported by new trial data and the CISNET mod-
median pack-year smoking history in NELSON trial participants was eling studies.
38 pack-years.11 Screening for lung cancer in persons at an earlier age and with
The NLST reported a relative risk reduction in lung cancer mor- fewer pack-years of smoking (ie, 20 pack-years) may also help par-
tality of 20% (95% CI, 6.8%-26.7%)9; a subsequent analysis of tially ameliorate racial disparities in screening eligibility. Data sug-
NLST data with additional follow-up and end point verification gest that Black persons who smoke have a higher risk of lung can-
reported a relative risk reduction of 16% (95% CI, 5%-25%).10 At cer than do White persons, and this risk difference is more
10 years of follow-up, the NELSON trial reported 181 lung cancer apparent at lower levels of smoking intensity.7 One recent analysis
deaths among participants in the screening group and 242 in of Southern Community Cohort Study participants found that 17%
the control group (incidence rate ratio [IRR], 0.75 [95% CI, of Black persons who smoke were eligible for lung cancer screen-
0.61-0.90]).11,24 The NLST also found a reduction in all-cause mor- ing based on the 2013 USPSTF eligibility criteria compared with
tality with LDCT screening compared with chest radiography (IRR, 31% of White persons who smoke. In the same study, among per-
0.93 [95% CI, 0.88-0.99]). Results of the other trials were impre- sons diagnosed with lung cancer, a significantly lower percentage
cise, without any statistically significant differences between of Black persons who smoke (32%) were eligible for screening
screening with LDCT and chest radiography or no screening.24 than were White persons (56%).29 Data also suggest that Latinx/
Evidence on screening interval comes from the NLST and the Hispanic persons who smoke accumulate fewer pack-years than
NELSON trial and CISNET modeling studies. The NLST screened an- White persons who smoke.30,31 A strategy of annually screening
nually for 3 years.9 The NELSON trial screened at intervals of 1 year, persons aged 50 to 80 years who have at least a 20 pack-year
then 2 years, then 2.5 years.11 The CISNET modeling studies sug- smoking history and currently smoke or have quit within the past
gest that annual screening with LDCT provides greater benefit in de- 15 years (A-50-80-20-15) would increase the relative percentage
creasing lung cancer mortality and in life-years gained compared with of persons eligible for screening by 87% overall—78% in non-
biennial screening.14 Hispanic White adults, 107% in non-Hispanic Black adults, and
Several lines of evidence suggest that screening for lung can- 112% in Hispanic adults compared with 2013 USPSTF criteria
cer in persons with fewer pack-years of smoking (ie, fewer than the (A-55-80-30-15).14 Similarly, a strategy of screening persons aged
30 pack-year eligibility criterion of the 2013 USPSTF recommenda- 50 to 80 years who have at least a 20 pack-year smoking history
tion) and at an earlier age can increase the benefits of screening. As and currently smoke or have quit within the past 15 years (A-50-
noted, the NELSON trial enrolled persons aged 50 to 74 years (about 80-20-15) would increase the relative percentage of persons eli-
one-fourth of participants were younger than 55 years) who had ac- gible for screening by 80% in men and by 96% in women, 14
cumulated fewer pack-years of smoking (half of a pack per day for because they accumulate fewer pack-years than men.32
more than 30 years or three-fourths of a pack per day for more than Simulation studies suggest that risk prediction models to
25 years).11 This trial provides empirical evidence for the benefit of determine eligibility for lung cancer screening could be associated
screening for lung cancer with LDCT in persons aged 50 to 55 years with reduced lung cancer deaths and the number of participants
and with lighter pack-year smoking histories. needed to screen to prevent 1 lung cancer death. The CISNET
The CISNET modeling studies also provided data that helped in- modeling studies commissioned by the USPSTF thus compared
form the pack-year eligibility criterion for lung cancer screening and the benefits and harms of screening programs based on risk pre-
the ages at which to start and stop screening. The USPSTF focused diction models vs risk factor–based screening (ie, using age and
on screening programs in the 1960 birth cohort (more representa- smoking history). The risk prediction models used were modified
tive of current smoking patterns compared with earlier cohorts) that versions of the PLCOm2012 model,33 the Lung Cancer Death Risk
yielded lung cancer mortality reductions at least as great as the 2013 Assessment Tool (LCDRAT) model,34 and the Bach model,35 lim-
USPSTF screening program (A-55-80-30-15). For screening pro- ited to age, sex (for those models that include sex as a variable,

966 JAMA March 9, 2021 Volume 325, Number 10 (Reprinted) jama.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
USPSTF Evidence Report: Screening for Lung Cancer US Preventive Services Task Force Clinical Review & Education

such as the LCDRAT and Bach models), smoking intensity, and gical procedures for false-positive results were reported in 0.5% to
smoking duration (and setting other potential variables such as 1.3% of all screened participants.24
race, education, body mass index, personal history of cancer, or In the NLST, false-positive results led to invasive procedures
family history of lung cancer to their reference value). Because age (needle biopsy, thoracotomy, thoracoscopy, mediastinoscopy, and
is an important risk factor for lung cancer, these risk prediction bronchoscopy) in 1.7% of patients screened. Complications oc-
models shifted screening to persons of older age and increased curred in 0.1% of patients screened, and death in the 60 days fol-
the number of lung cancer deaths averted, but screening occurs at lowing the most invasive procedure performed to evaluate a false-
older ages when there are fewer years to be gained. Thus, some positive result occurred in 0.007% of those screened.9 One study
risk prediction model–based screening programs were associated estimated that the use of Lung-RADS criteria would have pre-
with slightly increased life-years gained, while some were not or vented 23.4% of invasive procedures due to false-positive results.21
were associated with even slightly decreased life-years gained. In the CISNET modeling studies, the false-positive rate varied
Risk prediction models also were associated with increased the based on screening eligibility criteria. Relative to the 2013 USPSTF
number of over diagnosed lung cancers, which are more common criteria (A-55-80-30-15), the 2021 USPSTF criteria (A-50-80-20-
in older individuals.14 15) would result in 2.2 vs 1.9 false-positive results per person over a
It is possible that the use of a more complex risk prediction model lifetime of screening.14 Note that screening programs that start at
to determine eligibility might impose a barrier to wider implemen- younger ages or use a lower pack-year eligibility screen a larger total
tation and uptake of lung cancer screening, a service that currently number of persons.
has low uptake. Currently, there are no studies that have prospec- Determining the rate of overdiagnosis in screening trials is
tively compared the use of USPSTF criteria considering age, pack- challenging because the duration of follow-up affects the calcula-
year smoking history, and number of years since quitting vs risk pre- tion of excess, potentially over diagnosed, cancers in the screening
diction models as criteria for lung cancer screening, so it is uncertain vs control groups. Initially, the NLST reported 119 additional lung
whether using a risk prediction model would improve lung cancer cancers (1060 total cancers with LDCT vs 941 with chest radiogra-
detection and clinical outcomes. The International Lung Screening phy) after 3 screening rounds and 6.5 years of follow-up (IRR, 1.12
Trial (ILST), a prospective cohort study that is comparing the accu- [95% CI, 1.02-1.22]).9,24 With extended follow-up, the NLST found
racy of the PLCOm2012 model against the 2013 USPSTF criteria for no statistically significant difference between groups for overall
detecting lung cancer, may provide some evidence regarding this lung cancer incidence; however, this study had some methodologi-
issue.36 In summary, determining eligibility for lung cancer screen- cal limitations, including use of a different ascertainment method
ing using more complex risk prediction models may represent an during posttrial follow-up, lack of information on any posttrial
implementation barrier, and there is currently insufficient evi- screening that may have occurred in either the LDCT or chest radi-
dence to assess whether risk prediction model–based screening ography group, and missing data.39 In the NELSON trial, 40 excess
would improve outcomes relative to simply using the risk factors of lung cancers (344 cancers in the LDCT group vs 304 in the control
age and smoking history. group) were reported in the LDCT group after the a priori planned
10 years of follow-up; after 11 years of follow-up, there was an
Harms of Screening and Treatment excess of 14 cancers with LDCT.11
Harms of screening can include false-positive results leading to unnec- In the CISNET modeling studies, which account for lifetime fol-
essarytestsandinvasiveprocedures,overdiagnosis,radiation-induced low-up, the 2013 USPSTF screening program (A-55-80-30-15) would
cancer, incidental findings, and increases in distress or anxiety. result in 6.3% of screen-detected cases of lung cancer being over
The NLST reported false-positive rates of 26.3% for baseline, diagnosed lung cancers vs 6.0% lung cancers being overdiagnoses
27.2% for year 1, and 15.9% for year 2.9 The NELSON trial reported with the 2021 screening program (A-50-80-20-15).14
false-positive rates of 19.8% at baseline, 7.1% at year 1, 9.0% for males In the 9 publications reporting on radiation exposure associ-
at year 3, and 3.9% for males at year 5.5 of screening.11,37 An imple- ated with LDCT,24 the radiation exposure associated with 1 LDCT scan
mentation study through the Veterans Health Administration re- ranged from 0.65 to 2.36 mSv. For context, average annual back-
vealed a false-positive rate of 28.9% of veterans eligible for screen- ground radiation exposure in the US is 2.4 mSv. Two of the studies
ing (58% of those who were actually screened) at baseline.38 Both estimated the cumulative radiation exposure for participants un-
of these studies were conducted prior to the use of the Lung-RADS dergoing screening with LDCT. Using estimated radiation exposure
protocol for nodule classification, the use of which may reduce false- from screening and follow-up evaluations and estimates of the risk
positives, albeit at the cost of some false-negatives. One retrospec- of radiation-induced cancer deaths, the Italian Lung Cancer Screen-
tive study assessed how use of Lung-RADS would have changed the ing Trial (ITALUNG) estimated a lifetime risk of fatal cancer of 0.11
false-positive result rate in the NLST and found a false-positive rate cases per 1000 persons for LDCT after the 4 screening rounds,40
among baseline results for Lung-RADS of 12.8% (95% CI, 12.4%- and the Continuing Observation of Smoking Subjects study esti-
13.2%) vs 26.6% (95% CI, 26.1%-27.1%) for the NLST approach.21 mated a lifetime risk of 2.6 to 8.1 major cancers per 10 000 persons
The further workup of false-positive results can result in signifi- screened after 10 rounds of annual screening.41
cant harms such as additional imaging, biopsy, or surgical proce- The CISNET modeling studies found that lifetime estimates of
dures. Fourteen studies reported on the evaluation of false- radiation-related lung cancer deaths varied by eligibility criteria for
positive results. Among all patients screened, the percentage who screening. Relative to the 2013 USPSTF recommendation (A-55-80-
had a needle biopsy for false-positive results ranged from 0.09% 30-15), the 2021 USPSTF recommendation (A-50-80-20-15) would
to 0.56%. Complication rates from needle biopsy for false-positive be associated with an estimated 38.6 vs 20.6 radiation-related lung
results ranged from 0.03% to 0.07% of all patients screened. Sur- cancer deaths per 100 000 persons in the total population aged 45

jama.com (Reprinted) JAMA March 9, 2021 Volume 325, Number 10 967

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
Clinical Review & Education US Preventive Services Task Force USPSTF Evidence Report: Screening for Lung Cancer

to 90 years, or 1 death caused for every 13.0 vs 18.5 lung cancer on the effect of the current recommendation on eligibility for screen-
deaths avoided by screening.14 ing in Latinx/Hispanic persons. Last, the USPSTF added and up-
When comparing LDCT groups vs control groups for smoking dated resources and website links in the Additional Tools and
cessation or abstinence outcomes, evidence does not indicate that Resources section.
screening leads to lower rates of smoking cessation or continued ab-
stinence or to higher rates of relapse. Several studies suggest that, How Does the Evidence Fit With Biological Understanding?
compared with no screening, individuals who receive LDCT screen- Lung cancer is a proliferation of malignant cells that originate in lung
ing do not have worse health-related quality of life, anxiety, or dis- tissue. Smoking is the strongest risk factor for lung cancer. Older age
tress over 2 years of follow-up. However, screening participants who is also associated with increasing incidence of lung cancer. Lung can-
receive true-positive or indeterminate results may experience worse cer is classified into 2 major categories based on cell type and im-
health-related quality of life, anxiety, or distress in the short-term.24 munohistochemical and molecular characteristics: NSCLC, which col-
Studies reported a wide range of screening-related incidental lectively comprises adenocarcinoma, squamous cell carcinoma, and
findings that were deemed significant or required further evalua- large cell carcinoma, and small cell lung cancer. Screening is aimed
tion (4.4% to 40.7%), in part because of inconsistent definitions of at early detection of NSCLC rather than small cell lung cancer be-
what constitutes an incidental finding and which findings were clini- cause the latter is much less common and typically spreads too
cally significant.24 Older age was associated with a greater likeli- quickly to be reliably detected at an early, potentially curable stage
hood of incidental findings. Common incidental findings included by screening.
coronary artery calcification, aortic aneurysms, emphysema, and in- Currently, 79% of patients present with lung cancer that has
fectious and inflammatory processes. Other common findings were spread to regional lymph nodes or metastasized to distant sites. Only
masses, nodules, or cysts of the kidney, breast, adrenal gland, liver, 17% of patients present with localized disease. Patients with local-
thyroid, pancreas, spine, and lymph nodes. Cancers involving the kid- ized disease have a 59% 5-year survival rate, compared with 32%
ney, thyroid, or liver were ultimately diagnosed in 0.39% of NLST for those with regional spread and 6% for those with distant
participants in the LDCT group during screening.42 metastases.1 By leading to earlier detection and treatment, screen-
Incidental findings led to downstream evaluation, including con- ing for lung cancer can give patients a greater chance for cure.
sultations, additional imaging, and invasive procedures with asso-
ciated costs and burdens. The benefits of incidental detection of non-
lung cancer conditions and the balance of benefits and harms of
Research Needs and Gaps
incidental findings on LDCT screening remain uncertain.
• Implementation research addressing how best to increase the up-
Response to Public Comment take of lung cancer screening discussions in clinical practice is
A draft version of this recommendation statement was posted for needed, particularly among the populations at higher risk of death
public comment on the USPSTF website from July 7, 2020, to August from lung cancer or populations that are socially and economi-
3, 2020. Most comments generally agreed with the draft recom- cally disadvantaged.
mendation, although some requested broadening the eligibility cri- • Research is needed to evaluate whether, as lung cancer screening
teria for lung cancer screening and others mentioned that addi- is implemented in more diverse community settings, including
tional risk factors for lung cancer other than smoking exist or that among racial/ethnic minorities, among populations socially and eco-
lung cancer can occur in persons who never smoked. In response, nomically disadvantaged (for whom smoking prevalence and lung
the USPSTF acknowledges that there are risk factors for lung can- cancer incidence is higher), and in settings that screen greater num-
cer other than smoking; however, current evidence does not sup- bers of women, the balance of benefits and harms differs from
port the incorporation of these risk factors as determinants of eli- those observed in RCTs.
gibility for lung cancer screening. The USPSTF also acknowledges • Research to identify biomarkers that can accurately identify per-
that lung cancer can occur in persons who never smoked or among sons at high risk is needed to improve detection and minimize false-
persons who currently smoke or formerly smoked who do not meet positive results.
screening eligibility criteria. Nevertheless, smoking is the major risk • Research to identify technologies that can help more accurately
factor for lung cancer, all trials of screening for lung cancer have been discriminate between benign and malignant lung nodules
conducted among persons who smoke or were former smokers, and is needed.
trial and modeling data support the current USPSTF recommenda- • Research is needed on the benefits and harms of using risk pre-
tion as offering a reasonable balance of benefits and harms. diction models to select patients for lung cancer screening, includ-
Some comments suggested the use of more complex risk pre- ing whether use of risk prediction models represents a barrier to
diction models to determine eligibility for lung cancer screening. In wider implementation of lung cancer screening in primary care.
response, the USPSTF clarified language that use of these risk pre-
diction models might make implementation more difficult, and that
there are currently no lung cancer screening trials prospectively com-
Recommendations of Others
paring USPSTF eligibility criteria with risk prediction models. The
USPSTF also added a reference to the ILST, a prospective cohort The American Association for Thoracic Surgery recommends an-
study addressing this issue. nual lung cancer screening with LDCT for North Americans aged 55
In response to comments, the USPSTF also added information to 79 years with a 30 pack-year history of smoking. It also recom-
about the currently low uptake of lung cancer screening and data mends offering annual lung cancer screening with LDCT starting at

968 JAMA March 9, 2021 Volume 325, Number 10 (Reprinted) jama.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
USPSTF Evidence Report: Screening for Lung Cancer US Preventive Services Task Force Clinical Review & Education

age 50 years to persons with a 20 pack-year smoking history if there the past 15 years. It also recommends that screening not be per-
is an additional cumulative risk of developing lung cancer of 5% or formed for individuals with comorbidities that adversely influence
greater over the following 5 years.43 their ability to tolerate the evaluation of screen-detected findings
The American Cancer Society recommends annual lung cancer or tolerate treatment of an early-stage screen-detected lung can-
screening with LDCT for persons aged 55 to 74 years who are in fairly cer or that substantially limit their life expectancy.45
good health, have at least a 30 pack-year smoking history, and cur- The National Comprehensive Cancer Network recommends an-
rently smoke or have quit within the past 15 years. It also recom- nual screening for lung cancer with LDCT in persons aged 55 to 77
mends smoking cessation counseling for current smokers, shared years who have at least a 30 pack-year smoking history and cur-
decision-making about lung cancer screening, and that screening be rently smoke or have quit within the past 15 years or in persons 50
conducted in a high-volume, high-quality lung cancer screening and years or older who have at least a 20 pack-year smoking history and
treatment center.44 have at least 1 additional risk factor for lung cancer.46
The American College of Chest Physicians suggests that an- The American Academy of Family Physicians has concluded that
nual screening with LDCT should be offered to asymptomatic smok- the evidence is insufficient to recommend for or against screening
ers and former smokers aged 55 to 77 years who have smoked 30 for lung cancer with LDCT in persons at high risk of lung cancer based
pack-years or more and either continue to smoke or have quit within on age and smoking history.47

ARTICLE INFORMATION conflict-of-interest-disclosures. All members of the 2021. https://seer.cancer.gov/statfacts/html/lungb.


Accepted for Publication: January 27, 2021. USPSTF receive travel reimbursement and an html
honorarium for participating in USPSTF meetings. 2. Alberg AJ, Brock MV, Ford JG, et al.
The US Preventive Services Task Force (USPSTF) Dr Barry reported receiving grants and personal
members: Alex H. Krist, MD, MPH; Karina W. Epidemiology of lung cancer: diagnosis and
fees from Healthwise. management of lung cancer, 3rd ed: American
Davidson, PhD, MASc; Carol M. Mangione, MD,
MSPH; Michael J. Barry, MD; Michael Cabana, MD, Funding/Support: The USPSTF is an independent, College of Chest Physicians evidence-based clinical
MA, MPH; Aaron B. Caughey, MD, PhD; Esa M. voluntary body. The US Congress mandates that practice guidelines. Chest. 2013;143(5 suppl):e1S-
Davis, MD, MPH; Katrina E. Donahue, MD, MPH; the Agency for Healthcare Research and Quality e29S.
Chyke A. Doubeni, MD, MPH; Martha Kubik, PhD, (AHRQ) support the operations of the USPSTF. 3. Samet JM. Health benefits of smoking cessation.
RN; C. Seth Landefeld, MD; Li Li, MD, PhD, MPH; Role of the Funder/Sponsor: AHRQ staff assisted Clin Chest Med. 1991;12(4):669-679.
Gbenga Ogedegbe, MD, MPH; Douglas K. Owens, in the following: development and review of the 4. Key statistics for lung cancer. American Cancer
MD, MS; Lori Pbert, PhD; Michael Silverstein, MD, research plan, commission of the systematic Society. Accessed January 15, 2021. http://www.
MPH; James Stevermer, MD, MSPH; Chien-Wen evidence review from an evidence-based practice cancer.org/cancer/lung-cancer/about/key-statistics.
Tseng, MD, MPH, MSEE; John B. Wong, MD. center, coordination of expert review and public html
Affiliations of The US Preventive Services Task comment of the draft evidence report and draft
recommendation statement, and the writing and 5. Siegel RL, Miller KD, Jemal A. Cancer statistics,
Force (USPSTF) members: Fairfax Family Practice 2015. CA Cancer J Clin. 2015;65(1):5-29. doi:10.
Residency, Fairfax, Virginia (Krist); Virginia preparation of the final recommendation statement
and its submission for publication. AHRQ staff had 3322/caac.21254
Commonwealth University, Richmond (Krist);
Feinstein Institute for Medical Research at no role in the approval of the final recommendation 6. Procedure Manual. US Preventive Services Task
Northwell Health, Manhasset, New York statement or the decision to submit for publication. Force. Published 2015. Accessed January 21, 2021.
(Davidson); University of California, Los Angeles Disclaimer: Recommendations made by the https://uspreventiveservicestaskforce.org/
(Mangione); Harvard Medical School, Boston, USPSTF are independent of the US government. uspstf/procedure-manual
Massachusetts (Barry); University of California, They should not be construed as an official position 7. Haiman CA, Stram DO, Wilkens LR, et al. Ethnic
San Francisco (Cabana); Oregon Health & Science of AHRQ or the US Department of Health and and racial differences in the smoking-related risk of
University, Portland (Caughey); University of Human Services. lung cancer. N Engl J Med. 2006;354(4):333-342.
Pittsburgh, Pittsburgh (Davis); University of North Additional Contributions: We thank Howard doi:10.1056/NEJMoa033250
Carolina at Chapel Hill (Donahue); Mayo Clinic, Tracer, MD (AHRQ), who contributed to the writing 8. Stram DO, Park SL, Haiman CA, et al.
Rochester, Minnesota (Doubeni); George Mason of the manuscript, and Lisa Nicolella, MA (AHRQ), Racial/ethnic differences in lung cancer incidence in
University, Fairfax, Virginia (Kubik); University of who assisted with coordination and editing. the multiethnic cohort study: an update. J Natl
Alabama at Birmingham (Landefeld); University of Cancer Inst. 2019;111(8):811-819. doi:10.1093/jnci/
Virginia, Charlottesville (Li); New York University, Additional Information: The US Preventive
Services Task Force (USPSTF) makes djy206
New York (Ogedegbe); Stanford University,
Stanford, California (Owens); University of recommendations about the effectiveness of 9. Aberle DR, Adams AM, Berg CD, et al; National
Massachusetts Medical School, Worcester (Pbert); specific preventive care services for patients Lung Screening Trial Research Team. Reduced
Boston University, Boston, Massachusetts without obvious related signs or symptoms. It lung-cancer mortality with low-dose computed
(Silverstein); University of Missouri, Columbia bases its recommendations on the evidence of both tomographic screening. N Engl J Med. 2011;365(5):
(Stevermer); University of Hawaii, Honolulu the benefits and harms of the service and an 395-409. doi:10.1056/NEJMoa1102873
(Tseng); Pacific Health Research and Education assessment of the balance. The USPSTF does not 10. Pinsky PF, Church TR, Izmirlian G, Kramer BS.
Institute, Honolulu, Hawaii (Tseng); Tufts University consider the costs of providing a service in this The National Lung Screening Trial: results stratified
School of Medicine, Boston, Massachusetts (Wong). assessment. The USPSTF recognizes that clinical by demographics, smoking history, and lung cancer
decisions involve more considerations than histology. Cancer. 2013;119(22):3976-3983. doi:10.
Author Contributions: Dr Krist had full access to all evidence alone. Clinicians should understand the
of the data in the study and takes responsibility for 1002/cncr.28326
evidence but individualize decision-making to the
the integrity of the data and the accuracy of the specific patient or situation. Similarly, the USPSTF 11. de Koning HJ, van der Aalst CM, de Jong PA,
data analysis. The USPSTF members contributed notes that policy and coverage decisions involve et al. Reduced lung-cancer mortality with volume
equally to the recommendation statement. considerations in addition to the evidence of clinical CT screening in a randomized trial. N Engl J Med.
Conflict of Interest Disclosures: All authors benefits and harms. 2020;382(6):503-513. doi:10.1056/NEJMoa1911793
have completed and submitted the ICMJE 12. Hirales Casillas CE, Flores Fernández JM, Padilla
Form for Disclosure of Potential Conflicts of REFERENCES Camberos E, Herrera López EJ, Leal Pacheco G,
Interest. Authors followed the policy regarding 1. Cancer Stat Facts: lung and bronchus cancer. Martínez Velázquez M. Current status of circulating
conflicts of interest described at https://www. National Cancer Institute. Accessed January 15, protein biomarkers to aid the early detection of
uspreventiveservicestaskforce.org/Page/Name/

jama.com (Reprinted) JAMA March 9, 2021 Volume 325, Number 10 969

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
Clinical Review & Education US Preventive Services Task Force USPSTF Evidence Report: Screening for Lung Cancer

lung cancer. Future Oncol. 2014;10(8):1501-1513. Task Force recommendation statement. Ann Intern 36. Lim KP, Marshall H, Tammemägi M, et al; ILST
doi:10.2217/fon.14.21 Med. 2014;160(5):330-338. doi:10.7326/M13-2771 (International Lung Screening Trial) Investigator
13. Oken MM, Hocking WG, Kvale PA, et al; PLCO 24. Jonas D, Reuland DS, Reddy SM, et al. Consortium. Protocol and rationale for the
Project Team. Screening by chest radiograph and Screening for Lung Cancer With Low-Dose International Lung Screening Trial. Ann Am Thorac
lung cancer mortality: the Prostate, Lung, Computed Tomography: An Evidence Review for the Soc. 2020;17(4):503-512. doi:10.1513/AnnalsATS.
Colorectal, and Ovarian (PLCO) randomized trial. U.S. Preventive Services Task Force. Evidence 201902-102OC
JAMA. 2011;306(17):1865-1873. doi:10.1001/jama. Synthesis No. 198. Agency for Healthcare 37. van Klaveren RJ, Oudkerk M, Prokop M, et al.
2011.1591 Research and Quality; 2021. AHRQ publication Management of lung nodules detected by volume
14. Meza R, Jeon J, Toumazis I, et al. Evaluation of 20-05266-EF-1. CT scanning. N Engl J Med. 2009;361(23):2221-2229.
the Benefits and Harms of Lung Cancer Screening 25. Jonas DE, Reuland DS, Shivani SM, et al. doi:10.1056/NEJMoa0906085
With Low-Dose Computed Tomography: Screening for lung cancer with low-dose computed 38. Kinsinger LS, Anderson C, Kim J, et al.
A Collaborative Modeling Study for the U.S. tomography: updated evidence report and Implementation of lung cancer screening in the
Preventive Services Task Force. Agency for Healthcare systematic review for the US Preventive Services Veterans Health Administration. JAMA Intern Med.
Research and Quality; 2021. AHRQ publication 20- Task Force. JAMA. Published March 9, 2021. doi:10. 2017;177(3):399-406. doi:10.1001/jamainternmed.
05266-EF-2. 1001/jama.2021.0377 2016.9022
15. Meza R, Jeon J, Toumazis I, et al. Evaluation of 26. Pinsky PF, Gierada DS, Nath H, Kazerooni EA, 39. National Lung Screening Trial Research Team.
the benefits and harms of lung cancer screening Amorosa J. ROC curves for low-dose CT in the Lung cancer incidence and mortality with extended
with low-dose computed tomography: modeling National Lung Screening Trial. J Med Screen. 2013; follow-up in the National Lung Screening Trial.
study for the US Preventive Services Task Force. 20(3):165-168. doi:10.1177/0969141313500666 J Thorac Oncol. 2019;14(10):1732-1742. doi:10.1016/
JAMA. Published March 9, 2021. doi:10.1001/jama. 27. Yip R, Henschke CI, Yankelevitz DF, Smith JP. j.jtho.2019.05.044
2021.1077 CT screening for lung cancer: alternative definitions 40. Mascalchi M, Belli G, Zappa M, et al.
16. Non-Small Cell Lung Cancer Treatment of positive test result based on the National Lung Risk-benefit analysis of X-ray exposure associated
(PDQ®)–Health Professional Version. National Screening Trial and International Early Lung Cancer with lung cancer screening in the Italung-CT trial.
Cancer Institute. Updated November 18, 2020. Action Program databases. Radiology. 2014;273(2): AJR Am J Roentgenol. 2006;187(2):421-429. doi:10.
Accessed January 15, 2021. https://www.cancer. 591-596. doi:10.1148/radiol.14132950 2214/AJR.05.0088
gov/types/lung/hp/non-small-cell-lung-treatment- 28. Henschke CI, Yip R, Yankelevitz DF, Smith JP; 41. Rampinelli C, De Marco P, Origgi D, et al.
pdq International Early Lung Cancer Action Program Exposure to low dose computed tomography for
17. Yan TD, Black D, Bannon PG, McCaughan BC. Investigators. Definition of a positive test result lung cancer screening and risk of cancer: secondary
Systematic review and meta-analysis of in computed tomography screening for lung cancer: analysis of trial data and risk-benefit analysis. BMJ.
randomized and nonrandomized trials on safety a cohort study. Ann Intern Med. 2013;158(4):246-252. 2017;356:j347. doi:10.1136/bmj.j347
and efficacy of video-assisted thoracic surgery doi:10.7326/0003-4819-158-4-201302190-00004 42. Nguyen XV, Davies L, Eastwood JD, Hoang JK.
lobectomy for early-stage non-small-cell lung 29. Aldrich MC, Mercaldo SF, Sandler KL, Blot WJ, Extrapulmonary findings and malignancies in
cancer. J Clin Oncol. 2009;27(15):2553-2562. doi: Grogan EL, Blume JD. Evaluation of USPSTF lung participants screened with chest CT in the National
10.1200/JCO.2008.18.2733 cancer screening guidelines among African Lung Screening Trial. J Am Coll Radiol. 2017;14(3):
18. Zahnd WE, Eberth JM. Lung cancer screening American adult smokers. JAMA Oncol. 2019;5(9): 324-330. doi:10.1016/j.jacr.2016.09.044
utilization: a behavioral risk factor surveillance 1318-1324. doi:10.1001/jamaoncol.2019.1402 43. Jaklitsch MT, Jacobson FL, Austin JH, et al. The
system analysis. Am J Prev Med. 2019;57(2):250-255. 30. Kaplan RC, Bangdiwala SI, Barnhart JM, et al. American Association for Thoracic Surgery
doi:10.1016/j.amepre.2019.03.015 Smoking among U.S. Hispanic/Latino adults: the guidelines for lung cancer screening using low-dose
19. Siu AL; US Preventive Services Task Force. Hispanic community health study/study of Latinos. computed tomography scans for lung cancer
Behavioral and pharmacotherapy interventions for Am J Prev Med. 2014;46(5):496-506. doi:10.1016/j. survivors and other high-risk groups. J Thorac
tobacco smoking cessation in adults, including amepre.2014.01.014 Cardiovasc Surg. 2012;144(1):33-38. doi:10.1016/j.
pregnant women: U.S. Preventive Services Task 31. Bandiera FC, Assari S, Livaudais-Toman J, jtcvs.2012.05.060
Force recommendation statement. Ann Intern Med. Pérez-Stable EJ. Latino and Black smokers in the 44. Lung cancer screening guidelines. American
2015;163(8):622-634. doi:10.7326/M15-2023 Health and Retirement Study are more likely to Cancer Society. Accessed January 15, 2021. https://
20. Lung CT Screening Reporting & Data System quit: the role of light smoking. Tob Induc Dis. 2016; www.cancer.org/health-care-professionals/
(Lung-RADS). American College of Radiology. 14:23. doi:10.1186/s12971-016-0090-y american-cancer-society-prevention-early-
Accessed January 15, 2021. https://www.acr.org/ 32. Pinsky PF. Racial and ethnic differences in lung detection-guidelines/lung-cancer-screening-
Clinical-Resources/Reporting-and-Data-Systems/ cancer incidence: how much is explained by guidelines.html
Lung-Rads differences in smoking patterns? (United States). 45. Mazzone PJ, Silvestri GA, Patel S, et al.
21. Pinsky PF, Gierada DS, Black W, et al. Cancer Causes Control. 2006;17(8):1017-1024. doi: Screening for lung cancer: CHEST guideline and
Performance of Lung-RADS in the National Lung 10.1007/s10552-006-0038-2 expert panel report. Chest. 2018;153(4):954-985.
Screening Trial: a retrospective assessment. Ann 33. Tammemägi MC, Katki HA, Hocking WG, et al. doi:10.1016/j.chest.2018.01.016
Intern Med. 2015;162(7):485-491. doi:10.7326/M14- Selection criteria for lung-cancer screening. N Engl J 46. NCCN Guidelines for Patients: lung cancer
2086 Med. 2013;368(8):728-736. doi:10.1056/ screening. National Comprehensive Cancer
22. Owens DK, Davidson KW, Krist AH, et al; US NEJMoa1211776 Network. Published 2020. Accessed January 15,
Preventive Services Task Force. Primary care 34. Katki HA, Kovalchik SA, Petito LC, et al. 2021. https://www.nccn.org/patients/guidelines/
interventions for prevention and cessation of Implications of nine risk prediction models for content/PDF/lung_screening-patient.pdf
tobacco use in children and adolescents: US selecting ever-smokers for computed tomography 47. Clinical Preventive Service Recommendation:
Preventive Services Task Force recommendation lung cancer screening. Ann Intern Med. 2018;169(1): lung cancer. American Academy of Family
statement. JAMA. 2020;323(16):1590-1598. doi:10. 10-19. doi:10.7326/M17-2701 Physicians. Published 2013. Accessed January 15,
1001/jama.2020.4679 2021. https://www.aafp.org/patient-care/clinical-
35. Bach PB, Kattan MW, Thornquist MD, et al.
23. Moyer VA; US Preventive Services Task Force. Variations in lung cancer risk among smokers. J Natl recommendations/all/lung-cancer.html
Screening for lung cancer: U.S. Preventive Services Cancer Inst. 2003;95(6):470-478. doi:10.1093/
jnci/95.6.470

970 JAMA March 9, 2021 Volume 325, Number 10 (Reprinted) jama.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 10/21/2021
VOLUME 36 • NUMBER 11 • APRIL 10, 2018

JOURNAL OF CLINICAL ONCOLOGY O R I G I N A L R E P O R T

Nut Consumption and Survival in Patients With Stage III


Colon Cancer: Results From CALGB 89803 (Alliance)
Temidayo Fadelu, Sui Zhang, Donna Niedzwiecki, Xing Ye, Leonard B. Saltz, Robert J. Mayer, Rex B. Mowat,
Renaud Whittom, Alexander Hantel, Al B. Benson, Daniel M. Atienza, Michael Messino, Hedy L. Kindler, Alan
Venook, Shuji Ogino, Kimmie Ng, Kana Wu, Walter Willett, Edward Giovannucci, Jeffrey Meyerhardt, Ying Bao,
and Charles S. Fuchs

Author affiliations and support information


(if applicable) appear at the end of this A B S T R A C T
article.
Purpose
Published at jco.org on February 28, 2018.
Observational studies have reported increased colon cancer recurrence and mortality in patients
J.M., Y.B., and C.S.F. contributed equally with states of hyperinsulinemia, including type 2 diabetes, obesity, sedentary lifestyle, and high
to this work.
glycemic load diet. Nut intake has been associated with a lower risk of type 2 diabetes, metabolic
Nonfederal sponsors did not participate in syndrome, and insulin resistance. However, the effect of nut intake on colon cancer recurrence and
the design or conduct of the study;
collection, management, analysis, or
survival is not known.
interpretation of the data; or preparation, Patients and Methods
review, or approval of the article. The We conducted a prospective, observational study of 826 eligible patients with stage III colon cancer
views expressed in this article are those of
the authors and not an official position of
who reported dietary intake on food frequency questionnaires while enrolled onto a randomized
their affiliated institutions or funders. adjuvant chemotherapy trial. Using Cox proportional hazards regression, we assessed associations
Clinical trial information: NCT00003835.
of nut intake with cancer recurrence and mortality.
Corresponding author: Charles S. Fuchs, Results
MD, Yale Cancer Center, 333 Cedar St, After a median follow-up of 6.5 years, compared with patients who abstained from nuts, individuals
New Haven, CT 06510; e-mail: charles. who consumed two or more servings of nuts per week experienced an adjusted hazard ratio (HR) for
fuchs@yale.edu. disease-free survival of 0.58 (95% CI, 0.37 to 0.92; Ptrend = .03) and an HR for overall survival of 0.43
© 2018 by American Society of Clinical (95% CI, 0.25 to 0.74; Ptrend = .01). In subgroup analysis, the apparent benefit was confined to tree
Oncology. Creative Commons Attribution nut intake (HR for disease-free survival, 0.54; 95% CI, 0.34 to 0.85; Ptrend = .04; and HR for overall survival,
Non-Commercial No Derivatives 4.0
License.
0.47; 95% CI, 0.27 to 0.82; Ptrend = .04). The association of total nut intake with improved outcomes was
$ =
maintained across other known or suspected risk factors for cancer recurrence and mortality.

0732-183X/18/3611w-1112w/$20.00
Conclusion
Diets with a higher consumption of nuts may be associated with a significantly reduced incidence of
cancer recurrence and death in patients with stage III colon cancer.

J Clin Oncol 36:1112-1120. © 2018 by American Society of Clinical Oncology. Creative Commons
Attribution Non-Commercial No Derivatives 4.0 License: https://creativecommons.org/licenses/by-nc-nd/4.0/

plasma C-peptide or low insulin-like growth


INTRODUCTION
factor–binding protein-1 levels,9 suggesting
that the association between energy excess and
Recent prospective observational studies among increased risk of colon cancer recurrence may
patients with colon cancer suggest that diet and be mediated, in part, by long-term hyper-
lifestyle factors may significantly influence the insulinemia. In fact, increased coffee intake,
risk of colon cancer recurrence and death.1-6 In which has been associated with decreased risk
aggregate, these studies indicate that states of of T2D,10-14 lower plasma C-peptide levels,15,16
energy excess, including type 2 diabetes (T2D),7 and increased insulin sensitization,17,18 con-
obesity,6 sedentary lifestyle,2,5 Western-pattern ferred improved disease-free survival (DFS) in
diet,3 increased dietary glycemic load,1 and patients with stage III colon cancer.19
ASSOCIATED CONTENT
high intake of sugar-sweetened beverages,8 are In prospective cohort studies, increased nut
Appendix
DOI: https://doi.org/10.1200/JCO.
associated with an increased risk of colon intake has been associated with a reduced risk of
2017.75.5413 cancer recurrence and mortality. Moreover, T2D and metabolic syndrome20-23 and a re-
DOI: https://doi.org/10.1200/JCO.2017. increased cancer mortality was observed among duction in insulin resistance.20,24-27 Nuts are
75.5413 patients with colorectal cancer with elevated nutrient-dense foods that are rich in unsaturated

1112 © 2018 by American Society of Clinical Oncology


Nut Consumption and Stage III Colon Cancer Outcome

fatty acids, fiber, vitamins, minerals, and other bioactive substances


such as phenolic antioxidants and phytosterols.28-30 CALGB 89803
total enrollment
In light of evidence supporting a link between excess energy (N = 1,264)
balance, hyperinsulinemia, and increased recurrence in patients
with colon cancer, we prospectively examined the association of Did not complete Q1 (n = 169)
nut intake with cancer recurrence and mortality in a cohort of Enrolled prior to diet (n = 87)
and lifestyle incorporation
patients with stage III colon cancer enrolled onto a National into CALGB 89803
Cancer Institute–sponsored randomized clinical trial of adjuvant Refused to complete (n = 23)
chemotherapy. In the trial, detailed data on pathologic stage, or did not return Q1
performance status, postoperative treatment, and follow-up were Experienced cancer (n = 59)
recurrence, died, or were
prospectively captured. In addition, comprehensive data on diet removed from CALGB
and lifestyle were collected before any documentation of cancer 89803 before Q1
recurrence.
Completed Q1
(n = 1,095)

PATIENTS AND METHODS


Did not complete Q2
(n = 117)
Study Population
Patients in this prospective cohort participated in the National Cancer
Institute–sponsored Cancer and Leukemia Group B (now Alliance for
Clinical Trials in Oncology) 89803 adjuvant therapy trial for stage III
colon cancer, comparing therapy with weekly fluorouracil and leucovorin to Completed Q1 and Q2
weekly irinotecan, fluorouracil, and leucovorin (ClinicalTrials.gov identifier: (n = 978)
NCT000038350).31 Between April 1999 and May 2001, 1,264 patients were
enrolled. An amendment was introduced after enrolling 87 patients that
required enrollees to complete a self-administered semiquantitative food Excluded: Q2 invalid (n = 111)
frequency questionnaire (FFQ) that captured diet and lifestyle habits. The Caloric intake (Q2; n = 23)
questionnaires were administered midway through adjuvant therapy exclusion* and/or
(4 months after surgery; questionnaire 1 [Q1]) and again 6 months after left > 70 food items blank
completion of treatment (14 months after surgery; questionnaire 2 [Q2]). Experienced cancer (n = 88)
Patients were eligible if they underwent a complete surgical resection recurrence before
completing Q2
of the primary tumor within 56 days before trial entry, had no prior
chemotherapy or radiation therapy for treatment of the tumor, had re-
gional lymph node metastases without evidence of distant metastases, had Valid Q2
(n = 867)
a baseline Eastern Cooperative Oncology Group performance status of 0 to
2,32 and had adequate bone marrow, renal, and hepatic function.31 Because
of possible dietary modifications immediately after colectomy,33 we Excluded: ineligible (n = 41)
for analysis
a priori limited the primary analysis to nut intake reported on Q2. Patients
Did not answer (n = 9)
were excluded if they reported significantly abnormal caloric intake (, 600
both nut-related
or . 4,200 calories per day for men; , 500 or . 3,500 calories per day questions on Q2
for women), left . 70 food items blank, or left blank the nut Experienced cancer (n = 32)
consumption–related questions on Q2. Finally, patients were excluded if recurrence or died within
they had cancer recurrence or death before completion of Q2 or within 60 days after completing Q2
60 days of completing Q2 to avoid misattribution bias. The resulting
cohort included 826 eligible patients (Fig 1). The protocol was reviewed by Final sample
the institutional review board of each participating center, and all patients (N = 826)
provided study-specific informed consent.
Fig 1. Derivation of the study cohort. (*) Calorie exclusion: , 600 or . 4,200
calories per day for men and , 500 calories or . 3,500 calories per day for women.
Dietary Assessment Q1, questionnaire 1 (midway through adjuvant therapy); Q2, questionnaire 2
Patients in this analysis completed semiquantitative FFQs that in- (6 months after completion of adjuvant therapy). CALGB, Cancer and Leukemia
cluded 131 food items, vitamin and mineral supplements, and open-ended Group B.
sections for other supplements and foods not specifically listed.34,35 Par-
ticipants were asked how often, on average, over the previous 3 months they
Total nut intake was calculated as the weighted proportional summation of
consumed a specific food portion size, with up to nine possible responses,
tree nuts and peanuts. We similarly assessed self-reported peanut butter
which ranged from never to to six or more times per day. We computed
intake. In a previous cohort study evaluating the validity of our FFQ in
nutrient intake by multiplying the frequency of consumption of each food by
measuring intake of nuts and peanut butter, the correlation coefficients
the nutrient content of the specified portions.36 Nutrient values were energy
between dietary records and the FFQs were 0.75 and 0.75, respectively.38
adjusted using the residuals methods.37
On the questionnaire, we separately assessed self-reported intake of
1-oz servings of tree nuts and peanuts with the following eight ordered Tumor Assessments for KRAS, BRAF, PIK3CA, and TP53
categories for intake: none; less than once per month; one to three servings Mutations, Microsatellite Instability, and PTGS2 Expression
per month; one serving per week; two to four servings per week; five to six Polymerase chain reaction (PCR) and pyrosequencing targeted for
servings per week; one serving per day; and two or more servings per day. mutation hotspots in PIK3CA exons 9 and 20,39 BRAF codon 600,40 and

jco.org © 2018 by American Society of Clinical Oncology 1113


Fadelu et al

KRAS codons 12 and 13 were performed, as previously described.41,42 Mu- Statistics and Data Center and by the study chairperson following Alliance
tations in TP53 exons 5 to 8 were determined by Sanger sequencing and policies. All analyses were based on the study database frozen on November
sequencing by hybridization, as previously described.43 Microsatellite in- 9, 2009. See the Appendix (online only) for additional analyses.
stability (MSI) was assessed by PCR for 10 microsatellite markers; tumors with
instability in $ 50% of the loci were classified as MSI-high; for 28 patients
without PCR MSI results, those with loss of MLH1 or MSH2 expression were
classified as MSI-high, as previously described.44 PTGS2 (COX-2) expression RESULTS
was assessed by immunohistochemistry, as previously described.45
Baseline Characteristics
Study End Points Baseline characteristics by frequency of total nut consumption
The primary end point was DFS, defined as time from completion of are listed in Table 1. Patients who consumed more nuts were more
Q2 to tumor recurrence, occurrence of a new primary colon tumor, or frequently male, consumed more alcohol, and had a lower glycemic
death from any cause. We also assessed recurrence-free survival (RFS), load diet. Although nut users consumed greater total calories, nut
defined as time from completion of Q2 to tumor recurrence or occurrence users did not present with a higher body mass index. There were
of a new primary colon tumor. For RFS, patients who died without known
statistical differences in age, Western and prudent dietary patterns,
tumor recurrence were censored at last documented physician evaluation.
Finally, we assessed overall survival (OS), defined as the time from and dietary glycemic load by categories of nut intake, although
completion of Q2 to death from any cause. clear trends in these factors by nut intake were less apparent.

Statistical Analysis Impact of Nut Intake on Cancer Recurrence and Death


There was no statistically significant difference in OS or DFS in both Median follow-up time from completion of Q2 was 6.5 years.
arms of the randomized trial.31 Therefore, patient data from both arms During follow-up, 199 of the 826 patients experienced cancer
were combined and analyzed according to frequency categories of dietary recurrence or developed new primary tumors. Of the 826 patients,
intake. The primary analysis was done with total nut intake, combining 177 patients died; of those, 39 patients died without documented
intake of tree nuts and peanuts. For consistency with prior published
cancer recurrence.
studies on nut intake and to conserve statistical power for the analysis,
categories of nut intake were consolidated into the following five ordered Increasing total nut intake was associated with significant
categories: never, less than one serving per month, one to three servings per reduction in recurrence and mortality after adjusting for other
month, one serving per week, and two or more servings per week.46-49 predictors of cancer recurrence (Table 2). Compared with patients
Differences in distribution of baseline characteristics by nut consumption who abstained, individuals who consumed diets with two or more
categories were evaluated using the x2 test, except for continuous variables, servings of nuts per week experienced an adjusted hazard ratio
which were evaluated using the Kruskal-Wallis test. (HR) for DFS of 0.58 (95% CI, 0.37 to 0.92; Ptrend = .03). Higher
Cox proportional hazards regression analysis was used to determine
nut intake was also associated with a significant improvement in
the simultaneous affect of other variables potentially associated with each
outcome.50 The covariates in the model were fixed as measured on Q2. To OS (HR, 0.43; 95% CI, 0.25 to 0.74; Ptrend = .01) and a trend toward
account for the caloric content of nuts, our initial model adjusted for total improved RFS, which did not reach statistical significance (HR,
calorie intake.37 The final model further adjusted for potential con- 0.70; 95% CI, 0.42 to 1.16; Ptrend = .15). Unadjusted survival curves
founders including age, sex, depth of invasion through bowel wall, number are presented in Appendix Figures A1, A2, and A3 (online only).
of positive lymph nodes, baseline performance status, treatment group, We further examined the associations with nut intake models
body mass index (calculated as weight in kilograms divided by height in after controlling for other possible confounders (Western and
meters squared), physical activity (measured in metabolic equivalent task
hours per week), aspirin use, and glycemic load (as previously described).1
prudent dietary patterns, race, smoking, and alcohol use) and
Selection of covariates for the final model was based on clinical signifi- found that our findings were largely unchanged (Appendix Table
cance, previous studies, and degree of correlation with the exposure. A1, online only).
Covariates with missing variables were coded with indicator variables.
The statistical analysis was performed using nut consumption as
a continuous measure to minimize bias created by selected categorization.
Sensitivity Analyses
We tested for linear trends across frequency categories of intake by Because of possible dietary modifications immediately after
assigning each participant the median value for each frequency category colectomy,33 we a priori limited the primary analysis to nut intake
and modeling this value as a continuous variable, consistent with prior reported on Q2. Nonetheless, we repeated our analyses using the
studies.1,2,8,19 P values for trend were calculated using the Wald test of the cumulative average of nut intake on Q1 and Q2 and found that the
score variable. Secondary exploratory analysis was performed individually association with DFS was unchanged (Appendix Tables A2 and A3,
for peanuts and tree nuts; we further collapsed intake into four categories
online only).
to conserve power (never, , one serving per month, one to three servings
per month, and $ one serving per week). Stratified exploratory analyses We considered the possibility that changes in dietary habits
were also performed for other risk factors; the likelihood ratio test was used could reflect occult cancer or impending death; as such, our
to test for interaction. In addition, we performed several sensitivity an- primary analyses excluded patients who developed cancer re-
alyses to confirm the robustness of the results. The Cox regression models currence or died within 60 days of completing Q2. To further
were tested for and met the assumption of proportionality by both time- address this issue, we repeated the models after excluding re-
dependent covariate and Schoenfeld residuals methods. All analyses were currences or deaths within 180 days of Q2 completion (n = 783),
performed using SAS version 9.4 (SAS Institute, Cary, NC). A threshold
level of significance of P , .05 was considered statistically significant. All
and our results remained largely unchanged (HR, 0.54; 95% CI,
P values are two-sided and were not adjusted for multiple comparisons. 0.32 to 0.89; Ptrend = .02).
Data collection and management were conducted by the Alliance Statistics Furthermore, we considered that nut intake may be a marker
and Data Center. Data quality was ensured by review of data by the Alliance of dentition and oral health, potential confounders that were not

1114 © 2018 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Nut Consumption and Stage III Colon Cancer Outcome

Table 1. Baseline Characteristics: Total Nut Consumption (N = 826)


Servings of Nuts
, 1 per Month 1-3 per Month 1 per Week $ 2 per Week
Characteristic Never (n = 145) (n = 98) (n = 211) (n = 214) (n = 158) P*
Median nut intake, servings per week (IQR) 0 (0-0) 0.2 (0.2-0.2) 0.4 (0.4-0.4) 1.0 (0.7-1.0) 3.5 (3.0-6.0)
Median age, years (IQR) 64 (54-70) 60 (52-69) 58 (50-67) 59 (51-69) 62 (53-69) .01†
Male 74 (51.0) 46 (46.9) 114 (54.0) 129 (60.3) 103 (65.2) .02
Race .08
White 125 (86.2) 83 (84.7) 188 (89.1) 193 (90.2) 151 (95.6)
Black 11 (7.6) 9 (9.2) 17 (8.1) 10 (4.7) 5 (3.2)
Other 9 (6.2) 6 (6.1) 6 (2.8) 11 (5.1) 2 (1.3)
Performance status‡ .32
0 103 (71.0) 69 (70.4) 155 (73.5) 163 (76.2) 125 (79.1)
1-2 39 (26.9) 29 (29.6) 49 (23.2) 48 (22.4) 31 (19.6)
Unknown 3 (2.1) 0 (0.0) 7 (3.3) 3 (1.4) 2 (1.3)
No. of positive nodes .48
1-3 94 (64.8) 67 (68.4) 139 (65.9) 151 (70.6) 97 (61.4)
$4 48 (33.1) 31 (31.6) 66 (31.3) 60 (28.0) 59 (37.3)
Unknown 3 (2.1) 0 (0.0) 6 (2.8) 3 (1.4) 2 (1.3)
Tumor stage§ .61
T1, 2 29 (20.0) 15 (15.3) 33 (15.6) 32 (15.0) 29 (18.4)
T3, 4 116 (80.0) 83 (84.7) 177 (83.9) 182 (85.0) 129 (81.6)
Unknown 0 (0.0) 0 (0.0) 1 (0.5) 0 (0.0) 0 (0.0)
Differentiation .51
Well/moderate 108 (74.5) 75 (76.5) 154 (73.0) 171 (79.9) 122 (77.2)
Poor/undifferentiated 34 (23.4) 23 (23.5) 50 (23.7) 40 (18.7) 34 (21.5)
Unknown 3 (2.1) 0 (0.0) 7 (3.3) 3 (1.4) 2 (1.3)
Obstruction 32 (22.1) 18 (18.4) 47 (22.3) 50 (23.4) 34 (21.5) .91
Perforation 4 (2.8) 2 (2.0) 8 (3.8) 9 (4.2) 6 (3.8) .87
Treatment .18
FU+LV 82 (56.6) 44 (44.9) 97 (46.0) 115 (53.7) 76 (48.1)
CPT-11+FU+LV 63 (43.4) 54 (55.1) 114 (54.0) 99 (46.3) 82 (51.9)
Median calorie consumption, kcal/d (IQR) 1,624 (1,298-2,034) 1,547 (1,248-2,019) 1,692 (1,305-2,067) 1,755 (1,398-2,205) 2,162 (1,770-2,630) , .001†
Median BMI, kg/m2 (IQR) 28.7 (24.8-32.5) 27.7 (24.4-32.8) 28.7 (25.8-32.7) 28.8 (25.3-33.1) 27.9 (24.5-31.9) .28†
Median physical activity, MET-h/wk (IQR) 6.7 (0.6-17.2) 9.9 (2.3-20.2) 7.5 (1.9-21.5) 7.7 (2.1-21.3) 10.8 (2.7-26.4) .03†
Median caffeine, mg/d (IQR) 140 (30-318) 164 (46-352) 163 (73-356) 160 (46-327) 146 (51-301) .3†
Median alcohol, g/wk (IQR) 0.2 (0.1-14.2) 3.3 (0.1-56.2) 9.7 (0.1-48.9) 9.7 (0.1-71.7) 21.9 (0.1-96.0) , .001†
Median SSB, servings per week (IQR) 0.7 (0.0-3.4) 1.3 (0.4-5.5) 1.2 (0.2-4.5) 1 (0.2-4.0) 1.2 (0.2-4.0) .13†
Never-smokers 57 (39.3) 46 (46.9) 92 (43.6) 104 (48.6) 72 (45.6) .5
Aspirin use 27 (18.6) 8 (8.2) 25 (11.8) 33 (15.4) 28 (17.7) .1
Western diet, No. , median (%) 82 (56.6) 54 (55.1) 112 (53.1) 113 (52.8) 52 (32.9) , .001†
Prudent diet, No. , median (%) 81 (55.9) 63 (64.3) 132 (62.6) 87 (40.7) 51 (32.3) , .001†
Glycemic load, No. , median (%) 52 (35.9) 37 (37.8) 101 (47.9) 112 (52.3) 111 (70.3) , .001†

NOTE. Data presented as No. (%) unless otherwise indicated.


Abbreviations: BMI, body mass index; CPT-11, irinotecan; FU, fluorouracil; IQR, interquartile range; LV, leucovorin; MET, metabolic equivalent task; SSB, sugar-
sweetened beverage.
*By x2 test unless otherwise noted.
†By Kruskal-Wallis nonparametric test.
‡Baseline performance status: 0 indicates fully active; 1 indicates restricted in physically strenuous activity but ambulatory and able to carry out light work; 2 indicates
ambulatory and capable of all self-care but unable to carry out any work activities, up and about . 50% of waking hours.
§T1, 2 indicates level of invasion through the bowel wall not beyond the muscle layer; T3, 4 indicates level of invasion through the bowel wall beyond the muscle layer.

assessed in this cohort. To address this issue, we ran the models per week to nonconsumers (Fig 2). In this exploratory analysis, the
using a new reference group that combined the lowest intake association between total nut intake and DFS was consistent across
categories of never and less than one serving per month; the results strata of patient, disease, and treatment characteristics; these in-
remained largely unchanged. Individuals reporting two or more cluded strata of clinically relevant genomic alterations (MSI and
servings of nuts per week demonstrated an improvement in DFS KRAS, BRAF, and PIK3CA mutations). However, in these stratified
compared with the new reference group (HR, 0.57; 95% CI, 0.38 to analyses, statistical power to adequately detect differences was
0.87; Ptrend = .03). limited by the sample size, and such analyses should be considered
exploratory.
Stratified Analyses
We evaluated the influence of total nut intake on DFS across Subgroup Analyses
strata of other potential predictors and confounders of patient In separate analyses of the subtypes of nuts, we observed
outcome, comparing patients with an intake of two of more servings a significant improvement in both DFS and OS with increasing tree

jco.org © 2018 by American Society of Clinical Oncology 1115


Fadelu et al

Table 2. Associations Between Colon Cancer Recurrence and Mortality and Total Nut Consumption
Total Nut Consumption Categories of Intake (servings)
Outcome Never , 1 per Month 1-3 per Month 1 per Week $ 2 per Week Ptrend*
DFS
No. of events/total No. of patients 52/145 36/98 56/211 58/214 36/158
HR (95% CI), adjusted 1† Ref 1.03 (0.68 to 1.58) 0.72 (0.49 to 1.04) 0.71 (0.48 to 1.03) 0.58 (0.37 to 0.90) .02
HR (95% CI), adjusted 2‡ Ref 1.02 (0.66 to 1.57) 0.71 (0.48 to 1.04) 0.69 (0.47 to 1.02) 0.58 (0.37 to 0.92) .03
RFS
No. of events/total No. of patients 39/145 31/98 51/211 47/214 31/158
HR (95% CI), adjusted 1† Ref 1.18 (0.74 to 1.90) 0.88 (0.58 to 1.33) 0.77 (0.50 to 1.18) 0.68 (0.42 to 1.11) .08
HR (95% CI), adjusted 2‡ Ref 1.13 (0.70 to 1.82) 0.83 (0.54 to 1.27) 0.75 (0.48 to 1.15) 0.70 (0.42 to 1.16) .15
OS
No. of events/total No. of patients 44/145 27/98 43/211 39/214 24/158
HR (95% CI), adjusted 1† Ref 0.89 (0.55 to 1.43) 0.64 (0.42 to 0.98) 0.56 (0.36 to 0.86) 0.45 (0.27 to 0.75) .01
HR (95% CI), adjusted 2‡ Ref 0.91 (0.56 to 1.49) 0.64 (0.42 to 0.98) 0.54 (0.34 to 0.84) 0.43 (0.25 to 0.74) .01

Abbreviations: DFS, disease-free survival; HR, hazard ratio; OS, overall survival; Ref, reference; RFS, recurrence-free survival.
*Two-sided P value. Trend across consumption levels with the categorical median.
†Model adjusted 1: Adjusted using Cox proportional hazards regression for calorie intake.
‡Model adjusted 2: Adjusted using Cox proportional hazards regression further for age, sex, depth of invasion through bowel wall, number of positive lymph nodes,
baseline performance status, treatment group, body mass index, physical activity, aspirin use, and glycemic load.

nut consumption, whereas the associations with peanut intake did relevant intermediate markers including oxidation,25,66 endothelial
not reach statistical significance (Table 3). We similarly examined dysfunction,67 hyperglycemia,25 and insulin resistance.20,68 In
the influence of peanut butter consumption on patient outcome addition, preclinical studies suggest that the contents of nuts may
and found no association between peanut butter intake and DFS inhibit the growth of a colon cancer cell line69 and may decrease the
(Ptrend = .15), OS (Ptrend = .12), or RFS (Ptrend = .09). rate of colorectal cancer growth and angiogenesis in mice.70
Multiple prior observational studies in patients with colorectal
cancer, including analyses from our study population, suggest that
DISCUSSION states of excess energy balance (eg, obesity, T2D, sedentary lifestyle,
Western-pattern diet, greater glycemic load diet, and high sugar-
In this prospective cohort of patients with stage III colon cancer sweetened beverage intake) are associated with greater risk of
who received adjuvant chemotherapy, a diet with increased total cancer recurrence and mortality.1-3,8 Therefore, we hypothesize
nut intake was associated with a significant improvement in cancer that the effect of nuts on hyperinsulinemia and energy balance
recurrence or mortality (DFS) and all-cause mortality (OS). may, in part, explain the association between nut intake and
Moreover, these associations seemed to be independent of other improved patient outcome in our study. Nonetheless, there may be
predictors of patient outcome, diet, and lifestyle factors, and the other possible mechanisms through which nut intake may influ-
effect of total nut consumption was maintained across other ence survival in patients with colon cancer.
known or suspected risk factors of cancer recurrence. In contrast to There are several strengths of our analysis. Embedding the
tree nuts, we did not observe a significant association with peanuts, cohort within a randomized trial ensures equal distribution of po-
which are legumes, although the statistical power to assess peanut tential confounders. All patient had stage III colon cancer, minimizing
intake was limited. the effect of heterogeneity of the disease stage on the outcomes.
To our knowledge, this is the first study to examine the as- Treatment and patient follow-up were rigorous because they were
sociation between nut intake and colon cancer recurrence and prescribed and monitored by the clinical trial; the outcomes of death
survival. Prior observational cohort studies have explored the and cancer recurrence were prospectively collected through regular
relationship between nut intake and risk of developing colorectal detailed medical examinations during the course of the follow-up
cancer, finding inconsistent results.48,51,52 Other studies have re- period. Furthermore, detailed information about potential con-
ported reduced cancer-related mortality in association with in- founders and effect modifiers was collected prospectively.
creased nut intake49,53-55; however, the attributable mortality However, our study has a few limitations. Nut intake was self-
reductions in colorectal cancer or other individual cancer types reported, potentially subject to measurement errors, and some-
were not described in these studies. what limited in range of intake in this patient population. However,
Our data are consistent with a wealth of existing observational in previous validation studies, nut intake as recorded on our
and clinical trial data reporting health benefits of nut consumption questionnaire showed good concordance with independent dietary
on many chronic diseases, including reductions in the risk of T2D, records.38 Moreover, because dietary data were collected pro-
insulin resistance, metabolic syndrome, cardiovascular disease, and spectively, any misclassification of nut intake would underestimate
all-cause mortality.49,56-65 Nutrients in nuts, such as unsaturated a true association. Given the observational nature of this study, we
fatty acids, high-quality protein, fiber, vitamins, minerals, phy- cannot exclude the possibility that the associations between nut
tochemicals, and other bioactive substances, may confer potential intake and improved outcome are a result of confounding vari-
anticarcinogenic, anti-inflammatory, and antioxidant properties.28-30 ables or that error in the measurement of adjusted confounding
Clinical trials have demonstrated beneficial effects of nuts on variables could result in residual confounding. However, these

1116 © 2018 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Nut Consumption and Stage III Colon Cancer Outcome

Subgroup No. of Patients Ptrend Pinter

Age, years .07


< 60 399 .86
≥ 60 427 < .01
Sex .96
Male 466 .07
Female 360 .23
Treatment .35
FU+LV 414 .04
CPT-11+FU+LV 412 .31
Performance status .44
0 630 .14
1-2 196 .09
No. of positive lymph nodes .65
1-3 (N1) 562 .06
≥ 4 (N2) 264 .27
Body mass index .72
2 197 .32
< 25 kg/m
≥ 25 kg/m2 629 .04
Physcial activity .34
< 9 MET-h/wk 449 .03
≥ 9 MET-h/wk 377 .37
Glycemic load .64
< Median 413 .07
≥ Median 413 .38
Aspirin .20
No 705 .13
Yes 121 .05
Microsatellite .17
Stable 504 .19
Unstable 93 .08
KRAS .71
Wild type 289 .13
Mutant 152 .49
BRAF .29
Wild type 378 .23
Mutant 59 .15
PIK3CA .21
Wild type 370 .66
Mutant 50 .14
COX2 expression .80
Low 287 .38
High 135 .41

0 0.5 1 1.5 2 2.5

HR
Fig 2. Multivariable hazard ratios (HRs) and 95% CIs for cancer recurrence or mortality across strata of various factors. The analyses used five categories of total nut
intake (never, less than one serving per month, one to three servings per month, one serving per week, and two or more servings per week). The forest plot represents the
HRs of the comparison of never nut consumers versus consumers of two or more servings of nuts per week, adjusting for calorie intake, age, sex, depth of invasion through
bowel wall, number of positive lymph nodes, baseline performance status, treatment group, body mass index, physical activity, aspirin use, and glycemic load. P values are
two-sided; Pinter indicates P for interaction between strata and nut intake; Ptrend indicates P for trend across levels of nut intake. COX2, cyclooxygenase-2; CPT-11,
irinotecan; FU, fluorouracil; LV, leucovorin; MET-h/wk, metabolic equivalent task hours per week.

associations persisted even after controlling for known or sus- mortality of cardiovascular disease.71-77 Periodontal disease has
pected predictors of patient outcome. Furthermore, the associa- also been linked with a higher incidence of some solid cancers,78-85
tions remained largely consistent on multiple sensitivity analyses. although studies on the association with colon cancer incidence
Our cohort did not collect data on dentition. Poor dentition have yielded conflicting results79,82,86 and the effect on colon
may influence the intake of foods such as nuts, which are hard and cancer outcomes is largely unknown. Our additional sensitivity
require significant mastication. Observational studies have shown analysis to address this potential confounder, collapsing the two
an association between dental disease and increased incidence and lowest categories into a new control group, yielded consistent

jco.org © 2018 by American Society of Clinical Oncology 1117


Fadelu et al

Table 3. Associations Between Colon Cancer Recurrence and Mortality by Nut Subtype: Tree Nuts and Peanuts
Categories of Intake (servings)
Outcome Never , 1 per Month 1-3 per Month $ 1 per Week Ptrend*
Tree nuts
DFS
No. of events/total No. of patients 96/267 77/287 40/152 25/120
HR (95% CI), adjusted 1† Ref 0.72 (0.53 to 0.97) 0.66 (0.46 to 0.96) 0.53 (0.34 to 0.84) .02
HR (95% CI), adjusted 2‡ Ref 0.68 (0.51 to 0.93) 0.66 (0.45 to 0.96) 0.54 (0.34 to 0.85) .04
RFS
No. of events/total No. of patients 76/267 71/287 32/152 20/120
HR (95% CI), adjusted 1† Ref 0.84 (0.61 to 1.16) 0.67 (0.44 to 1.02) 0.55 (0.33 to 0.91) .03
HR (95% CI), adjusted 2‡ Ref 0.79 (0.57 to 1.10) 0.66 (0.43 to 1.01) 0.56 (0.33 to 0.94) .06
OS
No. of events/total No. of patients 77/267 55/287 28/152 17/120
HR (95% CI), adjusted 1† Ref 0.65 (0.46 to 0.92) 0.61 (0.39 to 0.94) 0.47 (0.27 to 0.80) .02
HR (95% CI), adjusted 2‡ Ref 0.62 (0.44 to 0.88) 0.58 (0.37 to 0.91) 0.47 (0.27 to 0.82) .04
Peanuts
DFS
No. of events/total No. of patients 70/215 78/280 51/183 39/148
HR (95% CI), adjusted 1† Ref 0.84 (0.61 to 1.16) 0.83 (0.58 to 1.19) 0.78 (0.52 to 1.17) .41
HR (95% CI), adjusted 2‡ Ref 0.87 (0.63 to 1.21) 0.85 (0.59 to 1.23) 0.81 (0.53 to 1.23) .46
RFS
No. of events/total No. of patients 55/215 66/280 44/183 34/148
HR (95% CI), adjusted 1† Ref 0.91 (0.64 to 1.30) 0.92 (0.62 to 1.37) 0.90 (0.58 to 1.40) .76
HR (95% CI), adjusted 2‡ Ref 0.94 (0.66 to 1.35) 0.93 (0.62 to 1.39) 0.97 (0.61 to 1.53) .98
OS
No. of events/total No. of patients 57/215 57/280 37/183 26/148
HR (95% CI), adjusted 1† Ref 0.75 (0.52 to 1.08) 0.73 (0.48 to 1.10) 0.62 (0.39 to 1.01) .15
HR (95% CI), adjusted 2‡ Ref 0.77 (0.53 to 1.12) 0.73 (0.48 to 1.11) 0.60 (0.37 to 0.98) .11

Abbreviations: DFS, disease-free survival; HR, hazard ratio; OS, overall survival; Ref, reference; RFS, recurrence-free survival.
*Two-sided P value. Trend across consumption levels with the categorical median.
†Model adjusted 1: Adjusted using Cox proportional hazards regression for calorie intake.
‡Model adjusted 2: Adjusted using Cox proportional hazards regression further for age, sex, depth of invasion through bowel wall, number of positive lymph nodes,
baseline performance status, treatment group, body mass index, physical activity, aspirin use, and glycemic load.

results. Nonetheless, as with many nutritional epidemiology


AUTHOR CONTRIBUTIONS
studies, the potential for residual confounding remains.
Finally, patients in randomized trials may differ from the
Conception and design: Leonard B. Saltz, Robert J. Mayer, Rex B. Mowat,
population at large. However, the distribution of dietary and
Alexander Hantel, Michael Messino, Hedy L. Kindler, Alan Venook, Walter
lifestyle habits reported by our cohort was similar to that of other Willett, Edward Giovannucci, Charles S. Fuchs
US cohorts,87 supporting generalizability of our results. Still, it is Financial support: Shuji Ogino
imperative to replicate our findings in other cohorts. Administrative support: Robert J. Mayer, Charles S. Fuchs
In conclusion, this prospective study of patients with stage III Provision of study materials or patients: Renaud Whittom, Michael
colon cancer suggests that a diet with increased nut consumption is Messino, Hedy L. Kindler
associated with a significant reduction in cancer recurrence and Collection and assembly of data: Renaud Whittom, Daniel M. Atienza,
Shuji Ogino, Charles S. Fuchs
mortality. Although the findings of our observational study do not Data analysis and interpretation: Temidayo Fadelu, Sui Zhang, Donna
establish causality, the results offer further support of the role of Niedzwiecki, Xing Ye, Al B. Benson, Alan Venook, Shuji Ogino, Kimmie
diet and lifestyle as modifiable risk factors of outcomes in patients Ng, Kana Wu, Walter Willett, Edward Giovannucci, Jeffrey Meyerhardt,
with colon cancer. Ying Bao, Charles S. Fuchs
Manuscript writing: All authors
Final approval of manuscript: All authors
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS Accountable for all aspects of the work: All authors
OF INTEREST

Disclosures provided by the authors are available with this article at


jco.org.

Findings from CALGB 89803. J Natl Cancer Inst 104: 3. Meyerhardt JA, Niedzwiecki D, Hollis D, et al:
REFERENCES 1702-1711, 2012 Association of dietary patterns with cancer re-
2. Meyerhardt JA, Heseltine D, Niedzwiecki D, et al: currence and survival in patients with stage III colon
1. Meyerhardt JA, Sato K, Niedzwiecki D, et al: Impact of physical activity on cancer recurrence and cancer. JAMA 298:754-764, 2007
Dietary glycemic load and cancer recurrence and survival in patients with stage III colon cancer: Findings 4. Meyerhardt JA, Niedzwiecki D, Hollis D, et al:
survival in patients with stage III colon cancer: from CALGB 89803. J Clin Oncol 24:3535-3541, 2006 Impact of body mass index and weight change after

1118 © 2018 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Nut Consumption and Stage III Colon Cancer Outcome

treatment on cancer recurrence and survival in pa- 22. O’Neil CE, Fulgoni VL III, Nicklas TA: Tree nut III colon cancer intergroup trial. J Natl Cancer Inst
tients with stage III colon cancer: Findings from consumption is associated with better adiposity 105:1789-1798, 2013
Cancer and Leukemia Group B 89803. J Clin Oncol measures and cardiovascular and metabolic syn- 40. Ogino S, Shima K, Meyerhardt JA, et al:
26:4109-4115, 2008 drome health risk factors in U.S. adults: NHANES Predictive and prognostic roles of BRAF mutation
5. Meyerhardt JA, Giovannucci EL, Holmes MD, 2005-2010. Nutr J 14:64, 2015 in stage III colon cancer: Results from intergroup
et al: Physical activity and survival after colorectal 23. Ros E: Health benefits of nut consumption. trial CALGB 89803. Clin Cancer Res 18:890-900,
cancer diagnosis. J Clin Oncol 24:3527-3534, 2006 Nutrients 2:652-682, 2010 2012
6. Dignam JJ, Polite BN, Yothers G, et al: Body 24. Hernández-Alonso P, Salas-Salvadó J, Baldrich- 41. Ogino S, Kawasaki T, Brahmandam M, et al:
mass index and outcomes in patients who receive Mora M, et al: Beneficial effect of pistachio con- Sensitive sequencing method for KRAS mutation
adjuvant chemotherapy for colon cancer. J Natl sumption on glucose metabolism, insulin resistance, detection by pyrosequencing. J Mol Diagn 7:
Cancer Inst 98:1647-1654, 2006 inflammation, and related metabolic risk markers: A 413-421, 2005
7. Meyerhardt JA, Catalano PJ, Haller DG, et al: randomized clinical trial. Diabetes Care 37:3098-3105, 42. Ogino S, Meyerhardt JA, Irahara N, et al: KRAS
Impact of diabetes mellitus on outcomes in pa- 2014 mutation in stage III colon cancer and clinical out-
tients with colon cancer. J Clin Oncol 21:433-440, 25. Jenkins DJ, Kendall CW, Josse AR, et al: Al- come following intergroup trial CALGB 89803. Clin
2003 monds decrease postprandial glycemia, insulinemia, Cancer Res 15:7322-7329, 2009
8. Fuchs MA, Sato K, Niedzwiecki D, et al: Sugar- and oxidative damage in healthy individuals. J Nutr 43. Warren RS, Atreya CE, Niedzwiecki D, et al:
sweetened beverage intake and cancer recurrence 136:2987-2992, 2006 Association of TP53 mutational status and gender
and survival in CALGB 89803 (Alliance). PLoS One 9: 26. Jiang R, Jacobs DR Jr, Mayer-Davis E, et al: with survival after adjuvant treatment for stage III
e99816, 2014 Nut and seed consumption and inflammatory colon cancer: Results of CALGB 89803. Clin Cancer
9. Wolpin BM, Meyerhardt JA, Chan AT, et al: markers in the multi-ethnic study of atherosclerosis. Res 19:5777-5787, 2013
Insulin, the insulin-like growth factor axis, and mor- Am J Epidemiol 163:222-231, 2006 44. Bertagnolli MM, Niedzwiecki D, Compton CC,
tality in patients with nonmetastatic colorectal can- 27. Sweazea KL, Johnston CS, Ricklefs KD, et al: et al: Microsatellite instability predicts improved re-
cer. J Clin Oncol 27:176-185, 2009 Almond supplementation in the absence of dietary sponse to adjuvant therapy with irinotecan, fluoro-
10. Bhupathiraju SN, Pan A, Manson JE, et al: advice significantly reduces C-reactive protein in uracil, and leucovorin in stage III colon cancer: Cancer
Changes in coffee intake and subsequent risk of type subjects with type 2 diabetes. J Funct Foods 10: and Leukemia Group B protocol 89803. J Clin Oncol
2 diabetes: Three large cohorts of US men and 252-259, 2014 27:1814-1821, 2009
women. Diabetologia 57:1346-1354, 2014 28. Kris-Etherton PM, Hu FB, Ros E, et al: The role 45. Chan AT, Ogino S, Fuchs CS: Aspirin and the
11. Ding M, Bhupathiraju SN, Chen M, et al: of tree nuts and peanuts in the prevention of coronary risk of colorectal cancer in relation to the ex-
Caffeinated and decaffeinated coffee consumption heart disease: Multiple potential mechanisms. J Nutr pression of COX-2. N Engl J Med 356:2131-2142,
and risk of type 2 diabetes: A systematic review and 138:1746S-1751S, 2008 2007
a dose-response meta-analysis. Diabetes Care 37: 29. González CA, Salas-Salvadó J: The potential of 46. Bao Y, Hu FB, Giovannucci EL, et al: Nut
569-586, 2014 nuts in the prevention of cancer. Br J Nutr 96: consumption and risk of pancreatic cancer in women.
12. Bhupathiraju SN, Pan A, Malik VS, et al: Caf- S87-S94, 2006 (suppl 2) Br J Cancer 109:2911-2916, 2013
feinated and caffeine-free beverages and risk of type 30. Pfundstein B, Haubner R, Würtele G, et al: 47. Wang W, Yang M, Kenfield SA, et al: Nut
2 diabetes. Am J Clin Nutr 97:155-166, 2013 Pilot walnut intervention study of urolithin bio- consumption and prostate cancer risk and mortality.
13. van Dam RM, Hu FB: Coffee consumption and availability in human volunteers. J Agric Food Chem Br J Cancer 115:371-374, 2016
risk of type 2 diabetes: A systematic review. JAMA 62:10264-10273, 2014 48. Yang M, Hu FB, Giovannucci EL, et al: Nut
294:97-104, 2005 31. Saltz LB, Niedzwiecki D, Hollis D, et al: Irino- consumption and risk of colorectal cancer in women.
14. Salazar-Martinez E, Willett WC, Ascherio A, tecan fluorouracil plus leucovorin is not superior to Eur J Clin Nutr 70:333-337, 2016
et al: Coffee consumption and risk for type 2 diabetes fluorouracil plus leucovorin alone as adjuvant treat- 49. Bao Y, Han J, Hu FB, et al: Association of nut
mellitus. Ann Intern Med 140:1-8, 2004 ment for stage III colon cancer: Results of CALGB consumption with total and cause-specific mortality.
15. Wu T, Willett WC, Hankinson SE, et al: Caf- 89803. J Clin Oncol 25:3456-3461, 2007 N Engl J Med 369:2001-2011, 2013
feinated coffee, decaffeinated coffee, and caffeine in 32. Zubrod CG, Schneiderman M, Frei E, et al: 50. Cox DR: Regression models and life-tables.
relation to plasma C-peptide levels, a marker of in- Appraisal of methods for the study of chemotherapy J R Stat Soc B 34:187-220, 1972
sulin secretion, in U.S. women. Diabetes Care 28: of cancer in man: Comparative therapeutic trial of 51. Yeh C-C, You S-L, Chen C-J, et al: Peanut
1390-1396, 2005 nitrogen mustard and triethylene thiophosphor- consumption and reduced risk of colorectal cancer in
16. Fung TT, Hu FB, Schulze M, et al: A dietary amide. J Chronic Dis 11:7-33, 1960 women: A prospective study in Taiwan. World J
pattern that is associated with C-peptide and risk of 33. Sturrock PR, Maykel JA: Postoperative Gastroenterol 12:222-227, 2006
colorectal cancer in women. Cancer Causes Control management, in colorectal surgery. https://www- 52. Jenab M, Ferrari P, Slimani N, et al: Associa-
23:959-965, 2012 clinicalkey-com.ezp-prod1.hul.harvard.edu/#!/content/ tion of nut and seed intake with colorectal cancer risk
17. Wedick NM, Brennan AM, Sun Q, et al: Effects book/3-s2.0-B9781437717242000040?scrollTo=% in the European Prospective Investigation into Can-
of caffeinated and decaffeinated coffee on biological 23hl0000956 cer and Nutrition. Cancer Epidemiol Biomarkers Prev
risk factors for type 2 diabetes: A randomized con- 34. Willett WC, Reynolds RD, Cottrell-Hoehner S, 13:1595-1603, 2004
trolled trial. Nutr J 10:93, 2011 et al: Validation of a semi-quantitative food frequency 53. Guasch-Ferré M, Bulló M, Martı́nez-González
18. Williams CJ, Fargnoli JL, Hwang JJ, et al: questionnaire: Comparison with a 1-year diet record. MÁ, et al: Frequency of nut consumption and mor-
Coffee consumption is associated with higher J Am Diet Assoc 87:43-47, 1987 tality risk in the PREDIMED nutrition intervention trial.
plasma adiponectin concentrations in women with or 35. Willett WC, Sampson L, Stampfer MJ, et al: BMC Med 11:164, 2013
without type 2 diabetes: A prospective cohort study. Reproducibility and validity of a semiquantitative food 54. Fernández-Montero A, Bes-Rastrollo M, Barrio-
Diabetes Care 31:504-507, 2008 frequency questionnaire. Am J Epidemiol 122:51-65, López MT, et al: Nut consumption and 5-y all-cause
19. Guercio BJ, Sato K, Niedzwiecki D, et al: 1985 mortality in a Mediterranean cohort: The SUN project.
Coffee intake, recurrence, and mortality in stage III 36. US Department of Agriculture: Composition of Nutrition 30:1022-1027, 2014
colon cancer: Results from CALGB 89803 (Alliance). Foods: Raw, Processed, and Prepared, 1963-1988. 55. Bonaccio M, Di Castelnuovo A, De Curtis A,
J Clin Oncol 33:3598-3607, 2015 Washington, DC, Department of Agriculture, US et al: Nut consumption is inversely associated with
20. Casas-Agustench P, López-Uriarte P, Bulló M, Government Printing Office, 1989 both cancer and total mortality in a Mediterranean
et al: Effects of one serving of mixed nuts on serum 37. Willett W, Stampfer MJ: Total energy intake: population: Prospective results from the Moli-sani
lipids, insulin resistance and inflammatory markers in Implications for epidemiologic analyses. Am J Epi- study. Br J Nutr 114:804-811, 2015
patients with the metabolic syndrome. Nutr Metab demiol 124:17-27, 1986 56. Jiang R, Manson JE, Stampfer MJ, et al: Nut
Cardiovasc Dis 21:126-135, 2011 38. Salvini S, Hunter DJ, Sampson L, et al: Food- and peanut butter consumption and risk of type 2
21. Holligan SD, West SG, Gebauer SK, et al: A based validation of a dietary questionnaire: The ef- diabetes in women. JAMA 288:2554-2560, 2002
moderate-fat diet containing pistachios improves fects of week-to-week variation in food consumption. 57. Villegas R, Gao Y-T, Yang G, et al: Legume and
emerging markers of cardiometabolic syndrome in Int J Epidemiol 18:858-867, 1989 soy food intake and the incidence of type 2 diabetes
healthy adults with elevated LDL levels. Br J Nutr 39. Ogino S, Liao X, Imamura Y, et al: Predictive in the Shanghai Women’s Health Study. Am J Clin
112:744-752, 2014 and prognostic analysis of PIK3CA mutation in stage Nutr 87:162-167, 2008

jco.org © 2018 by American Society of Clinical Oncology 1119


Fadelu et al

58. Pan A, Sun Q, Manson JE, et al: Walnut 68. Tapsell LC, Batterham MJ, Teuss G, et al: in the Glasgow Alumni Cohort. Heart 93:1098-1103,
consumption is associated with lower risk of type 2 Long-term effects of increased dietary poly- 2007
diabetes in women. J Nutr 143:512-518, 2013 unsaturated fat from walnuts on metabolic parame- 78. Migliorati CA: Periodontal diseases and can-
59. Fernández-Montero A, Bes-Rastrollo M, ters in type II diabetes. Eur J Clin Nutr 63:1008-1015, cer. Lancet Oncol 9:510-512, 2008
Beunza JJ, et al: Nut consumption and incidence of 2009 79. Michaud DS, Liu Y, Meyer M, et al: Periodontal
metabolic syndrome after 6-year follow-up: The SUN 69. Lux S, Scharlau D, Schlörmann W, et al: In vitro disease, tooth loss, and cancer risk in male health
(Seguimiento Universidad de Navarra, University of fermented nuts exhibit chemopreventive effects in professionals: A prospective cohort study. Lancet
Navarra Follow-up) cohort. Public Health Nutr 16: HT29 colon cancer cells. Br J Nutr 108:1177-1186, Oncol 9:550-558, 2008
2064-2072, 2013 2012 80. Abnet CC, Qiao Y-L, Mark SD, et al: Pro-
60. Singh PN, Fraser GE: Dietary risk factors for 70. Nagel JM, Brinkoetter M, Magkos F, et al: spective study of tooth loss and incident esophageal
colon cancer in a low-risk population. Am J Epidemiol Dietary walnuts inhibit colorectal cancer growth in and gastric cancers in China. Cancer Causes Control
148:761-774, 1998 mice by suppressing angiogenesis. Nutrition 28: 12:847-854, 2001
61. Djoussé L, Rudich T, Gaziano JM: Nut con- 67-75, 2012 81. Garrote LF, Herrero R, Reyes RMO, et al: Risk
sumption and risk of hypertension in US male phy- 71. DeStefano F, Anda RF, Kahn HS, et al: Dental factors for cancer of the oral cavity and oro-pharynx in
sicians. Clin Nutr 28:10-14, 2009 disease and risk of coronary heart disease and Cuba. Br J Cancer 85:46-54, 2001
62. Tsai C-J, Leitzmann MF, Hu FB, et al: A pro- mortality. BMJ 306:688-691, 1993 82. Hiraki A, Matsuo K, Suzuki T, et al: Teeth loss and
spective cohort study of nut consumption and the 72. Joshipura KJ, Hung H-C, Rimm EB, et al: risk of cancer at 14 common sites in Japanese. Cancer
risk of gallstone disease in men. Am J Epidemiol 160: Periodontal disease, tooth loss, and incidence of is- Epidemiol Biomarkers Prev 17:1222-1227, 2008
961-968, 2004 chemic stroke. Stroke 34:47-52, 2003 83. Abnet CC, Kamangar F, Dawsey SM, et al:
63. Tsai C-J, Leitzmann MF, Hu FB, et al: Frequent 73. Jimenez M, Krall EA, Garcia RI, et al: Peri- Tooth loss is associated with increased risk of gastric
nut consumption and decreased risk of cholecys- odontitis and incidence of cerebrovascular disease in non-cardia adenocarcinoma in a cohort of Finnish
tectomy in women. Am J Clin Nutr 80:76-81, 2004 men. Ann Neurol 66:505-512, 2009 smokers. Scand J Gastroenterol 40:681-687, 2005
64. Strate LL, Liu YL, Syngal S, et al: Nut, corn, and 74. Watt RG, Tsakos G, de Oliveira C, et al: Tooth 84. Boylan MR, Khalili H, Huang ES, et al: A pro-
popcorn consumption and the incidence of di- loss and cardiovascular disease mortality risk: Re- spective study of periodontal disease and risk of
verticular disease. JAMA 300:907-914, 2008 sults from the Scottish Health Survey. PLoS One 7: gastric and duodenal ulcer in male health pro-
65. Gopinath B, Buyken AE, Flood VM, et al: e30797, 2012 fessionals. Clin Transl Gastroenterol 5:e49, 2014
Consumption of polyunsaturated fatty acids, fish, and 75. Desvarieux M, Demmer RT, Rundek T, et al: 85. Hujoel PP, Drangsholt M, Spiekerman C, et al:
nuts and risk of inflammatory disease mortality. Am J Relationship between periodontal disease, tooth An exploration of the periodontitis-cancer associa-
Clin Nutr 93:1073-1079, 2011 loss, and carotid artery plaque: The Oral Infections tion. Ann Epidemiol 13:312-316, 2003
66. Torabian S, Haddad E, Rajaram S, et al: Acute and Vascular Disease Epidemiology Study (INVEST). 86. Momen-Heravi F, Babic A, Tworoger SS, et al:
effect of nut consumption on plasma total poly- Stroke 34:2120-2125, 2003 Periodontal disease, tooth loss and colorectal cancer
phenols, antioxidant capacity and lipid peroxidation. 76. Dietrich T, Jimenez M, Krall Kaye EA, et al: risk: Results from the Nurses’ Health Study. Int J
J Hum Nutr Diet 22:64-71, 2009 Age-dependent associations between chronic Cancer 140:646-652, 2017
67. Ma Y, Njike VY, Millet J, et al: Effects of walnut periodontitis/edentulism and risk of coronary heart 87. Michaud DS, Fuchs CS, Liu S, et al: Dietary
consumption on endothelial function in type 2 di- disease. Circulation 117:1668-1674, 2008 glycemic load, carbohydrate, sugar, and colorectal
abetic subjects: A randomized controlled crossover 77. Tu Y-K, Galobardes B, Smith GD, et al: As- cancer risk in men and women. Cancer Epidemiol
trial. Diabetes Care 33:227-232, 2010 sociations between tooth loss and mortality patterns Biomarkers Prev 14:138-147, 2005

Affiliations
Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/
Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health;
Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA; Donna
Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology
Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B.
Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal,
Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive
Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza,
Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San
Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT.
Support
Supported by the National Cancer Institute of the National Institutes of Health under Awards No. U10CA180821 and U10CA180882
(to Alliance for Clinical Trials in Oncology), U10CA180820 (to Eastern Cooperative Oncology Group–American College of Radiology
Imaging Network), and U10CA180888 (to Southwest Oncology Group). Also supported by Grants No. U10CA60138, U10CA138561,
U10CA180791, U10CA180836, U10CA180867, and UG1CA189858, and the Pharmacia & Upjohn Company, now Pfizer Oncology. C.S.F.,
K.N., J.M., D.N., and X.Y. are supported in part by grants from the National Cancer Institute (Grants No. K07 CA148894, R01 CA118553,
R01 CA149222, R01 CA169141, and P50 CA127003). Y.B. is supported in part by Grant No. P30 DK046200 from the National Institutes of
Health and the International Tree Nut Council Nutrition Research and Education Foundation. S.O. is supported in part by Grant No. R35
CA197735 from the National Institutes of Health.

nnn

1120 © 2018 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Nut Consumption and Stage III Colon Cancer Outcome

AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST


Nut Consumption and Survival in Patients With Stage III Colon Cancer: Results From CALGB 89803 (Alliance)
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are
self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more
information about ASCO’s conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/site/ifc.
Temidayo Fadelu Daniel M. Atienza
No relationship to disclose Employment: Virginia Oncology Associates
Sui Zhang Michael Messino
No relationship to disclose No relationship to disclose
Donna Niedzwiecki Hedy L. Kindler
No relationship to disclose Consulting or Advisory Role: Aduro Biotech, MedImmune, Bayer,
Genentech, Verastem, Celgene, GlaxoSmithKline, AstraZeneca, Merck
Xing Ye Research Funding: Aduro Biotech, AstraZeneca, Bayer, GlaxoSmithKline,
No relationship to disclose Merck, MedImmune, Verastem, Bristol-Myers Squibb, Eli Lilly
Leonard B. Saltz Expert Testimony: Aduro Biotech
Consulting or Advisory Role: Eli Lilly, McNeil PPC (I), Abbvie Alan Venook
Research Funding: Taiho Pharmaceutical Consulting or Advisory Role: Genentech
Robert J. Mayer Research Funding: Genentech (Inst), Bristol-Myers Squibb (Inst)
Honoraria: Taiho Pharmaceutical Patents, Royalties, Other Intellectual Property: Royalties from Now-
Consulting or Advisory Role: CASI Pharmaceuticals UptoDate for authoring and maintaining two chapters
Travel, Accommodations, Expenses: Genentech, Taiho Pharmaceutical,
Rex B. Mowat Roche
No relationship to disclose
Shuji Ogino
Renaud Whittom No relationship to disclose
Honoraria: AstraZeneca
Consulting or Advisory Role: Boehringer Ingelheim, AstraZeneca, Pfizer, Kimmie Ng
Bristol-Myers Squibb Consulting or Advisory Role: Bayer, Tarrex, Genentech, Eli Lilly
Research Funding: Astellas (Inst), Genentech (Inst) Research Funding: Gilead Sciences, Tarrex, Genentech (Inst)
Travel, Accommodations, Expenses: Seattle Genetics, Celgene Kana Wu
Alexander Hantel No relationship to disclose
Travel, Accommodations, Expenses: Genentech Walter Willett
Al B. Benson No relationship to disclose
Consulting or Advisory Role: Genentech, Bristol-Myers Squibb, Merck Edward Giovannucci
Serono, Celgene, Vicus Therapeutics, Pharmacyclics, Precision No relationship to disclose
Therapeutics, Taiho Pharmaceutical, Bayer, Alchemia, Infinity
Pharmaceuticals, Boehringer Ingelheim, Astellas Pharma, EMD Serono, Jeffrey Meyerhardt
IntegraGen, Guardant Health, Opsona Therapeutics, Lexicon, Novartis, Honoraria: Chugai Pharma
Boston Biomedical, Helsinn, Guerbet, Eli Lilly, Spectrum Pharmaceuticals, Consulting or Advisory Role: Genentech (Inst)
Advanced Accelerator Applications, Bayer/Onyx, TRM Oncology, DAVA
Pharmaceuticals, Sanofi, Eli Lilly/ImClone Ying Bao
Research Funding: Gilead Sciences (Inst), Amgen (Inst), Astellas Pharma No relationship to disclose
(Inst), Advanced Accelerator Applications (Inst), Bayer/Onyx (Inst), Charles S. Fuchs
Novartis (Inst), Alchemia (Inst), AVEO (Inst), Infinity Pharmaceuticals Leadership: CytomX Therapeutics
(Inst), Genentech (Inst), Merck Sharp & Dohme (Inst), Taiho Consulting or Advisory Role: Eli Lilly, Sanofi, Merck, Entrinsic Health,
Pharmaceutical (Inst) Five Prime Therapeutics, Agios, Gilead Sciences, Merrimack, Taiho
Travel, Accommodations, Expenses: Genentech, Eli Lilly/ImClone, Bayer, Pharmaceutical, KEW Group, Genentech
Sanofi, Spectrum Pharmaceuticals, AVEO, Gilead Sciences, Astellas
Pharma, Boehringer Ingelheim, DAVA Oncology, TRM Oncology,
Guardant Health, Helsinn, Guerbet, Boston Biomedical

jco.org © 2018 by American Society of Clinical Oncology


Fadelu et al

Appendix

Methods
In supplemental analyses, we offer additional multivariable models using Cox proportional hazards regression, whereby
additional covariates were added as part of sensitivity analyses. In successive models, Western and prudent dietary patterns (model 3)
and race (as a categorical variable: white, black, or other), smoking as binary (ever or never), and alcohol (as a continuous variable in
grams per day; model 4) were added to the final model that included age, sex, depth of invasion through bowel wall, number of positive
lymph nodes, baseline performance status, treatment group, body mass index (calculated as weight in kilograms divided by height in
meters squared), physical activity (measured in metabolic equivalent task hours per week), aspirin use, and glycemic load. Glycemic
load and Western and prudent dietary patterns were determined by methods previously described by our study cohort (Wu K, et al:
Cancer Causes Control 15:853-862, 2004; Fung T, et al: Arch Intern Med 163:309-314, 2003).1,3
Because of possible dietary modifications immediately after colectomy, we a priori limited the primary analysis to nut intake
reported on questionnaire 2 (Q2). In secondary analyses, we repeated the models using cumulative nut intake from questionnaire 1
(Q1) and Q2. Cumulative nut exposure average was calculated based on weighting proportional to the time between Q1 and Q2,
and then the time between Q2 and the outcome survival period, using previously published methods (Hu FB, et al: Am J Epidemiol
149:531-540, 1999).1 In this analysis, the follow-up period began from the completion of Q1. Consistent with previous analyses in
our study cohort, patients were excluded if they experienced cancer recurrence or death within 90 days of completing Q1 to prevent
misattribution bias.1,8,18 Survival estimate curves for disease-free survival, recurrence-free survival, and overall survival by nut
intake categories were calculated using the Kaplan-Meier methods, and the log-rank testing was used to compare survival between
groups (Kaplan EL, Meier P: J Am Stat Assoc 53:457-481, 1958).

1.0 +
Censor
0.9 Log-rank P = .0377

0.8
0.7
0.6
DFS

0.5
0.4 Nuts
Never
0.3 < 1 per month
0.2 1-3 per month
1 per week
0.1
≥ 2 per week

0 1 2 3 4 5 6 7 8
No. at risk Time (years)
Never 145 124 112 100 91 80 66 19 1
< 1 per month 98 87 70 68 62 55 46 4 0
1-3 per month 211 184 165 152 144 138 115 28 0
1 per week 214 196 174 166 150 139 117 22 0
≥ 2 per week 158 137 128 126 121 108 97 25 1

Fig A1. Kaplan-Meier curves for disease-free survival (DFS) by total nut con-
sumption. Curves depict survival for five categories of total nut intake (never, less
than one serving per month, one to three servings per month, one serving per
week, and two or more servings per week).

© 2018 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


Nut Consumption and Stage III Colon Cancer Outcome

1.0 +
Censor
0.9 Log-rank P = .1782

0.8
0.7
0.6

RFS
0.5
0.4 Nuts
Never
0.3
< 1 per month
0.2 1-3 per month
1 per week
0.1
≥ 2 per week

0 1 2 3 4 5 6 7 8

No. at risk Time (years)


Never 145 124 112 100 91 80 66 19 1
< 1 per month 98 87 70 68 62 55 46 4 0
1-3 per month 211 184 165 152 144 138 115 28 0
1 per week 214 196 174 165 149 139 117 22 0
≥ 2 per week 158 137 128 126 121 108 97 25 1

Fig A2. Kaplan-Meier curves for recurrence-free survival (RFS) by total nut
consumption. Curves depict survival for five categories of total nut intake (never,
less than one serving per month, one to three servings per month, one serving per
week, and two or more servings per week).

+
Censor
1.0 Log-rank P = .0052
0.9
0.8
0.7
0.6
OS

0.5
0.4 Nuts
Never
0.3
< 1 per month
0.2 1-3 per month
0.1 1 per week
≥ 2 per week

0 1 2 3 4 5 6 7 8

No. at risk Time (years)


Never 145 140 127 118 106 89 71 21 1
< 1 per month 98 96 87 81 76 67 53 6 0
1-3 per month 211 203 186 173 162 151 124 31 1
1 per week 214 208 198 185 169 155 125 27 1
≥ 2 per week 158 151 147 141 137 122 105 25 1

Fig A3. Kaplan-Meier curves for overall survival (OS) by total nut consumption. Curves
depict survival for five categories of total nut intake (never, less than one serving per
month, one to three servings per month, one serving per week, and two or more
servings per week).

jco.org © 2018 by American Society of Clinical Oncology


Fadelu et al

Table A1. Sensitivity Analyses Further Controlling for Additional Confounders: Dietary Pattern, Race, Smoking, and Alcohol Use
Total Nut Consumption Categories of Intake (servings)
Outcome Never , 1 per Month 1-3 per Month 1 per Week $ 2 per Week Ptrend*
DFS
No. of events/total No. of patients 52/145 36/98 56/211 58/214 36/158
HR (95% CI), model 3† Ref 1.03 (0.67 to 1.59) 0.71 (0.48 to 1.05) 0.71 (0.48 to 1.04) 0.60 (0.38 to 0.95) .05
HR (95% CI), model 4‡ Ref 1.02 (0.66 to 1.57) 0.69 (0.47 to 1.02) 0.69 (0.47 to 1.02) 0.59 (0.37 to 0.93) .05
RFS
No. of events/total No. of patients 39/145 31/98 51/211 47/214 31/158
HR (95% CI), model 3† Ref 1.15 (0.71 to 1.86) 0.84 (0.55 to 1.29) 0.76 (0.49 to 1.18) 0.72 (0.43 to 1.20) .18
HR (95% CI), model 4‡ Ref 1.13 (0.70 to 1.82) 0.82 (0.53 to 1.25) 0.75 (0.48 to 1.16) 0.70 (0.42 to 1.17) .16
OS
No. of events/total No. of patients 44/145 27/98 43/211 39/214 24/158
HR (95% CI), model 3† Ref 0.94 (0.58 to 1.54) 0.65 (0.42 to 0.99) 0.55 (0.35 to 0.85) 0.45 (0.26 to 0.78) .01
HR (95% CI), model 4‡ Ref 0.92 (0.56 to 1.50) 0.64 (0.41 to 0.98) 0.54 (0.34 to 0.85) 0.46 (0.26 to 0.78) .01

Abbreviations: BMI, body mass index; DFS, disease-free survival; HR, hazard ratio; OS, overall survival; Q2, questionnaire 2; Ref, reference; RFS, recurrence-free
survival.
*Two-sided P value. Trend across consumption levels with the categorical median.
†Model 3: Cox proportional hazards regression adjusting for age, sex, depth of invasion through bowel wall, number of positive lymph nodes, baseline performance
status, treatment group, and aspirin use (Q2), BMI (Q2), physical activity (Q2), dietary glycemic load (Q2), calorie intake (Q2), Western dietary pattern (Q2), and prudent
dietary pattern.
‡Model 4: Cox proportional hazards regression adjusting for age, sex, depth of invasion through bowel wall, number of positive lymph nodes, baseline performance
status, treatment group, and aspirin use (Q2), BMI (Q2), physical activity (Q2), glycemic load (Q2), and calorie intake (Q2), plus race (white, black, or other), smoking (never
or ever), and alcohol (grams per day, Q2).

Table A2. Consumption of Nuts on Q1 and Q2 in Servings per Day


Nut Type Q1 Mean (95% CI) Q1 Median Q2 Mean (95% CI) Q2 Median P
Peanuts 0.09 (0.08 to 0.11) 0.03 0.10 (0.09 to 0.11) 0.03 , .001
Tree nuts 0.07 (0.05 to 0.08) 0.03 0.08 (0.07 to 0.09) 0.03 , .001
Peanut butter 0.28 (0.25 to 0.32) 0.07 0.23 (0.21 to 0.26) 0.07 , .001

NOTE. P value generated from the paired Wilcoxon test for non-normality.
Abbreviations: Q1, questionnaire 1 (midway through adjuvant therapy); Q2, questionnaire 2 (6 months after completion of adjuvant therapy).

Table A3. Association Between Colon Cancer Recurrence and Mortality by Cumulative Average Total Nut Consumption of Q1 and Q2
Total Nut Consumption Categories of Intake (servings)
Outcome Never , 1 per Month 1-3 per Month 1 per Week $ 2 per Week Ptrend*
DFS
No. of events/total No. of patients 89/152 43/117 99/260 81/251 81/236
HR (95% CI), model 1† Ref 0.49 (0.34 to 0.70) 0.51 (0.39 to 0.69) 0.41 (0.30 to 0.56) 0.44 (0.32 to 0.60) .008
HR (95% CI), model 2‡ Ref 0.53 (0.36 to 0.76) 0.51 (0.38 to 0.68) 0.41 (0.30 to 0.56) 0.45 (0.33 to 0.62) .01
RFS
No. of events/total No. of patients 78/152 39/117 90/260 71/251 71/236
HR (95% CI), model 1† Ref 0.52 (0.35 to 0.76) 0.55 (0.40 to 0.74) 0.42 (0.30 to 0.58) 0.45 (0.33 to 0.63) .01
HR (95% CI), model 2‡ Ref 0.54 (0.36 to 0.79) 0.53 (0.39 to 0.73) 0.42 (0.30 to 0.58) 0.46 (0.32 to 0.64) .02
OS
No. of events/total No. of patients 74/152 32/117 78/260 62/251 64/236
HR (95% CI), model 1† Ref 0.44 (0.29 to 0.66) 0.49 (0.36 to 0.68) 0.38 (0.27 to 0.54) 0.43 (0.30 to 0.60) .02
HR (95% CI), model 2‡ Ref 0.48 (0.32 to 0.73) 0.48 (0.34 to 0.66) 0.38 (0.26 to 0.53) 0.43 (0.30 to 0.61) .03

NOTE. Survival time starts at Q1, and patients with events within 90 days of Q1 were excluded.
Abbreviations: DFS, disease-free survival; HR, hazard ratio; OS, overall survival; Q1, questionnaire 1 (midway through adjuvant therapy); Q2, questionnaire 2 (6 months
after completion of adjuvant therapy); Ref, reference; RFS, recurrence-free survival.
*Two-sided P value. Trend across consumption levels with the categorical median.
†Model 1: Cox proportional hazards regression adjusting for time-varying calorie consumption.
‡Model 2: Cox proportional hazards regression adjusting for age, sex, depth of invasion through bowel wall, number of positive lymph nodes, baseline performance
status, treatment group, regular aspirin use (both Q1 and Q2), and time-varying calorie consumption, body mass index, physical activity, and glycemic load.

© 2018 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY


EUROPEAN UROLOGY 70 (2016) 974–982

available at www.sciencedirect.com
journal homepage: www.europeanurology.com

Platinum Priority – Prostate Cancer


Editorial by Sarah K. Holt, George R. Schade and John L. Gore on pp. 983–984 of this issue

Ejaculation Frequency and Risk of Prostate Cancer:


Updated Results with an Additional Decade of Follow-up

Jennifer R. Ridera,b,1,*, Kathryn M. Wilson a,c,1, Jennifer A. Sinnott a,d, Rachel S. Kelly c,
Lorelei A. Mucci a,c, Edward L. Giovannucci a,c,e
a
Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; b Department of Epidemiology, Boston University School of Public
Health, Boston, MA, USA; c Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School,
Boston, MA, USA; d Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; e Department of Nutrition, Harvard T.H. Chan
School of Public Health, Boston, MA, USA

Article info Abstract

Article history: Background: Evidence suggests that ejaculation frequency may be inversely related to the risk
Accepted March 16, 2016 of prostate cancer (PCa), a disease for which few modifiable risk factors have been identified.
Objective: To incorporate an additional 10 yr of follow-up into an original analysis and to
comprehensively evaluate the association between ejaculation frequency and PCa, accounting for
Associate Editor:
screening, clinically relevant disease subgroups, and the impact of mortality from other causes.
Matthew Cooperberg Design, setting, and participants: A prospective cohort study of participants in the Health
Professionals Follow-up Study utilizing self-reported data on average monthly ejaculation
frequency. The study includes 31 925 men who answered questions on ejaculation frequency
Keywords:
on a 1992 questionnaire and followed through to 2010. The average monthly ejaculation
Epidemiology frequency was assessed at three time points: age 20–29 yr, age 40–49 yr, and the year before
Ejaculation questionnaire distribution.
Behavioral risk factors Outcome measurements and statistical analysis: Incidence of total PCa and clinically relevant
disease subgroups. Cox models were used to estimate hazard ratios (HRs) and 95% confidence
intervals (CIs).
Results and limitations: During 480 831 person-years, 3839 men were diagnosed with PCa.
Ejaculation frequency at age 40–49 yr was positively associated with age-standardized body
mass index, physical activity, divorce, history of sexually transmitted infections, and consump-
tion of total calories and alcohol. Prostate-specific antigen (PSA) test utilization by 2008, number
of PSA tests, and frequency of prostate biopsy were similar across frequency categories. In
multivariable analyses, the hazard ratio for PCa incidence for 21 compared to 4–7 ejaculations
per month was 0.81 (95% confidence interval [CI] 0.72–0.92; p < 0.0001 for trend) for frequency
at age 20–29 yr and 0.78 (95% CI 0.69–0.89; p < 0.0001 for trend) for frequency at age 40–49 yr.
Associations were driven by low-risk disease, were similar when restricted to a PSA-screened
cohort, and were unlikely to be explained by competing causes of death.
Conclusions: These findings provide additional evidence of a beneficial role of more frequent
ejaculation throughout adult life in the etiology of PCa, particularly for low-risk disease.
Patient summary: We evaluated whether ejaculation frequency throughout adulthood is
related to prostate cancer risk in a large US-based study. We found that men reporting higher
compared to lower ejaculatory frequency in adulthood were less likely to be subsequently
diagnosed with prostate cancer.
# 2016 European Association of Urology. Published by Elsevier B.V. All rights reserved.

1
These authors contributed equally to this work.
* Corresponding author. Department of Epidemiology, Boston University School of Public Health,
715 Albany Street, Boston, MA 02118, USA. Tel. +1 617 6385035; Fax: +1 617 5667805.
E-mail address: rider@bu.edu (J.R. Rider).
http://dx.doi.org/10.1016/j.eururo.2016.03.027
0302-2838/# 2016 European Association of Urology. Published by Elsevier B.V. All rights reserved.
EUROPEAN UROLOGY 70 (2016) 974–982 975

1. Introduction leaving 41 201 men. Of these, 9276 did not complete all three questions
on ejaculation frequency, leaving 31 925 men in the study population for
Prostate cancer (PCa) accounts for approximately 15% of all the current analysis. Nonresponders who provided information on
weight, physical activity, and diet appeared to be similar to the
new cancer diagnoses among men worldwide, and the
responders. Among participants who were alive in 2010, follow-up was
burden of disease continues to increase globally [1]. While
96% complete. All participants provided informed consent and the study
diet and physical activity may provide some promise for
was approved by the human subjects committee of the Harvard T.H.
secondary prevention [2–5], there are no evidence-based Chan School of Public Health, Boston.
recommendations to offer healthy adult men to reduce PCa
risk. The few established disease risk factors—age, race, 2.2. Exposure and covariate assessment
family history, and germline polymorphisms—are not
modifiable [6]. In 1992, participants were asked the following question: ‘‘On average,
Sexual behaviors represent potential modifiable risk how many ejaculations did you have per month during these ages?: ages
factors and may influence PCa development through a 20–29; ages 40–49; past year.’’ The frequency at each time point was
variety of specific mechanisms. One biological mechanism reported in the categories none, 1–3, 4–7, 8–12, 13–20, and >20 EPM. To
involves prostatic accumulation of potentially carcinogenic limit the burden for participants and because the question was designed
secretions, which may create more opportunity for PCa specifically to address the prostate stagnation hypothesis, no informa-
tion on the specific type of activity leading to ejaculation was requested.
development, sometimes referred to as the prostate
Information on potential confounders was ascertained in the 1992 ques-
stagnation hypothesis [7,8]. On the basis of this premise, a
tionnaire and most were updated on the biennial questionnaires
prospective report from the Health Professionals Follow-up
throughout follow-up. PSA testing was first assessed in the 1994 ques-
Study (HPFS) cohort published in 2004 found a statistically tionnaire; starting in 1994, men were also asked if they had an elevated
significant inverse association between monthly ejacula- PSA level and whether they had undergone a prostate biopsy or rectal
tion frequency and PCa risk based on 8 yr of follow-up [8]. ultrasound.
Compared to men reporting an average of 4–7 ejaculations
per month (EPM), the risk of PCa among men reporting 21 2.3. Outcome assessment
EPM in middle age was 50% lower. Although these initial
findings were intriguing, the strongest reduction in risk was For men reporting a diagnosis of PCa, we retrieved medical records and
noted for ejaculation frequency in the time period pathology reports to confirm the diagnosis and to obtain information on
immediately before questionnaire administration, raising age at diagnosis, PSA level, and tumor stage and grade. Cases were
concerns about the potential influence of undiagnosed PCa followed through biennial questionnaires to collect information on the
clinical course, including the development of metastases and treatments.
on the results.
Deaths were ascertained through repeated mailings and telephone calls
To confirm and build on these results [8], we conducted
to participants, as well as periodic searches of the National Death Index.
an updated study within the HPFS cohort with an additional
Cause of death was assigned following a review of death certificates,
10 yr of follow-up and 3839 PCa cases, more than double the information from the family, and medical records.
number included in the original report. This updated Total PCa incidence was the primary endpoint of interest. Men
analysis allows us to address possible reverse causation, diagnosed with stage T1a cancers were excluded from analyses. To
investigate the potential impact of PSA screening, and determine whether the association between ejaculation frequency and
determine whether the association between ejaculation PCa differed according to the clinical disease characteristics, we also used
frequency and PCa differed according to the clinical disease clinical information to group PCa diagnoses into four risk categories
characteristics, as has been observed for other PCa risk according to National Comprehensive Cancer Network (NCCN) guidelines
factors [9]. Finally, because ejaculation frequency may be an [10]. Locally advanced and metastatic disease categories were combined
owing to limited numbers. Men were assigned to the highest category for
indicator of health status and could be related to mortality
which they were eligible: low risk = T1/T2 tumor, PSA < 10 ng/ml,
from multiple causes, the current analysis considers the
Gleason score 6; intermediate risk = T1/T2 tumor, PSA 10–<20 ng/ml,
impact of competing causes of death on our findings. Thus,
Gleason score 7; high risk = T3 tumor, PSA 20–<50 ng/ml, Gleason score 8;
this updated analysis represents a comprehensive evalua- and regional or distant metastases = T4/N1/M1 tumor, PSA 50 ng/ml. To
tion of the association between ejaculation frequency and more carefully explore differences in risk for indolent and aggressive
PCa in a large US-based prospective cohort. disease, we also considered the following subgroups as secondary
analyses: lethal disease (defined as PCa death or metastases to bone or
2. Patients and methods other organs before the end of follow-up), advanced disease (stage T3b, T4
or N1 or M1 at diagnosis or lethal disease during follow-up), organ-
2.1. Study population confined disease (low-grade stage T1 or T2 and N0, M0 at diagnosis and no
progression to metastasis or death during follow-up); and categories of
The HPFS is an ongoing prospective cohort study among 51 529 US male Gleason score based on prostatectomy or biopsy pathology reports
health professionals [8]. In brief, cancer-free, predominantly Caucasian (Gleason  3 + 4 and Gleason  4 + 3).
(>91%) health professionals aged 40–75 yr were recruited in 1986 and
have been followed with biennial questionnaires on medical history and 2.4. Statistical analyses
lifestyle, including known or suspected cancer and chronic disease risk
factors, diet, use of supplements, and preventive behaviors. Ejaculation Person-time was calculated from the return date for the 1992 question-
frequency was assessed in the 1992 questionnaire, which was completed naire to the date of PCa diagnosis, death, or the end of the follow-up
by 46 213 men. Men with a diagnosis of cancer before 1992 (excluding period (January 31, 2010). Actuarial curves for PCa-free survival
non-melanoma skin cancer) were excluded from the analysis, were generated according to the ejaculation frequency category for
976 EUROPEAN UROLOGY 70 (2016) 974–982

age 40–49 yr using the Kaplan-Meier method. Cox proportional hazards reported at age 40–49 yr. We also investigated causes of death across
models were used to estimate the hazard ratio (HR) and 95% confidence ejaculation frequency categories to better understand the different
intervals (CI) for total PCa and for each of the clinical subgroups for each mortality rates. Analyses were run in R using the mstate package [16]. For
ejaculation frequency category. As in the 2004 report [8], 4–7 EPM was all analyses, p < 0.05 was considered statistically significant.
selected as the reference category as relatively few men reported an
average of 0–3 EPM. The top two categories were combined for some
3. Results
analyses owing to small numbers of men in the 21 EPM group. Age-
adjusted and multivariable models were evaluated. Age-adjusted
models are adjusted for age in months (as the time scale) and for
3.1. Baseline characteristics
calendar time. Multivariable models were additionally adjusted for: race
(Caucasian, African-America, Asian, other ancestry, missing); family Ejaculation frequency declined with age. The proportion of
history of PCa (yes/no); vigorous physical activity (quintiles); body mass men reporting average frequency of 13 EPM was 57% at age
index (BMI; <21, 21–<23, 23–<25, 25–<27.5, 27.5–<30, 30 kg/m2, 20–29 yr but dropped to 32% at age 40–49 yr. The Spearman
missing); height (quintiles); diabetes (yes/no); marital status (married, correlation between ejaculation frequency as an ordinal
divorced, other); intake of energy, processed meat, tomato sauce, variable and ages 20–29 and 40–49 yr was 0.66. Some 40% of
calcium, alcohol, and a-linolenic acid (all quintiles); multivitamin use men were in the same frequency category for ages 20–29 and
(yes, no, missing); smoking (never, quit >10 yr ago, quit 10 yr ago,
40–49 yr, and 47% of men moved down a single category from
current, missing); history of vasectomy (yes/no); and history of PSA
age 20–29 yr to age 40–49 yr.
testing (yes/no in the previous 2-yr questionnaire cycle). Statistical
The baseline age-standardized characteristics of the study
significance was evaluated based on a p trend estimated by assigning the
minimum frequency for each category. The missing indicator method
population (n = 31 925) according to average monthly
was used for missing data on most covariates [11]. All participants had ejaculation frequency at age 40–49 yr are presented in
baseline questionnaire data for food frequency, and missing data on Table 1. Having had a PSA test by 1994 or by 2008 was not
nutrients were carried forward from previous reported values. For monotonically associated with ejaculation frequency at age
activity, missing data were assumed to be in the lowest reference 40–49 yr, and the total number of PSA tests was similar across
category. For height, missing data were assumed to be in the middle frequency categories. Among 17 093 men who reported an
category. initial elevated PSA, the percentage who subsequently
reported prostate biopsy was similar across ejaculation
2.5. Sensitivity analyses and effect modification categories. Men reporting 21 EPM who were subsequently
diagnosed with PCa were somewhat less likely to undergo
If men with erectile dysfunction have lower ejaculatory frequency and
radical prostatectomy and more likely to report radiation
serious comorbidities associated with a higher risk of premature death
compared to men reporting lower frequencies.
from other causes, a spurious association between less frequent
Overall, associations between the covariates investigat-
ejaculation and reduced risk of PCa could result. To address this issue,
we performed a sensitivity analysis that excluded men who reported a
ed and ejaculation frequency were similar for frequency at
history of erectile dysfunction, defined as poor or very poor ability to age 20–29 yr and in the year before the questionnaire
maintain an erection without treatment during the period 1990–1994, (Supplementary Tables 1 and 2). Although the associations
as assessed in the 2000 questionnaire. To eliminate a possible effect of were not monotonic, there was some evidence that men in
undiagnosed disease on ejaculation frequency reported, we also the highest ejaculation frequency category in the year
performed analyses that excluded cases diagnosed within the first 4 before the questionnaire were less likely to have had a PSA
yr of follow-up. Finally, to address the fact that diagnostic intensity may screening test by 1994 (45.4% vs 52.6%) and by 2008 (87.5%
vary according to ejaculation frequency, we conducted a sensitivity vs 91.8%). However, the associations between covariates
analysis restricted to a PSA-screened subset of men who reported a PSA
and ejaculatory frequency remained similar to those for the
test before 1994 with follow-up from 1994 to 2010. Stratified analyses
overall cohort when analysis was restricted to the subset of
were performed to evaluate potential effect modification by age, BMI,
and vasectomy status. The statistical significance of effect modification
13 405 screened men who reported having had a PSA test in
was tested using likelihood ratio tests to compare models with and 1994 (Supplementary Table 3).
without interaction terms between the potential effect modifier and
ejaculation frequency. All aforementioned analyses were performed 3.2. Ejaculation frequency and PCa risk
using SAS statistical software (release 9.3; SAS Institute, Cary, NC, USA).
During 480 831 person-years of follow-up, a total of
2.6. Competing risks analysis 3839 incident PCa cases were diagnosed. As shown in
Figure 1, PCa was less frequently diagnosed among men in
PCa has a long natural history [12], is very sensitive to diagnostic the higher ejaculation frequency categories. The age-
intensity [13], and is often indolent [14]. Thus, to better understand the adjusted and multivariable-adjusted HRs according to
interplay between PCa and deaths due to other causes, we modeled these average monthly ejaculation frequency are presented in
events jointly using a multistate model in a semi-competing risks
Table 2. We also present results excluding 10 103 men who
framework [15], as shown in Supplementary Figure 1. Specifically, we
reported erectile dysfunction, leaving 21 822 men and
modeled PCa diagnoses as intermediate states and deaths due to PCa or
2704 total cases.
other causes as final states. We grouped men into the four NCCN risk
categories described above. Each transition hazard was modeled
Results for total PCa were similar for the age-adjusted
assuming that hazards are proportional across ejaculation frequency and multivariable analyses for all three time points at which
levels. We present results for a simple model using age as the time scale ejaculation frequency was assessed, and for the sensitivity
and considering event occurrence by levels of ejaculation frequency analysis excluding men with erectile dysfunction (Table 2).
EUROPEAN UROLOGY 70 (2016) 974–982 977

Table 1 – Age-standardized characteristics at baseline in 1992 for the 31 925 men from the Health Professionals Follow-up Study according
to reported ejaculation frequency at age 40–49 yra

Ejaculation frequency

0–3 EPM 4–7 EPM 8–12 EPM 13–20 EPM 21 EPM

Cases (n) 1713 7812 12147 7440 2813


Age (yr) 59.9 (10.4) 59.1 (9.6) 58.9 (9.1) 58.5 (8.9) 58.0 (8.9)
Height (cm) 178 (7) 178 (7) 178 (7) 178 (7) 178 (7)
Body mass index in 1992 (kg/m2) 25.6 (3.6) 25.5 (3.3) 25.8 (3.4) 26.0 (3.4) 26.3 (3.7)
Total activity (MET h/wk) 31 (35) 33 (38) 37 (39) 41 (47) 42 (46)
Total calorie intake (kcal/d) 1847 (583) 1890 (561) 1923 (576) 1968 (590) 2013 (619)
Alcohol intake (g/d) 8.4 (12.9) 9.4 (13.3) 10.4 (14.2) 11.3 (15.3) 12.2 (17.0)
Processed meat intake (servings/d) 0.3 (0.4) 0.3 (0.4) 0.3 (0.4) 0.3 (0.4) 0.3 (0.4)
Tomato sauce intake (servings/wk) 0.9 (0.9) 0.9 (0.9) 0.9 (0.9) 1.0 (1.0) 1.0 (1.0)
Calcium intake (mg/d) 908 (396) 916 (391) 909 (389) 908 (404) 927 (417)
a-Linolenic acid intake (g/d) 1.0 (0.4) 1.1 (0.4) 1.0 (0.4) 1.0 (0.4) 1.0 (0.4)
Divorced 4.9 4.1 5.1 7.8 11.8
Smoked in the past 10 yr 33.5 37.5 38.8 39.3 39.6
Self-reported history of syphilis or gonorrhea 1.2 2.6 3 3.3 4.6
Vasectomy history 18.2 25 27.6 27.2 27.9
Race
White 93.4 96.3 96.7 96.9 96.2
African-American 0.6 0.6 0.8 0.6 0.8
Asian 4 1.7 1.3 1.2 1.1
Other 2.1 1.3 1.2 1.4 2.0
Screening behavior
Had physical examination in past 2 yr 67.8 71.3 71.1 70.7 68.9
Had rectal examination in past 2 yr 65.4 67.4 68.1 67.1 66.3
Had PSA test by 1994 48.2 54.4 55.6 53.8 50.4
Had PSA test by 2008 90.9 92.7 93.1 92.2 90.6
Total periods with PSA test by 2008 (n)b 4.6 (2.6) 5.1 (2.5) 5.1 (2.5) 4.9 (2.6) 4.8 (2.6)
Had prostate biopsy after first report of elevated PSAc 47.2 47.4 48 46.7 47.2
Primary treatment among 3839 cases
Cases (n) 192 1041 1509 807 290
Radical prostatectomy 38.4 41.8 40.8 41.8 36.2
Radiation therapy 30.2 31.9 34.2 34.3 39.3
Hormone therapy 8.3 5.9 5.7 6.6 8.4
Active surveillance/none 8.4 8.2 8.3 6.7 5.1
Other 2.2 1.6 1.8 2.9 0.7
Missing treatment information 12.6 10.6 9.3 7.7 10.2

EPM = ejaculations per month; MET = metabolic equivalent of task; PSA = prostate-specific antigen.
a
Data are presented as the age-standardized mean (standard deviation) for continuous variables and the age-standardized percentage for categorical variables.
All characteristics except age and primary treatment are age-standardized to the distribution for the full cohort in 1992. Primary treatment is standardized to the
age distribution for all prostate cancer cases.
b
Number of 2-yr questionnaire cycles in which a PSA test was reported in the previous 2 yr (maximum = 8).
c
Among 17 093 men who reported ‘‘elevated PSA’’; based only on first report of elevated PSA.

Compared to men with an average monthly frequency of the rate was 4.49 cases/1000 person-years for 21 EPM and
4–7 ejaculations, men reporting 21 EPM at ages 20–29 and 8.35 cases/1000 person-years for 4–7 EPM (IRD 3.89 cases/
40–49 yr and in 1991 had a significantly lower risk of total 1000 person-years).
PCa, with a multivariable-adjusted HR of 0.81 (95% CI 0.72– Men who reported an average frequency of 21 EPM at
0.92), 0.78 (95% CI 0.69–0.89), and 0.76 (95% CI 0.61–0.94), both age 20–29 yr and age 40–49 yr experienced the same
respectively. Trend tests at each time point when excluding risk reduction for total PCa as men in the highest EPM
men in the lowest ejaculation frequency category, who may category at age 40–49 yr (HR 0.78, 95% CI 0.68–0.90). The
be more likely to have serious comorbidities, were similar to association appeared to be driven by frequency at age
results when considering all five categories (p < 0.0001 for 40–49 yr when frequency at both time points was included
ages 20–29 and 40–49 yr, p = 0.06 for 1991). in the same model. Compared to men with 4–7 EPM, the HR
The absolute PCa incidence rate for frequency at age for men with 13 EPM at age 40–49 yr was 0.85 (95% CI
20–29 yr was 6.56 cases/1000 person-years for 21 EPM 0.76–0.94; p = 0.005 for trend) after adjusting for frequency
and 8.95 cases/1000 person-years for 4–7 EPM (incidence at age 20–29 yr. The HR for 13 EPM compared to 4–7 EPM
rate difference [IRD] 2.39 cases/1000 person-years). For at age 20–29 yr was 0.95 (95% CI 0.83–1.08; p = 0.30 for
frequency at age 40–49 yr, the absolute rate was 6.74 cases/ trend) after adjusting for frequency at age 40–49 yrs.
1000 person-years for 21 EPM and 8.94 cases/1000 However, the correlation between frequencies at different
person-years for 4–7 EPM (IRD 2.20 cases/1000 person- time points makes it challenging to completely disentangle
years). For frequency in the year before the questionnaire, the associations.
978 EUROPEAN UROLOGY 70 (2016) 974–982
[(Fig._1)TD$IG]
A 1.0 Ejaculation frequency at any time point was not signifi-
cantly associated with diagnosis of high-risk PCa or
regional/distant metastases. However, for age 20–29 yr
0.8
there was a suggestion of an inverse association between
ejaculation frequency and local/distant metastases (HR
0.6 0.89, 95% CI 0.68–1.15; p = 0.07 for 13 vs 4–7 EPM).
Survival

The risk of both organ-confined and low-grade PCa was


significantly lower for 13 compared to 4–7 EPM for all
0.4 0 3 EPM
three time periods (Table 4). For high-grade PCa, there was a
4 7 EPM
8 12 EPM suggestion of higher risk for men in the lowest frequency
0.2 13 20 EPM category at age 20–29 yr (HR 1.32, 95% CI 0.91–1.92) and
21 EPM age 40–49 yr (HR 1.38, 95% CI 1.03–1.86). However, there
was some evidence that higher ejaculation frequency in the
0.0
year before questionnaire distribution is associated with
higher risk of advanced PCa (HR 1.37, 95% CI 1.00–1.86) or
B 1.00 lethal PCa (HR 1.48, 95% CI 1.02–2.15), but the trend tests
were only marginally significant (p values 0.11 for advanced
and 0.05 for lethal PCa). When we excluded men diagnosed
within the first 4 yr of follow-up to address the impact of
0.95 undiagnosed disease or early symptoms on the results, the
associations for 13 versus 4–7 EPM were attenuated for
Survival

both advanced PCa (HR 1.15, 95% CI 0.79–1.69; p = 0.36 for


trend) and lethal PCa (HR 1.19, 95% CI 0.73–1.94; p = 0.33
0.90 for trend; data not shown).
Sensitivity analyses, including analyses excluding men
diagnosed with PCa in the first 4 yr of follow-up and
analyses restricted to a screened cohort, produced results
similar to the overall findings (Supplementary Material and
0.85
Supplementary Tables 4 and 5). Several stratified analyses
0 4 8 12 16 were conducted to explore potential effect modifiers.
Time (yr) Results stratified by age at baseline, age at diagnosis, BMI
Number at risk: at diagnosis, and history of vasectomy provided no evidence
0 3 EPM: 1713 1609 1464 1283 1118 that any of these factors modified the association between
4 7 EPM: 7812 7381 6761 6117 5430 ejaculation frequency and PCa risk (data not shown).
8 12 EPM: 12147 11519 10695 9719 8573
13 20 EPM: 7440 7080 6589 6005 5365 3.3. Competing causes of death
21 EPM: 2813 2686 2519 2287 2046

Because ejaculation frequency may be an indicator of health


Fig. 1 – Kaplan-Meier curve for prostate cancer–free survival according
to ejaculation frequency category for age 40–49 yr (1992–2010). (A) Plot status, we fitted the multistate model in Supplementary
over the full prostate cancer–free survival range. (B) Magnified plot for Figure 1 to examine PCa incidence over time across the four
a restricted survival range. EPM = ejaculations per month.
NCCN risk groups and the cumulative incidence of lethal
PCa in light of other causes of death. None of the ejaculation
categories was significantly associated with changes in PCa-
specific survival after diagnosis, but categories for the
According to the four NCCN risk groups, 1585 cases had lowest (0–3 EPM) and highest (13 EPM) frequency had a
localized low-risk PCa, 1493 had localized intermediate-risk higher risk of other-cause mortality (Supplementary
disease, 604 had localized high-risk PCa, and 157 patients Tables 6 and 7). However, the predicted probability of
had evidence of regional or distant metastases at diagnosis. events over time for men who were cancer-free at age 50 yr
Information on clinical disease characteristics was missing (Supplementary Fig.) shows that the reduction in PCa risk in
for 434 (11%) men, who were classified in the two lowest the highest category cannot be fully explained by death
risk categories depending on whether their PCa diagnosis from other causes. Additional details on the results from
occurred after a PSA test (n = 336; 21.2% of the localized this analysis are included in the Supplementary Material.
low-risk group) or in the absence of a PSA test (n = 98; 6.6%
of the localized intermediate-risk group). For all three time 4. Discussion
periods, 13 EPM was associated with a significantly lower
risk (25–28%) of low-risk PCa in comparison to 4–7 EPM The results of this prospective cohort study involving
(Table 3). Ejaculation frequency at age 20–29 yr was 31 925 men, 18 yr of follow-up, and 3839 PCa cases offer
also significantly associated with intermediate-risk PCa additional evidence of a role for ejaculation frequency in
(p = 0.0003), with a 27% reduction for 13 versus 4–7 EPM. the etiology of PCa, particularly for low-risk disease.
EUROPEAN UROLOGY 70 (2016) 974–982 979

Table 2 – Hazard ratio for incidence of total prostate cancer among 31 925 men in the Health Professionals Follow-up Study according to
reported ejaculation frequency in different time periodsa

Ejaculation frequency

0–3 EPM 4–7 EPM 8–12 EPM 13–20 EPM 21 EPM p trend 13 EPM p trend

Age 20–29 yr
Cases (n) 137 438 1208 1303 753 2056
Age-adjusted HR (95% CI) 0.97 (0.80–1.17) 1.00 1.03 (0.92–1.15) 0.92 (0.83–1.03) 0.80 (0.71–0.90) <0.0001 0.87 (0.79–0.97) 0.0001
Age-adjusted no-ED HR (95% CI) 0.90 (0.70–1.15) 1.00 0.98 (0.86–1.12) 0.89 (0.78–1.01) 0.78 (0.67–0.89) <0.0001 0.84 (0.75–0.96) 0.0009
MV HR (95% CI) 0.99 (0.81–1.19) 1.00 1.03 (0.92–1.14) 0.92 (0.82–1.02) 0.81 (0.72–0.92) <0.0001 0.88 (0.79–0.97) 0.0002
MV no-ED HR (95% CI) 0.91 (0.71–1.17) 1.00 0.99 (0.86–1.13) 0.90 (0.78–1.02) 0.80 (0.69–0.92) <0.0001 0.86 (0.76–0.97) 0.002
Age 40–49 yr
Cases (n) 192 1041 1509 807 290 1097
Age-adjusted HR (95% CI) 0.87 (0.74–1.01) 1.00 0.91 (0.84–0.98) 0.80 (0.72–0.87) 0.77 (0.67–0.87) <0.0001 0.79 (0.72–0.86) <0.0001
Age-adjusted no-ED HR (95% CI) 0.91 (0.75–1.11) 1.00 0.93 (0.85–1.02) 0.80 (0.72–0.89) 0.80 (0.68–0.93) <0.0001 0.80 (0.72–0.88) <0.0001
MV HR (95% CI) 0.88 (0.76–1.03) 1.00 0.90 (0.83–0.98) 0.80 (0.73–0.88) 0.78 (0.69–0.89) <0.0001 0.80 (0.73–0.87) <0.0001
MV no-ED HR (95% CI) 0.91 (0.75–1.11) 1.00 0.93 (0.84–1.02) 0.81 (0.72–0.90) 0.82 (0.70–0.96) 0.0006 0.81 (0.73–0.90) 0.0002
Year before questionnaire (1991)
Cases (n) 1293 1299 831 322 94 416
Age-adjusted HR (95% CI) 1.03 (0.95–1.11) 1.00 0.95 (0.87–1.04) 0.90 (0.80–1.02) 0.73 (0.59–0.90) 0.0004 0.86 (0.76–0.96) 0.002
Age-adjusted no-ED HR (95% CI) 1.06 (0.96–1.17) 1.00 0.96 (0.87–1.06) 0.91 (0.80–1.05) 0.71 (0.56–0.90) 0.0005 0.86 (0.76–0.97) 0.002
MV HR (95% CI) 1.05 (0.97–1.13) 1.00 0.96 (0.87–1.05) 0.93 (0.82–1.05) 0.76 (0.61–0.94) 0.001 0.89 (0.79–0.99) 0.004
MV no-ED HR (95% CI) 1.07 (0.97–1.19) 1.00 0.96 (0.87–1.06) 0.94 (0.82–1.08) 0.74 (0.58–0.94) 0.002 0.89 (0.78–1.01) 0.007

EPM = ejaculations per month; HR = hazard ratio; CI = confidence interval; ED = erectile dysfunction; MV = multivariate.
a
Age-adjusted models are adjusted for age in months and calendar time. Multivariate models are additionally adjusted for: race; family history of prostate
cancer; vigorous physical activity (quintiles); body mass index (six categories); height (quintiles); diabetes; marital status; intake of energy, processed meat,
tomato sauce, calcium, alcohol, and a-linolenic acid (all quintiles); multivitamin use; smoking (never, quit >10 yr ago, quit 10 yr ago, current); history of
vasectomy; and history of PSA testing. No-ED models exclude men who reported poor or very poor ability to have and maintain an erection in the time period
1990–1994, for which analyses include 21 822 participants and 2704 prostate cancer events.

The absolute difference in PCa rate between 21 and An initial report published in 2004 for this cohort found
4–7 EPM was 2.39 cases/1000 person-years for frequency at that more frequent ejaculation was related to a lower risk of
age 20–29 yr, 2.20 cases/1000 person-years for frequency total PCa, with strongest associations for higher frequency
at age 40–49 yr, and 3.89 cases/1000 person-years for in the year before questionnaire distribution [8]. With an
frequency in the year before questionnaire distribution. additional decade of follow-up, we demonstrate that

Table 3 – Hazard ratio for prostate cancer by disease severity among 31 925 men in the Health Professionals Follow-up Study according to
reported ejaculation frequency at different timesa,b

0–3 EPM 4–7 EPM 8–12 EPM 13 EPM p value

Cases MV HR Cases MV HR Cases MV HR Cases MV HR for trend


(n) (95% CI) (n) (95% CI) (n) (95% CI) (n) (95% CI)

Age 20–29 yr
Low-risk disease 40 0.84 (0.59–1.18) 160 1.00 (Ref) 407 0.91 (0.75–1.09) 687 0.75 (0.63–0.89) 0.0006
Intermediate-risk disease 39 0.85 (0.60–1.20) 156 1.00 (Ref) 390 0.88 (0.73–1.06) 664 0.73 (0.61–0.88) 0.0003
High-risk disease 43 0.99 (0.70–1.39) 142 1.00 (Ref) 435 1.13 (0.93–1.36) 775 1.00 (0.84–1.20) 0.46
Regional/distant metastases 28 1.20 (0.77–1.87) 70 1.00 (Ref) 186 1.01 (0.77–1.33) 320 0.89 (0.68–1.15) 0.07
Age 40–49 yr
Low-risk disease 65 0.95 (0.72–1.23) 347 1.00 (Ref) 502 0.88 (0.77–1.01) 335 0.72 (0.61–0.83) <0.0001
Intermediate-risk disease 52 0.70 (0.52–0.94) 363 1.00 (Ref) 579 0.99 (0.87–1.13) 401 0.83 (0.72–0.96) 0.11
High-risk disease 39 1.13 (0.80–1.61) 160 1.00 (Ref) 217 0.85 (0.69–1.04) 188 0.89 (0.72–1.11) 0.17
Regional/distant metastases 7 0.61 (0.27–1.35) 44 1.00 (Ref) 55 0.85 (0.57–1.27) 51 0.96 (0.64–1.45) 0.65
Year before
questionnaire (1991)
Low-risk disease 365 1.05 (0.91–1.21) 446 1.00 (Ref) 303 0.94 (0.81–1.09) 135 0.75 (0.62–0.92) 0.002
Intermediate-risk disease 432 1.01 (0.89–1.16) 481 1.00 (Ref) 325 1.00 (0.87–1.15) 157 0.89 (0.74–1.07) 0.27
High-risk disease 232 1.18 (0.96–1.45) 180 1.00 (Ref) 129 1.20 (0.95–1.50) 63 1.12 (0.84–1.51) 0.94
Regional/distant metastases 72 0.94 (0.64–1.38) 48 1.00 (Ref) 15 0.59 (0.33–1.07) 22 1.82 (1.07–3.10) 0.19

EPM = ejaculations per month; MV = multivariate; HR = hazard ratio; CI = confidence interval; PSA = prostate-specific antigen.
a
Risk groups are based on National Comprehensive Cancer Network guidelines. Men were assigned to the highest category for which they were eligible:
low risk = T1/T2 tumor, PSA <10 ng/ml, Gleason score 6; intermediate risk = T1/T2 tumor, PSA 10–<20 ng/ml, Gleason score 7; High risk = T3 tumor, PSA level
20–<50 ng/ml, Gleason score 8; regional or distant metastases = T4/N1/M1 tumor, PSA >50 ng/ml.
b
Age-adjusted models are adjusted for age in months and calendar time. Multivariate models are additionally adjusted for: race; family history of prostate
cancer; vigorous physical activity (quintiles); body mass index (six categories); height (quintiles); diabetes; marital status; intake of energy, processed meat,
tomato sauce, calcium, alcohol, and a-linolenic acid (all quintiles); multivitamin use; smoking (never, quit >10 yrs ago, quit 10 yrs ago, current); history of
vasectomy; and history of PSA testing.
980 EUROPEAN UROLOGY 70 (2016) 974–982

Table 4 – Multivariable prostate cancer incidence by disease severity among 31 925 men in the Health Professionals Follow-up Study
according to reported ejaculation frequency at different timesa

Ejaculation frequency

0–3 EPM 4–7 EPM 8–12 EPM 13 EPM p value

Cases MV HR Cases MV HR Cases MV HR Cases MV HR for trend


(n) (95% CI) (n) (95% CI) (n) (95% CI) (n) (95% CI)

Age 20–29 yr
Low grade (GS  3 + 4) 76 0.88 (0.68–1.13) 283 1.00 (Ref) 785 1.00 (0.87–1.15) 1364 0.86 (0.76–0.98) 0.006
High grade (GS  4 + 3) 39 1.32 (0.91–1.92) 93 1.00 (Ref) 244 1.00 (0.78–1.27) 451 0.93 (0.74–1.16) 0.08
Organ-confined 98 1.04 (0.82–1.30) 309 1.00 (Ref) 877 1.04 (0.91–1.18) 1508 0.89 (0.78–1.00) 0.0008
Advanced 21 0.79 (0.48–1.29) 72 1.00 (Ref) 156 0.87 (0.65–1.15) 268 0.80 (0.62–1.05) 0.23
Lethal 14 0.63 (0.35–1.14) 57 1.00 (Ref) 113 0.83 (0.60–1.14) 200 0.82 (0.60–1.10) 0.71
Age 40–49 yr
Low grade (GS  3 + 4) 106 0.76 (0.62–0.93) 689 1.00 (Ref) 1010 0.90 (0.82–0.99) 703 0.76 (0.68–0.85) <0.0001
High grade (GS  4 + 3) 57 1.38 (1.03–1.86) 197 1.00 (Re) 318 1.01 (0.85–1.21) 255 0.99 (0.82–1.19) 0.23
Organ-confined 132 0.86 (0.71–1.03) 749 1.00 (Ref) 1126 0.93 (0.84–1.02) 785 0.78 (0.71–0.86) <0.0001
Advanced 33 0.98 (0.67–1.44) 146 1.00 (Ref) 185 0.83 (0.66–1.03) 153 0.85 (0.67–1.07) 0.16
Lethal 24 0.95 (0.61–1.49) 107 1.00 (Ref) 133 0.83 (0.64–1.08) 120 0.95 (0.73–1.24) 0.78
Year before
questionnaire (1991)
Low grade (GS  3 + 4) 778 1.04 (0.94–1.15) 869 1.00 (Ref) 587 0.97 (0.87–1.08) 274 0.82 (0.72–0.95) 0.003
High grade (GS  4 + 3) 284 1.08 (0.91–1.29) 264 1.00 (Ref) 180 1.07 (0.88–1.30) 99 1.13 (0.89–1.43) 0.68
Organ-confined 901 1.08 (0.98–1.18) 952 1.00 (Ref) 648 1.00 (0.90–1.11) 291 0.82 (0.72–0.94) 0.0008
Advanced 211 0.99 (0.79–1.22) 157 1.00 (Ref) 88 0.98 (0.75–1.28) 61 1.37 (1.00–1.86) 0.11
Lethal 169 0.95(0.74–1.22) 114 1.00 (Ref) 60 1.01 (0.74–1.39) 41 1.48(1.02–2.15) 0.05

EPM = ejaculations per month; MV = multivariable; HR = hazard ratio; CI = confidence interval; GS = Gleason score.
a
Organ-confined disease: stage T1/2N0M0 at diagnosis that did not progress to metastasis or death during the follow-up period. Advanced disease: stage
T3b/4 or N1 or M1 at diagnosis or prostate cancer death or distant metastasis during follow-up. Lethal disease: prostate cancer death or metastases to bone
or other organs before the end of the follow-up period.

ejaculation frequency at three different time points during increase of only 1.8% in the risk of dying from other causes
adulthood is associated with statistically significant modest by age 80 yr, while their decrease in PCa risk is 3.8%. Thus, in
reductions in risk of total PCa. The association with both cases the reduction in PCa risk may be partly explained
frequency at age 20–29 yr became more pronounced with by premature death due to other causes, but the reduction
additional follow-up, while the associations with frequency among men reporting high ejaculation frequency seemingly
at age 40–49 yr and in the year before the questionnaire cannot be explained by this effect alone.
remained statistically significant but were somewhat Several important limitations of our study should be
attenuated. Taken together with the fact that strong inverse noted. Ascertainment of the exposure relied on reporting of
associations remained after excluding men diagnosed in the sexual activity in the past. This may introduce measure-
first 4 yr of follow-up, the updated results are unlikely to ment error, particularly in the reporting of frequency at age
be strongly influenced by the effects of undiagnosed disease 20–29 yr. However, the use of an anonymized questionnaire
on ejaculation frequency. may have resulted in more accurate reporting of sexual
Importantly, our findings were robust to adjustment for behaviors than a one-on-one interview [17]. Previous
time-varying factors such as BMI, physical activity, and diet studies suggest that the validity of data on sensitive
that differed with ejaculation frequency and that have also information is further improved by: (1) an understanding
been associated with PCa and its progression [5], as well as among participants that their data will be kept confidential,
other factors associated with PCa risk in this cohort. Because most likely true in this large ongoing cohort in which men
men in the higher ejaculation frequency categories had had already responded to at least one and potentially two
some exposure patterns that might put them at higher risk previous questionnaires; and (2) the avoidance of implying
of morbidity and mortality due to other causes—higher BMI, a ‘‘normal’’ response category [18]. Most importantly,
greater alcohol consumption, and more frequent history of however, the data were collected prospectively, preventing
smoking and sexually transmitted infections—we were differential misclassification (ie, recall bias). Thus, our
concerned that the reduction in PCa risk we observed in this results can be considered conservative estimates of the true
group might be attributable to premature death from other association.
causes among men who may have had undiagnosed PCa. While the utilization of a large prospective study has
Thus, a strength of our study is the consideration of a model numerous advantages, the clinical information available for
for semi-competing risks. From this model, the increase in men at the time of their PCa diagnosis is more limited than
risk of death by age 80 yr among men with the lowest in clinical settings. Thus, we are not able to distinguish very
ejaculation frequency is 3.8%, while the reduction in PCa low-risk disease from low-risk disease in subgroup
risk is 2.2%. By comparison, men reporting 13 EPM have an analyses. The abundance of data on potential confounders
EUROPEAN UROLOGY 70 (2016) 974–982 981

is an advantage of working within the well-annotated HPFS more frequent ejaculation may reduce the development of
cohort, but we still cannot rule out residual confounding by prostatic intraluminal crystalloids, which have been asso-
other lifestyle factors. Furthermore, our cohort consisted ciated with higher risk of PCa [34,35]. Higher ejaculatory
primarily of Caucasian men and the frequency of ejaculation frequency may be linked to lowering of psychological
may vary across populations. However, the results may still tension and central sympathetic nervous system suppres-
be generalizable to other men, as we would not expect a sion, which could dampen the stimulation of prostate
true biological association between ejaculation frequency epithelial cell division [36]. Given the lack of modifiable risk
and PCa to differ by race or ethnicity. factors identified for PCa to date, the specific biological
The literature exploring the role of sexual activity in the mechanisms underlying these associations are worthy of
etiology of PCa is inconsistent [7,19–30]. Previous studies further investigation.
are primarily retrospective case-control studies, raising
concerns about recall bias, especially given that erectile 5. Conclusions
dysfunction, ejaculatory dysfunction, and decreased libido
are common consequences of both PCa and its treatment This large prospective study provides the strongest
[31,32]. Moreover, few previous studies have considered evidence to date of a beneficial role of ejaculation in
ejaculation frequency per se, with most utilizing proxies of prevention of PCa, a disease for which relatively little is
sexual activity, such as age at first marriage, marital status, understood about etiology generally and knowledge of
number of sexual partners, and number of children. Few modifiable risk factors is particularly scant. The results are
previous studies have examined associations according to robust to adjustment for many dietary, lifestyle, and
tumor grade or stage despite their particular importance for screening behaviors, but additional work on the underlying
PCa; spurious associations with more favorable disease may biological mechanisms should be undertaken to corrobo-
result from confounding by early detection. We do, in fact, rate these findings given the potential for residual
find that the inverse association with overall PCa is driven confounding. More frequent ejaculation in the absence of
by low-risk disease, which could indicate that more risky sexual behaviors could represent an important means
sexually active men might undergo less screening and of reducing the profound medical costs and physical and
follow-up testing. This alternative explanation for our psychological side effects of unnecessary diagnosis and
findings is especially plausible given the potential resulting treatment of low-risk tumors, even though it appears to be
side effects of PCa and its treatment on sexual function. less strongly associated with aggressive disease.
However, PSA screening history and biopsy utilization after
elevated PSA were quite similar across the ejaculation Author contributions: Jennifer R. Rider had full access to all the data in
frequency categories. Moreover, the results were consistent the study and takes responsibility for the integrity of the data and the
even when we restricted the analysis to a screened cohort accuracy of the data analysis.
and PSA history was taken into account. Nonetheless, we
Study concept and design: Rider, Mucci, Giovannucci.
cannot rule out residual confounding by screening or post- Acquisition of data: Rider, Wilson, Mucci, Giovannucci.
screening biopsy behaviors. Analysis and interpretation of data: Rider, Wilson, Sinnott, Mucci,
Our results identified suggestive but not statistically Giovannucci.
significant associations between higher ejaculation fre- Drafting of the manuscript: Rider, Wilson.
quency in the year before the questionnaire and both Critical revision of the manuscript for important intellectual content: Rider,
advanced and lethal PCa. However, the findings appear to be Wilson, Sinnott, Kelly, Mucci, Giovannucci.
driven by men diagnosed in the period immediately Statistical analysis: Rider, Wilson, Sinnott, Kelly.
Obtaining funding: Giovannucci.
following the questionnaire. The attenuated association
Administrative, technical, or material support: None.
in sensitivity analyses excluding men diagnosed in the first
Supervision: Mucci, Giovannucci.
4 yr of follow-up, together with the fact that these
Other: None.
suggestive positive associations were only found for
ejaculation frequency in the year before the questionnaire Financial disclosures: Jennifer R. Rider certifies that all conflicts of
distribution and not at younger ages, is consistent with men interest, including specific financial interests and relationships and
with undiagnosed aggressive PCa experiencing symptoms affiliations relevant to the subject matter or materials discussed in the
manuscript (eg, employment/affiliation, grants or funding, consultan-
that promoted more frequent ejaculation. While we are not
cies, honoraria, stock ownership or options, expert testimony, royalties,
aware of any literature supporting ejaculation for relief of
or patents filed, received, or pending), are the following: None.
PCa symptoms, it nonetheless seems unlikely that these
suggestive associations with advanced and lethal disease Funding/Support and role of the sponsor: The Health Professionals
reflect causality. Follow-Up Study is supported in part by grants UM1 CA167552, P01
In addition to the prostate stagnation hypothesis [7], a CA055075, P01 CA133891, and P01 CA141298. Jennifer R. Rider, Kathryn
number of mechanisms have been proposed to explain an M. Wilson, and Lorelei A. Mucci are supported by Prostate Cancer
Foundation Young Investigator Awards. The sponsors played no role in
inverse association between ejaculation frequency and PCa.
the study.
More frequent ejaculation may influence the function of
peripheral-zone epithelial cells, hindering the metabolic Acknowledgments: We are grateful to the participants and staff of the
switch from citrate secretion to citrate oxidation known to Health Professionals Follow-up Study for their valuable contributions. In
occur early in prostate tumorigenesis [33]. Alternatively, addition, we would like to thank the following state cancer registries for
982 EUROPEAN UROLOGY 70 (2016) 974–982

their cooperation and assistance: AL, AZ, AR, CA, CO, CT, DE, FL, GA, ID, IL, [16] R Core Team. R: a language and environment for statistical com-
IN, IA, KY, LA, ME, MD, MA, MI, NE, NH, NJ, NY, NC, ND, OH, OK, OR, PA, RI, puting. Vienna, Austria: R Foundation for Statistical Computing;
SC, TN, TX, VA, WA, and WY. JRR and KMW assume full responsibility for 2014.
analyses and interpretation of these data. Finally, we are indebted to [17] Fenton KA, Johnson AM, McManus S, Erens B. Measuring sexual
David Havelick for his enthusiastic support of this project. behaviour: methodological challenges in survey research. Sex
Transm Infect 2001;77:84–92.
[18] Tourangeau R, Yan T. Sensitive questions in surveys. Psychol Bull
Appendix A. Supplementary data 2007;133:859–83.
[19] Sarma AV, McLaughlin JC, Wallner LP, et al. Sexual behavior,
Supplementary data associated with this article can be sexually transmitted diseases and prostatitis: the risk of prostate
found, in the online version, at http://dx.doi.org/10.1016/j. cancer in black men. J Urol 2006;176:1108–13.
eururo.2016.03.027. [20] Fernandez L, Galan Y, Jimenez R, et al. Sexual behaviour, history of
sexually transmitted diseases, and the risk of prostate cancer: a
case-control study in Cuba. Int J Epidemiol 2005;34:193–7.
References
[21] Hayes RB, Pottern LM, Strickler H, et al. Sexual behaviour, STDs and
[1] Soerjomataram I, Lortet-Tieulent J, Parkin DM, et al. Global burden risks for prostate cancer. Br J Cancer 2000;82:718–25.
of cancer in 2008: a systematic analysis of disability-adjusted life- [22] Dennis LK, Dawson DV. Meta-analysis of measures of sexual activi-
years in 12 world regions. Lancet 2012;380:1840–50. ty and prostate cancer. Epidemiology 2002;13:72–9.
[2] Gardner JR, Livingston PM, Fraser SF. Effects of exercise on treat- [23] Honda GD, Bernstein L, Ross RK, Greenland S, Gerkins V, Henderson
ment-related adverse effects for patients with prostate cancer BE. Vasectomy, cigarette smoking, and age at first sexual inter-
receiving androgen-deprivation therapy: a systematic review. J Clin course as risk factors for prostate cancer in middle-aged men. Br J
Oncol 2014;32:335–46. Cancer 1988;57:326–31.
[3] Kenfield SA, Stampfer MJ, Giovannucci E, Chan JM. Physical activity [24] Spence AR, Rousseau MC, Parent ME. Sexual partners, sexually
and survival after prostate cancer diagnosis in the Health Profes- transmitted infections, and prostate cancer risk. Cancer Epidemiol
sionals Follow-up Study. J Clin Oncol 2011;29:726–32. 2014;38:700–7.
[4] Richman EL, Kenfield SA, Stampfer MJ, Paciorek A, Carroll PR, Chan [25] Rotkin ID. Studies in the epidemiology of prostatic cancer: expand-
JM. Physical activity after diagnosis and risk of prostate cancer ed sampling. Cancer Treat Rep 1977;61:173–80.
progression: data from the cancer of the prostate strategic urologic [26] Ahmadi H, Allameh F, Baradaran N, et al. Circulating sex hormones
research endeavor. Cancer Res 2011;71:3889–95. play no role in the association between sexual activity and the risk
[5] Wilson KM, Giovannucci EL, Mucci LA. Lifestyle and dietary factors of prostate cancer. J Sex Med 2011;8:905–13.
in the prevention of lethal prostate cancer. Asian J Androl [27] Giles GG, Severi G, English DR, et al. Sexual factors and prostate
2012;14:365–74. cancer. BJU Int 2003;92:211–6.
[6] Discacciati A, Wolk A. Lifestyle and dietary factors in prostate [28] Hayes RB, de Jong FH, Raatgever J, et al. Physical characteristics and
cancer prevention. Recent Results Cancer Res 2014;202:27–37. factors related to sexual development and behaviour and the risk
[7] Isaacs JT. Prostatic structure and function in relation to the etiology for prostatic cancer. Eur J Cancer Prev 1992;1:239–45.
of prostatic cancer. Prostate 1983;4:351–66. [29] Rosenblatt KA, Wicklund KG, Stanford JL. Sexual factors and the risk
[8] Leitzmann MF, Platz EA, Stampfer MJ, Willett WC, Giovannucci E. of prostate cancer. Am J Epidemiol 2001;153:1152–8.
Ejaculation frequency and subsequent risk of prostate cancer. JAMA [30] Oishi K, Okada K, Yoshida O, et al. A case-control study of prostatic
2004;291:1578–86. cancer in Kyoto, Japan: sexual risk factors. Prostate 1990;17:269–79.
[9] Giovannucci E, Liu Y, Platz EA, Stampfer MJ, Willett WC. Risk factors [31] Saitz TR, Serefoglu EC, Trost LW, Thomas R, Hellstrom WJ. The pre-
for prostate cancer incidence and progression in the Health Profes- treatment prevalence and types of sexual dysfunction among
sionals Follow-up Study. Int J Cancer 2007;121:1571–8. patients diagnosed with prostate cancer. Andrology 2013;1:859–63.
[10] National Comprehensive Cancer Network. Clinical practice guide- [32] Choo R, Long J, Gray R, Morton G, Gardner S, Danjoux C. Prospective
lines in oncology: prostate cancer. Fort Washington, PA: NCCN; 2016. survey of sexual function among patients with clinically localized
[11] Greenland S, Finkle WD. A critical look at methods for handling prostate cancer referred for definitive radiotherapy and the impact
missing covariates in epidemiologic regression analyses. Am J of radiotherapy on sexual function. Support Care Cancer 2010;18:
Epidemiol 1995;142:1255–64. 715–22.
[12] Popiolek M, Rider JR, Andren O, et al. Natural history of early, [33] Costello LC, Franklin RB. The clinical relevance of the metabolism of
localized prostate cancer: a final report from three decades of prostate cancer; zinc and tumor suppression: connecting the dots.
follow-up. Eur Urol 2013;63:428–35. Mol Cancer 2006;5:17.
[13] Thompson IM, Pauler DK, Goodman PJ, et al. Prevalence of prostate [34] Svatek RS, Karam JA, Rogers TE, Shulman MJ, Margulis V, Benaim EA.
cancer among men with a prostate-specific antigen level 4.0 ng Intraluminal crystalloids are highly associated with prostatic ade-
per milliliter. N Engl J Med 2004;350:2239–46. nocarcinoma on concurrent biopsy specimens. Prostate Cancer
[14] Jahn JL, Giovannucci EL, Stampfer MJ. The high prevalence of Prostatic Dis 2007;10:279–82.
undiagnosed prostate cancer at autopsy: implications for epidemi- [35] Del Rosario AD, Bui HX, Abdulla M, Ross JS. Sulfur-rich prostatic
ology and treatment of prostate cancer in the prostate-specific intraluminal crystalloids: a surgical pathologic and electron probe
antigen era. Int J Cancer 2015;137:2795–802. X-ray microanalytic study. Hum Pathol 1993;24:1159–67.
[15] Putter H, Fiocco M, Geskus RB. Tutorial in biostatistics: competing [36] Newman HF, Reiss H, Northup JD. Physical basis of emission,
risks and multi-state models. Stat Med 2007;26:2389–430. ejaculation, and orgasm in the male. Urology 1982;19:341–50.
original article Annals of Oncology 20: 542–549, 2009
doi:10.1093/annonc/mdn644
Published online 6 January 2009

A phase III randomized, double-blind,


placebo-controlled trial of gabapentin in the
management of hot flashes in men (N00CB)
C. L. Loprinzi1*, A. C. Dueck1, B. S. Khoyratty2, D. L. Barton1, S. Jafar3, K. M. Rowland Jr4,
P. J. Atherton1, G. W. Marsa5, W. H. Knutson6, J. D. Bearden III7, L. Kottschade1 & T. R. Fitch8
1
Division of Medical Oncology, Mayo Clinic and Mayo Foundation, Rochester; 2Metro-Minnesota Community Clinical Oncology Program, St Louis Park; 3Michigan
Cancer Research Consortium, Ann Arbor; 4Carle Cancer Center CCOP, Urbana; 5Toledo Community Hospital Oncology Program CCOP, Toledo; 6Duluth CCOP,
Duluth, MN; 7Upstate Carolina CCOP, Spartanburg; 8Mayo Clinic Arizona, Scottsdale, USA

Received 2 July 2008; revised 26 August 2008; accepted 27 August 2008

Introduction: Hot flashes represent a significant problem in men undergoing androgen deprivation therapy.
Materials and methods: Via a prospective, double-blind, placebo-controlled clinical trial, men with hot flashes, on
a stable androgen deprivation therapy program for prostate cancer, received a placebo or gabapentin at target doses
of 300, 600, or 900 mg/day. Hot flash frequencies and severities were recorded daily during a baseline week and for
4 weeks while the patients took the study medication.
Results: In the 214 eligible patients who began the study drug on this trial, comparing the fourth treatment week to
the baseline week, mean hot flash scores decreased in the placebo group by 4.1 units and in the three increasing
dose gabapentin groups by, 3.2, 4.6, and 7.0 units. Comparing the three combined gabapentin arms to the placebo
original
article

arm did not result in significant hot flash differences. Wilcoxon rank-sum P values for change in hot flash scores and
frequencies after 4 weeks of treatment were 0.10 and 0.02, comparing the highest dose gabapentin arm to the
placebo arm, respectively. The gabapentin was well tolerated in this trial.
Conclusion: These results support that gabapentin decreases hot flashes, to a moderate degree, in men with
androgen ablation-related vasomotor dysfunction.
Key words: hot flashes, men, prostate cancer

introduction Anecdotal experience reported a reduction of hot flashes in five


women and one man [7]. Subsequent pilot trials also suggested
Hot flashes can be a prominent problem in men with prostate efficacy [8, 9].
cancer undergoing androgen deprivation therapy. Whether Given this promising evidence and the similar treatment
androgen deprivation therapy is carried out by medical or benefits of some drugs for hot flashes in men and women, it
surgical means, hot flashes affect up to 75% of patients [1, 2]. was hypothesized that gabapentin would be effective against hot
There are limited therapeutic options for treating men with flashes in men receiving androgen ablation therapy. This
hot flashes. While clonidine moderately helps hot flashes in current clinical trial was developed to study the efficacy and
women, a randomized, placebo-controlled, double-blind trial side-effects of three relatively low gabapentin doses in this
demonstrated no significant benefit in men [3]. Placebo- patient population.
controlled trials evaluating estrogen or progestational agents
have demonstrated efficacy in treating male hot flashes, with
about a 75% reduction seen with hormones versus a 25% materials and methods
reduction with a placebo [4, 5]. Nonetheless, these hormonal
treatments can have side-effects. Estrogen can cause breast patient eligibility characteristics
tenderness and enlargement. Progestational agents have been Men with a history of prostate cancer, who were on a stable program of
associated with rising prostate-specific antigen levels in some androgen ablation hormone therapy for the prior 4 weeks and who were
men with prostate cancer [6]. not planning on discontinuing hormone therapy for 5 weeks, were eligible
Gabapentin is an antiseizure agent which has been for this trial if they had bothersome hot flashes. Bothersome hot flashes
utilized for the treatment of a number of pain syndromes. were defined by their occurrence of at least 14 times per week and of
sufficient severity to make the patient desire therapeutic intervention, with
*Correspondence to: Dr C. L. Loprinzi, Division of Medical Oncology, Mayo Clinic, 200 First
the hot flashes being present for at least 1 month before study entry.
Street SW, Rochester, MN 55905, USA. Tel: +1-507-284-8964; Fax: +1-507-284-1803; Patients were required to have an estimated life expectancy of at least
E-mail: cloprinzi@mayo.edu 6 months and a good performance status.

ª The Author 2009. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
All rights reserved. For permissions, please email: journals.permissions@oxfordjournals.org
Annals of Oncology original article
Eligible patients could not have had a history of any significant renal completed daily for 5 weeks, symptom experience diaries to be completed
insufficiency (defined as a serum creatinine of 1.5 times normal in the prior weekly for 5 weeks, and the 30-item Profile of Mood States-Brief
2 years) nor could they have any of the following concurrent (£4 weeks) or (POMS-B) [11] form to be completed at the end of the baseline week and
planned therapies: antineoplastic chemotherapy, androgens, estrogens, or then at the end of 4 weeks of treatment. Hot flash definitions were
progestational agents. They could not have used gabapentin previously.
provided to each patient, derived from other men who had participated in
Antidepressant use was allowed if the patient had been on a stable dose for
a previous hot flash trial [12]. The hot flash diary was similar to
at least 1 month and did not plan to modify this treatment during the
a validated diary that has been utilized in a series of clinical trials which
ensuing 5 weeks.
All patients provided written informed consent for participating in this have now involved >1500 subjects [13]. This diary questionnaire inquired
trial, monitored by local Internal Review Boards as mandated by United regarding the numbers of mild, moderate, severe, and very severe hot
States Federal regulations. Patients were stratified before randomization on flashes per 24-hour period. The weekly symptom experience diaries asked
the basis of the patient’s reported number of hot flashes per day (2–3 versus about the severity of potential side-effects of gabapentin on a 0–10 scale
4–9 versus ‡10) and the reported duration of hot flash symptoms (<9 including appetite loss, undesirable appetite increase, nausea, mouth
versus ‡9 months). Patients were then randomized, using a dynamic dryness, fatigue, dizziness, trouble walking/balance problems, muscle
allocation method that balances the marginal distributions of the pain, trouble concentrating, constipation, sleepiness, sleeping troubles,
stratification factors [10], to receive one of four treatment schedules: blurry vision, nervousness, and mood changes. It also inquired about
gabapentin 300 mg at bedtime for 28 days, versus gabapentin 300 mg at
hot flash distress, satisfaction with hot flash control, quality of life (QoL),
bedtime for 7 days and then 300 mg twice daily for 21 days, versus
and how much hot flashes affected QoL, each on a 0–10 scale. The
gabapentin 300 mg at bedtime for 7 days then 300 mg twice daily for 7 days
POMS-B asked about the severity of weekly mood changes. Throughout
and then 300 mg three times daily for 14 days, versus a placebo for
the presented results, all patient-reported outcomes have been
28 days. Patients receiving placebo were divided into three groups so that
one group received one tablet at bedtime for 28 days, a second group transformed onto a 0–100 scale with 0 indicating the most negative
received one tablet at bedtime for 7 days then one tablet b.i.d. for 21 days, outcome and 100 indicating the most positive outcome, for ease of
and a third group received one tablet at bedtime for 7 days then one comparability. On the last day of each treatment week, patients were
tablet b.i.d. for 7 days then one tablet t.i.d. for 14 days. The assigned asked about their compliance with taking the study medication by asking
treatment was administered following the completion of a 1-week period them how many tablets they were taking per day.
to collect baseline hot flash data. All treatment assignments were blinded Each patient was to be contacted by telephone weekly for the first 5 weeks
to the patients and clinical investigators, with only the North Central to document compliance, encourage completion of the questionnaires, and
Cancer Treatment Group (NCCTG) randomization office, the pharmacists, address questions.
and the study statisticians having access to the drug assignments for On the last scheduled day of the randomized double-blind treatment,
individual patients. Gabapentin and matching placebo tablets were each patient, after assuring that he had completed his questionnaires, was
provided by Pfizer Corporation, New York. allowed to be told which dose of gabapentin he received and was
At study entry, the patient was given a booklet that contained patient subsequently given the choice of whether he would like to continue with
instructions, hot flash definitions, hot flash diary questionnaires to be gabapentin or to start active drug if he was on the placebo.

Table 1. Baseline eligible patient characteristics

Placebo (n = 53) 300 mg/day (n = 54) 600 mg/day (n = 53) 900 mg/day (n = 54) P valuea
Age 0.89
Mean (standard deviation) 70 (8.89) 69 (8.53) 70 (7.02) 69 (7.69)
Median 71 69 69 72
Range (47-87) (50-89) (58-86) (52-85)
Race/ethnicity 0.66
White 49 (93%) 49 (91%) 49 (93%) 50 (93%)
Black or African-American 3 (6%) 5 (9%) 3 (6%) 4 (7%)
Asian 1 (2%) 0 (0%) 0 (0%) 0 (0%)
Hispanic 0 (0%) 0 (0%) 1 (2%) 0 (0%)
ECOG performance score 0.39
0 41 (77%) 47 (87%) 42 (79%) 40 (74%)
1 12 (23%) 7 (13%) 11 (21%) 14 (26%)
No. of hot flashes per day 0.62
1: 2–3 7 (13%) 5 (9%) 4 (7%) 10 (19%)
2: 4–9 22 (42%) 26 (48%) 28 (53%) 23 (43%)
3: ‡10 24 (45%) 23 (43%) 21 (40%) 21 (39%)
Duration of hot flashes (months) 0.99
1: <9 31 (58%) 33 (61%) 31 (58%) 32 (59%)
2: ‡9 22 (42%) 21 (39%) 22 (42%) 22 (41%)
a
P values for Kruskal–Wallis test for age and chi-squared test for all other variables.
ECOG, Eastern Cooperative Oncology Group.

Volume 20 | No. 3 | March 2009 doi:10.1093/annonc/mdn644 | 543


original article Annals of Oncology

statistical methods previous data, equates to 2.5 units of hot flash score. Cohen identifies such
Methods used to analyze the data were similar to those used for a previous an effect as being of moderate size [15]. For each Wilcoxon rank-sum
dose-finding hot flash study of venlafaxine [14]. The primary end point comparison, the Hodges–Lehmann (HL) shift estimator (median of all
in this current study was change from baseline in hot flash score after pairwise differences) with 95% confidence interval (CI) is reported herein.
4 weeks of treatment. The hot flash score was computed for each patient Additional analyses included Kruskal–Wallis testing of the primary end
using data provided on the hot flash diary by assigning points (1 = mild, 2 = point across the three gabapentin arms (i.e. dose levels) followed by
moderate, 3 = severe, and 4 = very severe) to each hot flash based on pairwise comparisons using Wilcoxon rank-sum tests among the three
reported severity, summing the points for each day, and averaging the daily gabapentin arms. Additionally, the primary end point for each gabapentin
scores across each week of the study. arm was compared with the placebo arm, using Wilcoxon rank-sum tests.
A sample size of 50 patients per arm was required to provide at least 80% Secondary analyses included analysis of hot flash frequency similar to
power to detect a clinically meaningful difference in the primary end point that carried out using hot flash score, where a clinically meaningful
between the collective gabapentin arm (150 patients) and the placebo difference equates to 1 hot flash per day. CIs were constructed for the
arm (50 patients) with a two-sided 5% type I error rate using the Wilcoxon median reductions in hot flash score and frequency by arm, and plots
rank-sum test. For comparisons between any pair of arms, the power were created of average hot flash scores and frequencies (as percent of
was 66% for detecting the same clinically meaningful difference in the baseline) during this double-blind study by treatment arm. Additionally,
primary end point using a Wilcoxon rank-sum test with a two-sided 5% changes at week 4, from baseline values, in side-effects as measured by the
type I error rate. For this study, a clinically meaningful difference in the symptom experience diary, mood (POMS-B), hot flash distress, satisfaction
primary end point was defined as 0.5 standard deviations, which, based on with hot flash control, QoL, and hot flash effect on QoL tools were

223
randomized
patients

8 withdrew
prior to
receiving study
medication

215 started
study
medication

54 assigned to 53 assigned to 54 assigned to


54 assigned to
gabapentin gabapentin gabapentin
Placebo
300 mg/d 600 mg/d 900 mg/d

1 ineligible 46 with 42 with 43 with


due to use of evaluable hot evaluable hot evaluable hot
megestrol flash data for flash data for flash data for
acetate the 5 weeks the 5 weeks the 5 weeks

46 with
evaluable hot
flash data for
the 5 weeks

Figure 1. Consort diagram of patient flow in this trial.

544 | Loprinzi et al. Volume 20 | No. 3 | March 2009


Annals of Oncology original article
compared between the gabapentin arms and the placebo arm using 0 (0, 0), Wilcoxon P = 0.002; constipation HL (95% CI) =
Wilcoxon rank-sum tests. Two-sided P values £0.05 were considered 0 (0, 0), Wilcoxon P = 0.02], with the significant differences
statistically significant throughout. favoring the gabapentin arms (i.e. patients on the placebo arm
The impact of the missing data on conclusions was investigated using reported more trouble with both appetite loss and
imputation. Analyses were first carried out using all available data and then constipation). In comparing the highest gabapentin dose arm
carried out on datasets with missing values imputed using the last value to the placebo arm, the significant differences included
carried forward and the average value carried forward. Conclusions were
appetite loss [HL (95% CI) = 0 (0, 0), Wilcoxon P = 0.02),
consistent throughout. Results of the analyses using all available data are
fatigue [HL (95% CI) = 10 (0, 20), Wilcoxon P = 0.05), and
presented in the following section.
indigestion/belching [HL (95% CI) = 0 (0, 10), Wilcoxon P =
As confirmatory analyses to the prespecified primary and secondary
0.02), each, again favoring the gabapentin arm. The
analyses, hot flash scores (and frequencies) at week 4 were compared using
proportion of patients ending active treatment early due to
analysis of covariance with baseline scores (frequencies) as the covariate.
Results of these analyses were very similar to those of our primary and
side-effects was not significantly different in any of the four
secondary analyses (results not shown). study arms or in the combined gabapentin arms as compared
with the placebo arm (Fisher’s exact test; 5.9% versus 4.3%,
P = 0.69).
results Hot flash distress changes are illustrated in Figure 3. The
difference between the combined gabapentin arms and the
From 12 February 2002 to 9 November 2006, 223 patients were
placebo arm in change from baseline at week 4 was significant
entered on this clinical trial from 45 institutions in the United
for satisfaction with hot flash control [HL (95% CI) = 10 (0,
States. Eight patients canceled before receiving study treatment
20), Wilcoxon P = 0.03) and the effect of hot flashes on QoL
and one patient, on the placebo arm, was deemed ineligible due
[HL (95% CI) = 10 (0, 20), Wilcoxon P = 0.01), with the
to concomitant treatment with megestrol acetate, leaving 214
eligible patients. Baseline characteristics (Table 1) were well
balanced across the treatment arms. The flow of patients on this 120
A
trial is illustrated in Figure 1.
Changes from baseline hot flash scores and frequencies 100
(percent of baseline)
Hot flash frequency

during the fourth treatment week were available for 177 of 214 Placebo
80 300
(83%) patients—46 of 54 (85%), 46 of 54 (85%), 42 of 53 600
(79%), and 43 of 54 (80%) patients for the placebo arm and the 60 900
300, 600, and 900 mg/day gabapentin arms, respectively.
40
Changes in hot flash score from the baseline week over the four
treatment weeks are illustrated in Figure 2 and Table 2. The 20
primary comparison of change from baseline in hot flash score
0
after 4 weeks of treatment between the collective gabapentin
Baseline 1 2 3 4
arm and the placebo arm was not statistically significant [HL
Week
(95% CI) = 20.6 (22.3, 1.0); Wilcoxon P = 0.48]. However,
the comparisons of changes from baseline hot flash score and B 120
frequency at week 4 between the 900 mg/day gabapentin arm 100
(percent of baseline)

(the highest gabapentin dose arm) and the placebo were


Hot flash score

Placebo
associated with Wilcoxon rank-sum P values of 0.10 and 0.02 80
300
[HL (95% CI) of 21.9 (24.1, 0.3) and 21.4 (22.7, 20.3)], 600
60
respectively. P values for all pairwise Wilcoxon rank-sum 900
comparisons among the four treatment arms appear in Table 3. 40
There was no apparent difference in efficacy between sets of
20
patients grouped by whether they had 10 or more baseline hot
flashes per day, versus less. 0
With regard to side-effects, the only statistically significant Baseline 1 2 3 4
differences between the combined gabapentin arms and the Week
placebo arm were appetite loss and constipation [change
Figure 2. Mean changes from baseline for hot flash scores (A) and
during week 4, from baseline; appetite loss HL (95% CI) =
frequencies (B) for all study arms.

Table 2. Median percent decreases in hot flash score and frequency during treatment week 4, from baseline

Measure Median % change from baseline to week 4 (95% confidence interval)


Placebo 300 mg/day 600 mg/day 900 mg/day
Hot flash frequency 21.5 (11.3–30.9) 22.8 (12.1–33.0) 31.8 (16.5–40.5) 45.5 (31.1–50.6)
Hot flash score 27.0 (12.1–36.1) 29.7 (13.1–36.9) 33.8 (22.2–47.1) 44.4 (35.2–56.3)

Volume 20 | No. 3 | March 2009 doi:10.1093/annonc/mdn644 | 545


original article Annals of Oncology

Table 3. Wilcoxon rank-sum comparisons of change from baseline at week 4 for hot flash score and frequency

Placebo 300 mg/day 600 mg/day 900 mg/day Collective gabapentin Hodges–Lehmann Wilcoxon
(n = 46) (n = 46) (n = 42) (n = 43) (n = 131) shift estimator P value
Table values are median changes from baseline at week 4 (95% confidence
interval)
Hot flash score Collective gabapentin 22.8 23.3 20.6 (22.3, 1.0) 0.48
versus placeboa
300 mg/day 22.8 22.4 0.1 (21.7, 2.2) 0.80
versus placebo
600 mg/day 22.8 23.1 20.3 (22.6, 1.6) 0.72
versus placebo
900 mg/day 22.8 24.3 21.9 (24.1, 0.3) 0.10
versus placebo
300 mg/day 22.4 23.1 0.7 (21.3, 2.9) 0.44
versus 600 mg/day
300 mg/day 22.4 24.3 2.1 (0.0, 4.1) 0.05
versus 900 mg/day
600 mg/day 23.1 24.3 1.4 (20.9, 3.4) 0.19
versus 900 mg/day
Hot flash frequency Collective gabapentin 21.6 22.1 20.6 (21.6, 0.3) 0.19
versus placebo
300 mg/day 21.6 21.8 20.1 (21.3, 1.0) 0.75
versus placebo
600 mg/day 21.6 22.0 20.3 (21.6, 1.0) 0.60
versus placebo
900 mg/day 21.6 22.6 21.4 (22.7, 20.3) 0.02
versus placebo
300 mg/day 21.8 22.0 0.3 (21.0, 1.6) 0.73
versus 600 mg/day
300 mg/day 21.8 22.6 1.3 (0.1, 2.7) 0.03
versus 900 mg/day
600 mg/day 22.0 22.6 1.1 (20.3, 2.5) 0.10
versus 900 mg/day
a
Primary comparison.

25 Wilcoxon P = 0.003], with the significant differences again


Change in hot flash distress

900 favoring the gabapentin arm. Overall QoL (as changes during
20 week 4, from baseline) was not significantly different between
600
the gabapentin arm and the placebo arm [HL (95% CI) =
15
0 (0, 10), Wilcoxon P = 0.49] or between the highest dose of
10
gabapentin and the placebo arm [HL (95% CI) = 0 (0, 10),
300
Wilcoxon P = 0.42).
5 Placebo The differences between the combined gabapentin arms and
the placebo arm in changes during week 4, from the baseline
0 week, were insignificant (all Wilcoxon P > 0.05) for the
Baseline 1 2 3 4
POMS-B Total Mood Disturbance and all subscale scores
Week (tension/anxiety, depression/dejection, confusion/
Figure 3. Median changes from baseline in hot flash distress, for each bewilderment, fatigue/inertia, and anger/hostility) except for
study arm (higher scores are better, on a total scale of 100 points). vigor/activity [HL (95% CI) = 5 (0, 10), Wilcoxon P = 0.03],
with the significant difference in favor of the combined
significant differences favoring the gabapentin arms. The gabapentin arms. The difference between the highest
differences between the highest gabapentin dose arm and the gabapentin dose arm and the placebo arm in change during
placebo arm in changes in the week 4 values, from the baseline week 4, from the baseline week, was insignificant (all Wilcoxon
week, were significant for hot flash distress [HL (95% CI) = 10 P > 0.05) for the POMS-B Total Mood Disturbance and all
(0, 20), Wilcoxon P = 0.04), satisfaction with hot flash subscale scores except for depression/dejection [HL (95% CI) =
control [HL (95% CI) = 10 (0, 30), Wilcoxon P = 0.03), and 5 (0, 10), Wilcoxon P = 0.02), with the significant difference
the effect of hot flashes on QoL [(95% CI) = 10 (0, 20), again favoring the gabapentin arm.

546 | Loprinzi et al. Volume 20 | No. 3 | March 2009


Annals of Oncology original article
Compliance information, obtained from patient self- 51% reduction of hot flashes with gabapentin, compared with a
reports, appears in Table 4. During each week of treatment, 26% reduction with a placebo (P < 0.0001) [18].
compliance rates for patients who did not drop out of the The gabapentin was extremely well tolerated in this trial,
study were high (‡89% for each of the individual treatment without any evidence of side-effects being observed more than
regimens and ‡95% for the combination of all study what was seen in the placebo group. Similar results were seen in
arms) during this double-blind study. There were no a placebo-controlled trial looking at gabapentin for treatment
significant differences in compliance rates across the of chemotherapy neuropathy, with this trial utilizing up to 2700
gabapentin arms and matching placebo regimens at each mg/day in divided doses [19].
treatment week. The mechanism of action of gabapentin’s effect against hot
The proportion of patients choosing to continue on with flashes is not clear. It has been hypothesized that modulation of
gabapentin (or start taking gabapentin if initially on the placebo calcium currents may be involved [17]. Nonetheless, there is
arm) at the end of the double-blind study was 78%, 77%, 56%, not definitive information regarding the mechanism for its
and 62% in the placebo and three increasing gabapentin dose efficacy.
arms. It is worth providing a few words with regards to the
The placebo arm was broken up into three subsets, based on methodology of this trial. The utilized methodology has been
whether the target dose was 1, 2, or 3 tablets per day. When validated [13] and has been used in a series of Mayo Clinic/
comparing these three arms to each other, there were no NCCTG clinical trials involving both women and men with
substantial differences between them in terms of hot flash hot flashes [14, 20–25]. A similar methodology has also been
scores (which, during the fourth treatment week were 78%, used by investigators in other hot flash trials [16, 26, 27].
80%, and 72% of the baseline week in the three subsets, With regards to trial length, it might be argued that the
respectively), toxicity, or compliance (Table 4). duration of treatment in this trial (4 weeks) is shorter than
what has been utilized in other trials evaluating hot flash
treatment regimens. Nonetheless, trials that investigated
discussion nonhormonal agents for hot flashes over 8–12 weeks nicely
The efficacy of gabapentin seen in this current trial parallels the demonstrate that there is a plateau effect on the curves from
efficacy seen with similar doses in women with hot flashes, with 4 weeks on to 6–12 weeks [16, 17, 25–27].
each of these revealing an 50% reduction in hot flashes at Another methodology question deals with the placebo
a gabapentin dose of 300 mg given three times daily [16, 17]. program that was utilized in this trial. The placebo arm could
A cross study comparison of the results from this current trial be broken up into three subsets, based on whether the target
and the results of the two trials in women with 900 mg of dose was 1, 2, or 3 tablets per day. One might argue that the
gabapentin per day is illustrated in Figure 4, demonstrating patients getting more tablets per day might demonstrate greater
similarities among the three different trials. Recent data from placebo or nocebo effects or be less compliant. Nonetheless,
another trial in women also revealed very similar results, with a the results of this trial did not illustrate a difference in efficacy,

Table 4. Patient-reported compliance: the placebo arms are designated as those that had target doses of 1, 2, or 3 tablets per day to correspond to
gabapentin doses of 300, 600, and 900 mg/day, respectively

Placebo (1/day) Placebo (2/day) Placebo (3/day) 300 mg/day 600 mg/day 900 mg/day Total P valuea
Compliance week 1 0.21
Missing 0 0 0 2 1 3 6
Yes 17 (100%) 15 (94%) 16 (94%) 49 (100%) 42 (89%) 43 (96%) 182 (95%)
No 0 (0%) 1 (6%) 1 (6%) 0 (0%) 5 (11%) 2 (4%) 9 (5%)
Compliance week 2 0.31
Missing 0 0 0 1 1 2 4
Yes 16 (100%) 15 (94%) 17 (100%) 49 (100%) 44 (98%) 44 (100%) 185 (99%)
No 0 (0%) 1 (6%) 0 (0%) 0 (0%) 1 (2%) 0 (0%) 2 (1%)
Compliance week 3 0.33
Missing 0 0 0 2 0 3 5
Yes 16 (100%) 14 (93%) 17 (100%) 48 (98%) 42 (93%) 43 (100%) 180 (97%)
No 0 (0%) 1 (7%) 0 (0%) 1 (2%) 3 (7%) 0 (0%) 5 (3%)
Compliance week 4 0.16
Missing 0 1 0 6 2 3 12
Yes 15 (100%) 13 (93%) 17 (100%) 44 (100%) 39 (91%) 41 (98%) 169 (97%)
No 0 (0%) 1 (7%) 0 (0%) 0 (0%) 4 (9%) 1 (2%) 6 (3%)

Compliant patient status was defined by the patient stating that he took the correct number of daily pills, with the given percentage applying to all patients
that answered that question.
a
Each week is a 2 · 6 two-way table [compliance (yes, no) versus arm (placebo 1/day, placebo 2/day, placebo 3/day, 300 mg/day, 600 mg/day, 900 mg/day)]
and each P value is for a chi-squared test with 5 degrees of freedom (for the week).

Volume 20 | No. 3 | March 2009 doi:10.1093/annonc/mdn644 | 547


original article Annals of Oncology

Men current funding


900 mg/d P=0.10
Placebo Public Health Service (CA-25224, CA-37404, CA-35103, CA-
Guttuso et al
63849, CA-63848, CA-35195, CA-35272, CA-35269, CA-35101,
900 mg/d CA-60276, CA-52352, CA-37417, CA-35448); National Cancer
P=0.02
Placebo Institute (U10CA037404).
Pandya et al
900 mg/d
P=0.0001
acknowledgements
Placebo
The content is solely the responsibility of the authors and does
0 20 40 60 80 100 120 not necessarily represent the official views of the National
Residual hot flash score at 4 weeks (%) Cancer Institute or the National Institutes of Health. This study
was conducted as a collaborative trial of the North Central
Figure 4. Cross study comparisons of data from two studies of gabapentin Cancer Treatment Group and Mayo Clinic. Additional
in women [16, 17] and the current one in men. Error bars represent 95% participating institutions include Iowa Oncology Research
confidence intervals. Association CCOP, Des Moines, IA 50314 (Roscoe F. Morton,
MD); Rapid City Regional Oncology Group, Rapid City, SD
59709 (Richard C. Tenglin, MD); CentraCare Clinic, St Cloud,
presumed toxicity, or compliance among the three subsets of
MN 56301 (Harold E. Windschitl, MD); Quain and Ramstad
the placebo arm (Figure 4).
Clinic, Bismarck, ND 58506 (Edward J. Wos, DO); Sioux
It is reasonable to ask whether this current project should be
Community Cancer Consortium, Sioux Falls, SD 57105 (Loren
considered as a positive clinical study. On the one hand, it
K. Tschetter, MD); Missouri Valley Cancer Consortium,
could be argued that the primary analysis written in the
Omaha, NE 68106 (Gamini S. Soori, MD); Siouxland
statistical section of this protocol, that being a comparison of
Hematology-Oncology Associates, Sioux City, IA 51105
the combined gabapentin arms to the placebo arm, did not
(Donald B. Wender, MD); Wichita Community Clinical
reveal a statistically significant difference, and thus this is
Oncology Program, Wichita, KS 67214-3882 (Shaker R. Dakhil,
a negative clinical study. On the other hand, it can easily be
MD); Illinois Oncology Research Assn. CCOP, Peoria, IL
argued that this is not the best analytical means to look at
61615-7828 (John W. Kugler, MD); Montana Cancer
a study such as this. It was expected that there was likely to be
Consortium, Billings, MT 59101 (Benjamin T. Marchello, MD);
a dose–response effect for the chosen low doses of gabapentin
Cedar Rapids Oncology Project CCOP, Cedar Rapids, IA 52403
and that only the higher doses might have shown a positive
(Martin Wiesenfeld, MD); Allegheny General Hospital,
result, as was seen in another similarly designed clinical trial
Pittsburgh, PA 15212 (Jane M. Raymond, MD); Columbus
conducted by the NCCTG [14] and in a placebo-controlled
CCOP, Columbus, OH 53215 (J. Philip Kuebler, MD, PhD);
double-blind, randomized trial in women examining
Hawaii Minority-Based CCOP (William S. Loui, MD); and
gabapentin at 300 and 900 mg/day [16]. This was, in fact, what
Howard University Cancer Center, Washington, DC 20060
was observed.
(Lucile Adams-Campbell, PhD).
Facts that support that gabapentin moderately decreases
hot flashes in men include a statistically significant comparison
of hot flash frequencies and a trend for improvement in hot references
flash scores between the 900 mg/day and placebo arms;
1. Schow DA, Renfer LG, Rozanski TA, Thompson IM. Prevalence of hot flushes
statistically significant improvements of hot flash frequencies during and after neoadjuvant hormonal therapy for localized prostate cancer.
and scores between the 900 mg/day and 300 mg/day arms; that South Med J 1998; 91: 855–857.
dose–response relationships are apparent in Figures 1 and 2; 2. Charig CR, Rundle JS. Flushing. Long-term side effect of orchiectomy in
that the results in men look so similar to what was seen in three treatment of prostatic carcinoma. Urology 1989; 33: 175–178.
trials in women [16–18]; that there were significant differences 3. Loprinzi C, Goldberg RM, O’Fallon JR et al. Transdermal clonidine for
for satisfaction with hot flash control and the effect of hot ameliorating postorchiectomy hot flashes. J Urol 1993; 151: 634–636.
flashes on QoL for the combined gabapentin arms compared 4. Loprinzi CL, Michalak JC, Quella SK et al. Megestrol acetate for the prevention of
with the placebo arm, and that there were significant differences hot flashes. N Engl J Med 1994; 331: 347–352.
between the highest gabapentin dose arm and the placebo 5. Atala A, Amin M, Harty JI. Diethylstilbestrol in treatment of postorchiectomy
arm in changes in the week 4 values, from the baseline week, for vasomotor symptoms and its relationship with serum follicle-stimulating
hot flash distress, satisfaction with hot flash control, and the hormone, luteinizing hormone, and testosterone. Urology 1992; 39: 108–110.
effect of hot flashes on QoL. 6. Burch PA, Loprinzi CL. Prostate-specific antigen decline after withdrawal of low-
Thus, it appears reasonable to utilize gabapentin in clinical dose megestrol acetate. J Clin Oncol 1999; 17: 1087–1088.
practice for hot flashes in men, noting that it would be ideal 7. Guttuso TJ Jr. Gabapentin’s effects on hot flashes and hypothermia. Neurology
to have further data to substantiate its benefit. Nonetheless, 2000; 54: 2161–2163.
the magnitude of benefit with this therapy appears to be 8. Pandya KJ, Thummala AR, Griggs JJ et al. Pilot study using gabapentin for
relatively moderate. More information about gabapentin’s tamoxifen-induced hot flashes in women with breast cancer. Breast Cancer Res
ability to relieve hot flashes in men is desirable. Better therapies Treat 2004; 83: 87–89.
for alleviating hot flashes in men on androgen deprivation 9. Loprinzi C, Barton D, Sloan J et al. Pilot evaluation of gabapentin for treating hot
therapy are still needed. flashes. Mayo Clin Proc 2002; 77: 1155–1163.

548 | Loprinzi et al. Volume 20 | No. 3 | March 2009


Annals of Oncology original article
10. Pocock SJ, Simon R. Sequential treatment assignment with balancing for 20. Goldberg RM, Loprinzi CL, O’Fallon JR et al. Transdermal clonidine for
prognostic factors in the controlled clinical trial. Biometrics 1975; 31: 103–115. ameliorating tamoxifen-induced hot flashes. J Clin Oncol 1994; 12: 155–158.
11. McNair DM, Loor M, Droppleman LF. Profile of Mood States Manual. New York: 21. Barton DL, Loprinzi CL, Quella SK et al. Prospective evaluation of vitamin E for
Multi-Health Systems 2003. hot flashes in breast cancer survivors. J Clin Oncol 1998; 16: 495–500.
12. Quella S, Loprinzi CL, Dose AM. A qualitative approach to defining ‘‘hot flashes’’ 22. Quella S, Loprinzi C, Barton D. Evaluation of soy phytoestrogens for the
in men. Urol Nurs 1994; 14: 155–158. treatment of hot flashes in breast cancer survivors. J Clin Oncol 1999; 16:
13. Sloan JA, Loprinzi CL, Novotny PJ et al. Methodologic lessons learned from hot 495–500.
flash studies. J Clin Oncol 2001; 19: 4280–4290.
23. Loprinzi CL, Sloan JA, Perez EA et al. Phase III evaluation of fluoxetine for
14. Loprinzi CL, Kugler JW, Sloan JA et al. Venlafaxine in management of hot flashes
treatment of hot flashes. J Clin Oncol 2002; 20: 1578–1583.
in survivors of breast cancer: a randomised controlled trial. Lancet 2000; 356:
2059–2063. 24. Pockaji B, Gallagher J, Loprinzi C et al. Phase III double-blinded, randomized trial
to evaluate the use of black cohosh in the treatment of hot flashes: a North
15. Cohen J. Statistical Power Analysis for the Behavioral Sciences. Hillsdale, NJ:
Central Cancer Treatment Group Study. Proc Am Soc Clin Oncol 2005; 23:
Lawrence Erlbaum Associates, Inc 1988.
732S.
16. Pandya KJ, Morrow GR, Roscoe JA et al. Gabapentin for hot flashes in 420
women with breast cancer: a randomised double-blind placebo-controlled trial. 25. Loprinzi C, Levitt R, Sloan J et al. Medroxyprogesterone acetate (MPA) versus
Lancet 2005; 366: 818–824. venlafaxine for hot flashes: a North Central Cancer Treatment Group Trial.
17. Guttuso T Jr., Kurlan R, McDermott MP, Kieburtz K. Gabapentin’s effects on hot 2005 Annual Meeting of the American Society of Clinical Oncol. J Clin Oncol
flashes in postmenopausal women: a randomized controlled trial. Obstet Gynecol 2005 vol 23, No 16s:732s (Abstr 8014).
2003; 101: 337–345. 26. Stearns V, Beebe KL, Iyengar M, Dube E. Paroxetine controlled release in the
18. Butt DA, Lock M, Lewis JE et al. Gabapentin for the treatment of menopausal hot treatment of menopausal hot flashes: a randomized controlled trial. JAMA 2003;
flashes: a randomized controlled trial. Menopause 2008; 15: 310–318. 289: 2827–2834.
19. Rao RD, Michalak JC, Sloan JA et al. Efficacy of gabapentin in the management of 27. Stearns V, Slack R, Greep N et al. Paroxetine is an effective treatment for hot
chemotherapy-induced peripheral neuropathy: a phase 3 randomized, double-blind, flashes: results from a prospective randomized clinical trial. J Clin Oncol 2005;
placebo-controlled, crossover trial (N00C3). Cancer 2007; 110: 2110–2118. 23: 6919–6930.

Volume 20 | No. 3 | March 2009 doi:10.1093/annonc/mdn644 | 549


Author's Accepted Manuscript

Prophylactic Sildenafil Citrate Improves Selected Aspects of Sexual Function in


Men Treated by Radiotherapy for Prostate Cancer

Michael J. Zelefsky, Daniel Shasha, Rebekah Dunn Branco, Marisa Kollmeier,


Raymond E. Baser, Xin Pei, Ronald Ennis, Richard Stock, Natan Bar-Chama, John
P. Mulhall

PII: S0022-5347(14)00355-3
DOI: 10.1016/j.juro.2014.02.097
Reference: JURO 11150

To appear in: The Journal of Urology


Accepted Date: 21 February 2014

Please cite this article as: Zelefsky MJ, Shasha D, Branco RD, Kollmeier M, Baser RE, Pei X, Ennis R,
Stock R, Bar-Chama N, Mulhall JP, Prophylactic Sildenafil Citrate Improves Selected Aspects of Sexual
Function in Men Treated by Radiotherapy for Prostate Cancer, The Journal of Urology® (2014), doi:
10.1016/j.juro.2014.02.097.

DISCLAIMER: This is a PDF file of an unedited manuscript that has been accepted for publication. As a
service to our subscribers we are providing this early version of the article. The paper will be copy edited
and typeset, and proof will be reviewed before it is published in its final form. Please note that during the
production process errors may be discovered which could affect the content, and all legal disclaimers
that apply to The Journal pertain.

All press releases and the articles they feature are under strict embargo until uncorrected proof of
the article becomes available online. We will provide journalists and editors with full-text copies of the
articles in question prior to the embargo date so that stories can be adequately researched and written.
The standard embargo time is 12:01 AM ET on that date.
ACCEPTED MANUSCRIPT

Original Article

Prophylactic Sildenafil Citrate Improves Selected Aspects of Sexual Function in Men Treated

by Radiotherapy for Prostate Cancer

PT
Michael J. Zelefsky, MD,* Daniel Shasha, MD, Rebekah Dunn Branco, MA, MBA,

Marisa Kollmeier, MD, Raymond E. Baser, MS, Xin Pei, PhD, Ronald Ennis, MD, Richard

RI
Stock, MD, Natan Bar-Chama, MD, and John P Mulhall, MD, MSc, FECSM, FACS

SC
From The Department of Radiation Oncology and Urology (MJZ, RBD, MK, XP, JPM),

U
Memorial Sloan-Kettering Cancer Center, the Department of Radiation Oncology (DS, RE),
AN
Continuum Cancer Center of New York, the Department of Radiation Oncology and Urology

(RS, NB-C), Mt Sinai Medical Center, and the Department of Epidemiology and Biostatistics
M

(REB), Memorial Sloan-Kettering Cancer Center, New York, New York


D
TE

* Correspondence: Michael J. Zelefsky, MD, Memorial Sloan-Kettering Cancer Center, 1275

York Avenue, New York, NY 10065. Phone: 212-639-6802. Fax: 212-639-8876. E-mail:
EP

zelefskm@mskcc.org.
C

Number of words: 2594


AC

Running Title: Prophylactic Sildenafil for ED after RT

Key Words: Prostate Cancer, Radiotherapy, Sildenafil Citrate, Erectile Dysfunction

1
ACCEPTED MANUSCRIPT

Funding/Support: Pfizer provided an educational grant that was used for research

assistance support and supplied drug for the study.

PT
RI
U SC
AN
M
D
TE
C EP
AC

2
ACCEPTED MANUSCRIPT

ABSTRACT

Purpose: Adjuvant daily sildenafil citrate (SC) during and after radiotherapy for prostate cancer

for erectile function preservation was studied.

Methods: A randomized, prospective trial of 279 patients with localized prostate cancer treated

PT
with radiotherapy compared daily SC (50 mg) to placebo (2:1 randomization).

RI
Medication/placebo was initiated 3 days pretreatment and continued daily for 6 months. Patients

completed the International Index of Erectile Function (IIEF) including the erectile function

SC
domain (EF) and International Prostate Symptom Score questionnaires pretherapy and the Rand

36-Item Health Survey at 3, 6, 9, 12, 18, and 24 months post-radiotherapy. All IIEF domains

were scored.
U
AN
Results: At 12 months, EF scores were better for SC (P=0.018) than placebo, 73% of SC vs 50%

of placebo patients had mild/no erectile dysfunction (ED) (P=0.024), and the SC arm had
M

superior overall satisfaction (P=0.028) and IIEF total scores (P=0.044). At 24 months, EF and
D

IIEF scores were no longer significantly better for SC (P=0.172 and 0.09, respectively), yet
TE

overall satisfaction scores were higher (P=0.032). SC sexual desire scores were higher (P=0.049)

at 24 months despite completing drug therapy 18 months prior. At 24 months, 81.6% of SC and
EP

56.0% of placebo patients (P=0.045) had functional erections (with or without ED medication).

Conclusions: Daily SC during and after radiotherapy for prostate cancer was associated with
C

improved overall sexual function compared with placebo for various sexual function domains.
AC

This is the largest randomized, prospective, controlled trial to demonstrate utility of a PDE5

inhibitor as a rehabilitation strategy in prostate cancer radiation therapy patients.

3
ACCEPTED MANUSCRIPT

ERECTILE dysfunction (ED) is recognized as a significant treatment-related complication after

definitive treatment for prostate cancer such as surgery and radiotherapy (RT). ED is a significant

concern for patients as they select therapy, and can negatively impact overall quality of life. After

RT, the overall reported ED incidence is 24-59%, and sexual function often begins to decline only

PT
12-18 months following completion of therapy.[1]

RI
The importance of penile rehabilitation after prostate cancer therapy has been evaluated

by several investigators [2-6], based on recognition that preservation of corpus cavernosal

SC
endothelial and smooth muscle integrity might improve long-term sexual function. Several

animal studies have demonstrated a positive effect for regular use of a PDE5i such as sildenafil

U
citrate (SC) through neuro- and vascular-protective effects on tissues involved in erectile
AN
function.[7] In animals chronically treated with PDE5i, reduced deposition of collagen within the

corpora cavernosa and endothelial cell preservation has been observed in penile tissues.[8]
M

Clinical studies suggest a benefit to rehabilitation in radical prostatectomy (RP) patients. In


D

one retrospective case-matched study,[4] RP patients were given early SC within 6 months of

surgery or after 6 months from surgery. Improved erectile function was noted post-therapy
TE

among early-SC patients compared with historical controls (58% vs 30%). In a randomized trial
EP

evaluating penile rehabilitation among RP patients, 628 men were randomized to receive

placebo, nightly vardenafil, or on-demand vardenafil. The study had a 9-month double-blind
C

treatment period, and the primary outcome measure was the percentage of subjects with IIEF-EF
AC

score ≥22. No statistically significant differences were observed between groups.[5]

For RP patients, a significant component of post-treatment ED is related to neural injury,

but the etiology of post-RT function appears to be more related to vascular (endothelial)

dysfunction.[9] We hypothesized that, in RT patients, adjuvant SC may demonstrate more

significant improvements given its recognized vascular-protective effects. In one study that was

4
ACCEPTED MANUSCRIPT

grossly underpowered, including 27 patients randomized, no benefits were observed for the use

of prophylactic SC.[10] To prospectively study the impact of prophylactic SC among prostate RT

patients with pretherapy functional erections, we undertook the current study. The primary

objective was to determine if there is a role for penile rehabilitation among potent patients

PT
receiving definitive RT for prostate cancer, specifically if prophylactic daily SC during and after

RI
RT can preserve sexual function 24 months from the start of RT.

SC
MATERIALS AND METHODS

This is a double-blind, placebo-controlled trial to assess whether concurrent and adjuvant SC in

U
conjunction with RT for clinically localized prostate cancer will improve sexual function
AN
following RT at 24 months post-treatment.
M

Patient Population
D

Two hundred seventy-nine patients with organ-confined prostate cancer were randomized.
TE

Eligible patients included those who were to receive definitive treatment with external beam

radiation therapy (EBRT), brachytherapy, or brachytherapy combined with EBRT, who also had
EP

a baseline International Index of Erectile Function (IIEF) score ≥17. Following enrollment of

≈50 patients, eligibility criteria were expanded to broaden accrual, allowing patients receiving
C

neo-adjuvant androgen-deprivation therapy (ADT), starting within 1 month prior to enrollment.


AC

These patients were included to determine if a benefit would be observed despite the sexual

depressive effects associated with ADT.

Patients using erectogenic agents >4 times per month or those taking 0.8 mg of

tamsulosin daily pretherapy were excluded. Also excluded were patients with a clinically

5
ACCEPTED MANUSCRIPT

significant penile deformity, a history of penile implant, or prior history of prostate cancer

therapy, or if SC was contraindicated. The patients were enrolled across three institutions. There

were 279 randomized patients and 202 initiated treatment with the study drug or placebo,

including 218 and 162, respectively, at Memorial Sloan-Kettering Cancer Center (MSKCC); 48

PT
and 30 at Continuum Cancer Center; and 13 and 10 at Mt Sinai School of Medicine.

RI
This research protocol was approved by the institutional review board from each

participating institution and all participants provided written informed consent prior to

SC
enrollment.

Those not taking the study drug either decided against RT following randomization or

U
otherwise withdrew without taking the first dose. Table 1 provides baseline demographic,
AN
disease, and comorbidity information for participants who initiated therapy. The median EBRT

dose was 86.4 Gy when given as monotherapy and 50.4 Gy when given with brachytherapy.
M

There were no significant differences between the sildenafil and placebo groups as far as the
D

prescription dose levels delivered to the prostate (i.e. percentage who underwent EBRT alone
TE

versus BRT or combined modality treatment). When ADT was used, luteinizing hormone-

releasing hormone agonist in a neoadjuvant and concurrent setting was often given in
EP

combination with anti-testosterone therapy, and in general ADT was discontinued after

completion of RT.
C
AC

Treatment

Patients were randomized 2:1 to receive daily orally administered study therapy with SC 50 mg

or placebo. For ADT patients, study drug was initiated simultaneously with or within 1 month

after starting of ADT. Participants not receiving ADT began study therapy between 3 days prior

6
ACCEPTED MANUSCRIPT

to starting RT through 2 weeks following the first fraction of radiation or implantation of

brachytherapy. All participants were instructed to continue daily administration of SC or placebo

until 6 months following the start of RT. Patients were asked to keep a daily pill diary to

document compliance.

PT
RI
Assessments and Follow-up

Participants were evaluated for changes in sexual function, urinary symptoms, quality-of-life

SC
measures, and toxicities for the duration of study therapy and for 2 years after initiation of RT.

Urinary symptoms and quality of life were measured through patient-reported questionnaires

U
(International Prostate Symptom Score (IPSS), and the Rand 36-Item Health Survey (QOL),
AN
respectively). Sexual function was assessed by the IIEF-6 and by patient self-reports at follow-up

visits. IIEF domains were evaluated and scored for each treatment, including Erectile Function
M

(EF), Orgasmic Function (OF), Sexual Desire (SD), Intercourse Satisfaction (IS), and Overall
D

Satisfaction (OS). Patient characteristics including age, use of brachytherapy, use of ADT, and
TE

baseline IIEF scores were balanced between treatment groups.

Participants were assessed for these outcomes and quality-of-life measures at baseline
EP

and 3, 6, 9, 12, 18, and 24 months following start of RT. Treatment-related toxicities were

assessed throughout the course of therapy. Baseline assessments occurred in the 4-week interval
C

prior to starting study drug.


AC

Response Criteria

The primary objective was to determine if prophylactic SC used during and after a course of RT

(EBRT and/or brachytherapy with or without ADT) can prevent loss of spontaneous erectile

7
ACCEPTED MANUSCRIPT

function 2 years from the start of RT, evaluated based on responses to the IIEF at 2 years post-

RT. We also assessed the response among only patients not treated with ADT at both 1 and 2

years. A four-point change in IIEF score vs baseline was considered significant in erectile

function.[11] An additional analysis was performed on patients treated at MSKCC, where the

PT
information was more readily available to assess erectile function based on participant self-report

RI
at regular clinical follow-ups, and graded according to the Common Terminology Criteria for

Adverse Events (CTCAE) v 3.0. Participants with grade 0-1 ED were considered fully

SC
functional, grade 2 ED partially potent, and grade 3 ED non-functional.

Acute and late urinary side effects were assessed by IPSS scores and participant self-

U
report in the clinic. Acute toxicity was defined as any toxicity occurring within 4 months from
AN
the completion of therapy. Late toxicities were defined as those observed after 4 months
M

following completion of therapy.


D

Statistical Methods

Outcome analysis was based on the modified intention-to-treat principle in which data from all
TE

randomized patients who received RT were analyzed. Baseline demographic and medical
EP

characteristics of the treatment groups were compared using χ2 tests for categorical variables and

t-tests for continuous variables.


C

Average IIEF total and subscale scores and IPSS total scores were summarized for each
AC

treatment arm at baseline and each follow-up assessment time (e.g., 12-months, 24-months, etc.)

using median values. Score variability was summarized by the first and third quartiles of the

score distributions. Wilcoxon rank sums tests assessed statistical differences between study arms

in their score distributions at each assessment time. Differences were evaluated with two-sided

tests at baseline and one-sided tests at each follow-up, with the one-sided alternative hypothesis

8
ACCEPTED MANUSCRIPT

that SC patients had more functional scores than placebo patients. Proportions of MSKCC

patients potent at clinical follow-ups corresponding to the study assessment times, and

proportions of patients experiencing acute and late GU toxicities were compared using Fisher

exact tests. For each scale, effect of treatment arm on scores assessed at 12 months and later,

PT
controlling for baseline score and age, was evaluated using linear regression models estimated

RI
with generalized estimating equations accounting for multiple scores per patient (i.e., at 12, 18,

and 24 months). For all tests, P≤0.05 was considered statistically significance, while P≤0.10 was

SC
considered marginally statistically significant.

U
RESULTS
AN
Among patients treated with or without ADT, SC patients tended to have higher EF and IIEF total

scores at 24 months than placebo patients, but these differences were not significant (Table 2). SC
M

patients had significantly higher OS subscale scores at 24 months (P=0.048).

As shown in Table 3, ADT patients tended to experience worse erectile function outcomes
D

than non-ADT patients (Table 4), regardless of treatment arm. Additionally, the IIEF total and
TE

subscale scores among the ADT patients were considerably more variable at each time-point

than among non-ADT patients. These data suggest that sexually depressive effects of ADT
EP

mitigated potential benefits of prophylactic SC.


C

For the rest of the analysis we focused on the overwhelming majority of patients not
AC

receiving ADT (90%). In these there were no significant erectile function differences between

treatment arms at baseline. In general, SC patients had significantly better scores for all domains

at 3 and 6 months post-therapy, with these differences diminishing with time (Table 5, Figs 1 and

2). However, significant differences persisted at 12 and 24 months. At 12 months, EF scores of

SC patients (median, 26.0; IQR, 19.3-29.0) were significantly better (P=0.018) than those of

9
ACCEPTED MANUSCRIPT

placebo patients (21.5, 14.0-28.0), and 73% of SC patients had only mild or no ED by EF score

vs 50% of placebo patients (P=0.024). The SC arm reported superior overall satisfaction (OS)

subscale (P=0.028) and IIEF total scores (P=0.044) at 12 months. EF scores were no longer

significantly better at 24 months for SC patients (P=0.172), but OS scores were significantly

PT
higher (P=0.032) and IIEF scores marginally but not significantly higher (P=0.097). SC patients

RI
also experienced significantly higher sexual desire (SD) scores at 24 months (P=0.049)

compared with placebo despite having completed drug therapy 18 months prior.

SC
At 3 and 6 months, 91.4% and 78.4% of SC patients reported functional erections and were

not taking any medications for ED at their physician visits vs 63.6% and 47.6% for placebo (p =

U
0.015 and p = 0.022, respectively). At 24 months, 81.6% of SC and 56.0% of placebo patients (p =
AN
0.045) reported functional erections (with or without ED medication). We observed no differences

in erectile function outcomes among EBRT patients compared with brachytherapy or a combined-
M

modality treatment.
D

There were no significant differences in acute or late treatment-related toxicities between

the groups. Among SC patients, 20.7% had no acute GU toxicity, 52.6% had grade 1 and 26.7%
TE

had grade 2-3 acute GU toxicities vs 11.7% of control patients with no toxicity, 54.5% with
EP

grade 1, and 33.8% with grade 2-3 acute GU toxicity (P=0.194). 39.7%, 41.3%, and 19.0% of SC

patients experienced no, grade 1, and grade 2-3 late toxicities, respectively, compared with
C

11.7%, 54.5%, and 33.8% of control patients, respectively (P=0.576). The two study arms also
AC

had similar IPSS scores at baseline and at each follow-up study assessment. Drug-related

symptoms were more often reported in the SC group compared with the placebo cohort.

Specifically, SC patients more often complained of dizziness (11% vs 0%), headaches (18% vs

5%), blurred vision (6% vs 5%), and flushing (17% vs 8%).

10
ACCEPTED MANUSCRIPT

DISCUSSION

Our findings are the first indications in a randomized trial of evidence for a benefit using

prophylactic SC in prostate cancer patients treated with RT. We observed significant

improvements in various sexual function domains at 6 and 12 months post-therapy, and

PT
specifically for OS and SD. In addition, these differences persisted as far as 24 months after

RI
therapy. We view these important findings as proof of the principle that prophylactic SC can play

a critical role in erectile function preservation for prostate cancer patients treated with RT. The

SC
greatest improvement was at 6 and 12 months after treatment, suggesting that our 6-month usage

of prophylactic SC may not be of adequate duration. At 24 months however the differences were

U
less pronounced yet with persistently improved desire and satisfaction in the SC arm noted, and
AN
at 24 months, 82% of SC and 56.0% of placebo patients (p = 0.045) reported functional erections

(with or without ED medication)..A longer course of SC (> 6 months) may be required to provide
M

even better functional outcomes beyond 12 months from therapy. Only further prospective
D

studies will clarify this issue.


TE

Our results support the concept that PDE5 inhibitors might limit the damage radiation

inflicts on vascular tissue in cavernosal tissue. Desouza et al published data that unequivocally
EP

demonstrated the positive effect of sildenafil on endothelial function.[12] In patients with ED

and type 2 diabetes, sildenafil improved flow-mediated dilation. This has also been demonstrated
C

using tadalafil.[13] The duration of effect of these agents on endothelial function was shown to
AC

be longer than the presence of either drug within the blood, suggesting that the impact of PDE5

inhibitors on endothelium at the cellular level may be self-sustaining. Furthermore, all

commercially available PDE5i have been shown to generate endothelial progenitor cells from the

bone marrow,[14] suggesting another mechanism by which PDE5i may protect endothelium. As

patients in this study were not prospectively followed with penile duplex Doppler studies, we

11
ACCEPTED MANUSCRIPT

could not validate our hypothesis that the erectile function improvements we observed with SC

was related to improved vascular function. We recognize a limitation of this study is that not all

patients underwent serial testosterone measurements which could have detected in some

testosterone deficiency syndrome. Nevertheless, we would not expect, in the non-hormonal

PT
cohort of patients (where the greatest benefit of prophylactic SC was observed) any differences

RI
in testosterone reductions as a result of localized radiotherapy to the prostate.

PDE5i have been used as the backbone to penile rehabilitation interventions following

SC
RP.[4] While definitive data does not yet exist proving their utility in this population, two of

three randomized, placebo-controlled trials have demonstrated a signal that regular PDE5i use

U
results in improved rates of men returning to their baseline preoperative erectile function
AN
(sildenafil)[15] and earlier return to PDE5i response (tadalafil).[16] Therefore, it seems

reasonable to explore such agents in RT patients to limit the negative effects of RT on


M

endothelium and improve erectile function preservation. Schiff et al analyzed patients who
D

underwent prostate brachytherapy who subsequently used PDE5 inhibitors on a regular


TE

basis.[17] Patients were stratified into early-use (<1 year post-brachytherapy) and late-use groups

(>1 year post-brachytherapy). Higher rates of erectile function preservation, based on sexual
EP

health inventory for men (SHIM) scores, were seen in the early-use group.

Our study also suggests that prophylactic SC was not as beneficial among patients treated
C

with ADT in conjunction with RT. Due to the limited number of these patients in our study, it is
AC

difficult to draw conclusions about the value of penile rehabilitation among patients with a

depressed libido as a result of ADT. Further studies in this population are warranted and longer

duration of adjuvant usage well beyond testosterone recovery may be necessary to achieve more

optimal outcomes.

12
ACCEPTED MANUSCRIPT

CONCLUSIONS

Our findings indicate that daily use of SC during and for 6 months after initiation of

prostate cancer RT is associated with improved sexual function outcomes. Patients reported

PT
significantly improved sexual functions for SC compared to placebo. Radiotherapy-induced

RI
trauma to the vascular supply of the cavernosal tissue may undergo rehabilitation and repair

during administration of SC. Longer SC usage post-RT may further improve sexual function and

SC
will require further prospective studies.

U
AN
M
D
TE
C EP
AC

13
ACCEPTED MANUSCRIPT

Abbreviations and Acronyms

ADT = androgen-deprivation therapy

EBRT = external beam radiation therapy

ED = erectile dysfunction

PT
EF = erectile function

RI
GU = genitourinary

IIEF = International Index of Erectile Function

SC
IPSS = International Prostate Symptom Score

IQR = interquartile range

IS = intercourse satisfaction
U
AN
MSKCC = Memorial Sloan-Kettering Cancer Center

OF = orgasmic function
M

OS = overall satisfaction
D

PSA = prostate-specific antigen


TE

Q1 = first quartile

Q3 = third quartile
EP

QOL = quality of life

RP = radical prostatectomy
C

RT = radiotherapy
AC

SC = sildenafil citrate

SD = sexual desire

14
ACCEPTED MANUSCRIPT

REFERENCES

1. van der Wielen GJ, Mulhall JP, Incrocci L (2007) Erectile dysfunction after radiotherapy

for prostate cancer and radiation dose to the penile structures: a critical review. Radiother Oncol

84:107-113

PT
2. Montorsi F, Guazzoni G, Strambi LF, Da Pozzo LF, Nava L, Barbieri L, Rigatti P,

RI
Pizzini G, Miani A (1997) Recovery of spontaneous erectile function after nerve-sparing radical

retropubic prostatectomy with and without early intracavernous injections of alprostadil: results

SC
of a prospective, randomized trial. J Urol 158:1408-1410

3. Mulhall J, Land S, Parker M, Waters WB, Flanigan RC (2005) The use of an erectogenic

U
pharmacotherapy regimen following radical prostatectomy improves recovery of spontaneous
AN
erectile function. J Sex Med 2:532-540; discussion 540-532

4. Mulhall JP, Parker M, Waters BW, Flanigan R (2010) The timing of penile rehabilitation
M

after bilateral nerve-sparing radical prostatectomy affects the recovery of erectile function. BJU
D

Int 105:37-41
TE

5. Montorsi F, Brock G, Lee J, Shapiro J, Van Poppel H, Graefen M, Stief C (2008) Effect

of nightly versus on-demand vardenafil on recovery of erectile function in men following


EP

bilateral nerve-sparing radical prostatectomy. Eur Urol 54:924-931

6. McCullough AR, Hellstrom WG, Wang R, Lepor H, Wagner KR, Engel JD (2010)
C

Recovery of erectile function after nerve sparing radical prostatectomy and penile rehabilitation
AC

with nightly intraurethral alprostadil versus sildenafil citrate. J Urol 183:2451-2456

7. Mulhall JP, Slovick R, Hotaling J, Aviv N, Valenzuela R, Waters WB, Flanigan RC

(2002) Erectile dysfunction after radical prostatectomy: hemodynamic profiles and their

correlation with the recovery of erectile function. J Urol 167:1371-1375

15
ACCEPTED MANUSCRIPT

8. Vignozzi L, Filippi S, Morelli A, Ambrosini S, Luconi M, Vannelli GB, Donati S,

Crescioli C, Zhang XH, Mirone V, Forti G, Maggi M (2006) Effect of chronic tadalafil

administration on penile hypoxia induced by cavernous neurotomy in the rat. J Sex Med 3:419-

431

PT
9. Zelefsky MJ, Eid JF (1998) Elucidating the etiology of erectile dysfunction after

RI
definitive therapy for prostatic cancer. Int J Radiat Oncol Biol Phys 40:129-133

10. Ilic D, Hindson B, Duchesne G, Millar JL (2013) A randomised, double-blind, placebo-

SC
controlled trial of nightly sildenafil citrate to preserve erectile function after radiation treatment

for prostate cancer. J Med Imaging Radiat Oncol 57:81-88

11.
U
Mulhall J (2003) Deciphering erectile dysfunction drug trials. J Urol 170:353-358
AN
12. Desouza C, Parulkar A, Lumpkin D, Akers D, Fonseca VA (2002) Acute and prolonged

effects of sildenafil on brachial artery flow-mediated dilatation in type 2 diabetes. Diabetes Care
M

25:1336-1339
D

13. Rosano GM, Aversa A, Vitale C, Fabbri A, Fini M, Spera G (2005) Chronic treatment
TE

with tadalafil improves endothelial function in men with increased cardiovascular risk. Eur Urol

47:214-220; discussion 220-212


EP

14. Foresta C, De Toni L, Magagna S, Galan A, Garolla A (2010) Phosphodiesterase-5

inhibitor tadalafil acts on endothelial progenitor cells by CXCR4 signalling. Curr Drug Deliv
C

7:274-282
AC

15. Padma-Nathan H, McCullough AR, Levine LA, Lipshultz LI, Siegel R, Montorsi F,

Giuliano F, Brock G (2008) Randomized, double-blind, placebo-controlled study of

postoperative nightly sildenafil citrate for the prevention of erectile dysfunction after bilateral

nerve-sparing radical prostatectomy. Int J Impot Res 20:479-486

16
ACCEPTED MANUSCRIPT

16. Montorsi F, Aversa A, Moncada I, Perimenis P, Porst H, Barker C, Shane M, Sorsaburu S

(2011) A randomized, double-blind, placebo-controlled, parallel study to assess the efficacy and

safety of once-a-day tadalafil in men with erectile dysfunction who are naïve to PDE5 inhibitors.

J Sex Med 8:2617-2624

PT
17. Schiff JD, Bar-Chama N, Cesaretti J, Stock R (2006) Early use of a phosphodiesterase

RI
inhibitor after brachytherapy restores and preserves erectile function. BJU Int 98:1255-1258

U SC
AN
M
D
TE
C EP
AC

17
ACCEPTED MANUSCRIPT

Table 1. Demographic characteristics and baseline comorbidities data for patients who initiated

study therapy

Placebo Sildenafil Citrate

PT
N = 77 N = 125

RI
No. % No. %

SC
Demographics

Age (years)

≤65 50
U 65% 83 66%
AN
>65 27 35% 42 34%

Pretreatment PSA
M

<10 67 87% 113 90%


D

10-20 9 12% 9 7%
TE

>20 1 1% 1 1%

Gleason score
EP

6 44 57% 75 60%

7 29 38% 47 38%
C

>7 4 5% 3 2%
AC

RT type

EBRT 17 22% 25 20%

Brachytherapy 33 43% 57 46%

Combined EBRT + 27 35% 43 34%

18
ACCEPTED MANUSCRIPT

brachytherapy

Hormone therapy

No ADT 69 90% 112 90%

ADT 8 10% 13 10%

PT
Baseline comorbidities

RI
Diabetes 5 6% 4 3%

Hypertension 33 43% 44 35%

SC
Heart disease 9 12% 12 10%

Smoking history 32 42% 57 46%

U
ADT, androgen-deprivation therapy; EBRT, external beam radiotherapy; PSA prostate-
AN
specific antigen; and RT, radiotherapy.
M
D
TE
C EP
AC

19
ACCEPTED MANUSCRIPT

Table 2. Erectile Function (EF), International Index of Erective Function (IIEF) Total, and

Overall Satisfaction (OS) scores at baseline, 6, 12, and 24 months after therapy according to

treatment arm among patients treated with or without androgen-deprivation therapy

PT
Placebo Sildenafil Citrate

Time Points Median [Q1; Q3] Median [Q1; Q3] P Value

RI
Baseline EF 28.00 [25.00;29.50] 28.00 [25.00;30.00] 0.493

IIEF total 64.00 [56.75;69.00] 64.00 [57.00;71.00] 0.577

SC
OS 8.00 [7.00;10.00] 9.00 [7.25;10.00] 0.232

U
Month 6 EF 25.00 [11.00;28.00]
AN 27.00 [19.75;30.00] 0.036

IIEF total 58.00 [35.00;64.00] 59.00 [49.00;67.75] 0.091

OS 7.50 [4.25;8.00] 8.00 [7.00;9.00] 0.013


M

Month 12 EF 20.70 [13.25;27.75] 25.00 [18.50;29.00] 0.024

IIEF total 51.00 [34.50;63.50] 58.00 [41.50;66.75] 0.070


D

OS 6.00 [4.00;8.00] 8.00 [4.00;9.00] 0.069


TE

Month 24 EF 24.00 [8.75;29.00] 24.50 [14.00;29.00] 0.262

IIEF total 54.50 [29.75;64.75] 58.00 [39.00;65.00] 0.186


EP

OS 6.00 [4.00;8.00] 8.00 [5.00;9.00] 0.048


C

Q1, first quartile; Q3, third quartile. Baseline P values are from two-sided Wilcoxon rank sums
AC

tests. Post-baseline P values are from one-sided Wilcoxon rank sums tests with one-sided

alternative hypothesis that sildenafil citrate patients had higher scores than placebo patients.

20
ACCEPTED MANUSCRIPT

Table 3. Erectile Function (EF), International Index of Erective Function (IIEF) Total, and

Overall Satisfaction (OS) scores at baseline, 6, 12 and 24 months after therapy according to

treatment arm in the androgen-deprivation therapy patients

PT
Placebo Sildenafil Citrate

Time Points Median [Q1; Q3] Median [Q1; Q3] P Value

RI
Baseline EF 26.00 [20.50;27.50] 27.00 [25.50;29.00] 0.173

IIEF total 56.00 [49.00;63.00] 61.00 [44.00;67.00] 0.965

SC
OS 7.00 [5.00;9.00] 6.00 [4.00;10.00] 0.624

U
Month 6 EF 22.50 [8.75;25.75]
AN 18.00 [6.50;24.50] 0.628

IIEF total 56.00 [56.00;57.00] 43.00 [17.00;51.00] 0.959

OS 7.00 [6.00;9.00] 5.50 [4.50;6.50] 0.908


M

Month 12 EF 9.00 [2.50;15.75] 17.00 [1.00;23.00] 0.274

IIEF total 22.00 [9.75;36.75] 19.00 [5.50;44.50] 0.666


D

OS 4.00 [3.50;5.00] 3.00 [2.25;5.25] 0.707


TE

Month 24 EF 21.00 [10.50;26.50] 15.00 [10.80;24.00] 0.711

IIEF total 52.00 [32.50;61.75] 38.50 [19.85;59.50] 0.759


EP

OS 6.50 [4.25;8.00] 7.00 [2.50;8.00] 0.566


C

Q1, first quartile; Q3, third quartile. Baseline P values are from two-sided Wilcoxon rank sums
AC

tests. Post-baseline P values are from one-sided Wilcoxon rank sums tests with one-sided

alternative hypothesis that sildenafil citrate patients had higher scores than placebo patients.

21
ACCEPTED MANUSCRIPT

Table 4. Erectile Function (EF), IIEF total, and Overall Satisfaction (OS) scores at baseline, 6,

12, and 24 months after therapy according to treatment arm in the non-androgen-deprivation

therapy patients

Time Placebo Sildenafil Citrate

PT
points Median [Q1; Q3] Median [Q1; Q3] P Value

RI
Baseline EF 28.00 [25.00; 30.00] 28.00 [25.00; 30.00] 0.727

IIEF total 66.00 [58.00; 69.50] 65.00 [58.00; 71.00] 0.741

SC
OS 8.00 [7.25; 10.00] 9.00 [8.00; 10.00] 0.259

Month 6 EF 25.00 [11.50; 28.00] 27.00 [20.00; 30.00] 0.021

IIEF total
U
58.50 [34.50; 64.00] 60.50 [51.75; 69.00] 0.030
AN
OS 8.00 [4.00; 8.00] 8.00 [7.00; 9.50] 0.003

Month 12 EF 21.50 [14.00; 28.00] 26.00 [19.25; 29.00] 0.018


M

IIEF total 53.00 [37.70; 64.50] 60.00 [44.50; 67.50] 0.043


D

OS 7.00 [5.00; 8.00] 8.00 [5.25; 9.75] 0.027


TE

Month 24 EF 25.00 [9.00; 29.00] 25.00 [17.00; 29.00] 0.172

IIEF total 54.50 [33.00; 65.00] 58.50 [45.75; 66.00] 0.097


EP

OS 6.00 [4.50; 8.00] 8.00 [5.50; 9.50] 0.033

Q1, first quartile; Q3, third quartile. Baseline P values are from two-sided Wilcoxon rank sums
C

tests. Post-baseline P values are from one-sided Wilcoxon rank sums tests with one-sided
AC

alternative hypothesis that sildenafil citrate patients had higher scores than placebo patients.

22
ACCEPTED MANUSCRIPT

Table 5. Linear regressions fit using generalized estimating equations testing effect of sildenafil

citrate (SC) arm on all scores 12 months or longer after radiotherapy for Erectile Function (EF),

IIEF Total, and Overall Satisfaction (OS) among non-androgen-deprivation therapy patients,

controlling for baseline scores and age

PT
Outcome Predictor Regression Standard

RI
Coefficient Error P Value

EF Intercept 19.879 0.996 <0.001

SC
Baseline EF1 0.739 0.163 <0.001

Age1 -0.055 0.077 0.473

SC Arm2 1.63
U 1.241 0.189
AN
IIEF Total Intercept 48.963 2.34 <0.001

Baseline IIEF
M

0.816 0.133 <0.001


Total1

Age1 -0.091 0.173 0.599


D

SC Arm2 4.145 2.773 0.135


TE

OS Intercept 6.55 0.289 <0.001

Baseline OS1 0.583 0.109 <0.001


EP

Age1 -0.005 0.022 0.827

SC Arm2 0.667 0.359 0.063


C

1
Baseline scores and age were centered around their means.
AC

2
The regression coefficients for the predictor “SC Arm” represent the average differences

between the SC and placebo arms in the scale scores assessed at 12 months or later from the start

of radiotherapy, controlling for baseline score and age. Positive values indicate better scores in

the SC arm.

23
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
C EP
AC

Fig 1. International Index of Erectile Function (IIEF) median scores and inter-quartile ranges

according to the assigned arms during the 2-year study period among the non-androgen-

deprivation patients.

24
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
C EP
AC

Fig 2. Erectile function (EF) median scores and inter-quartile ranges according to the assigned

arms during the 2-year study period among the non-androgen-deprivation patients.

25
Prostate Cancer and Prostatic Disease (2014) 17, 180–186 OPEN
& 2014 Macmillan Publishers Limited All rights reserved 1365-7852/14
www.nature.com/pcan

ORIGINAL ARTICLE
A double-blind, placebo-controlled randomised trial evaluating the
effect of a polyphenol-rich whole food supplement on PSA
progression in men with prostate cancer—the UK NCRN Pomi-T
study
R Thomas1,2,3, M Williams1, H Sharma1, A Chaudry2 and P Bellamy3

BACKGROUND: Polyphenol-rich foods such as pomegranate, green tea, broccoli and turmeric have demonstrated anti-neoplastic
effects in laboratory models involving angiogenesis, apoptosis and proliferation. Although some have been investigated in small,
phase II studies, this combination has never been evaluated within an adequately powered randomised controlled trial.
METHODS: In total, 199 men, average age 74 years, with localised prostate cancer, 60% managed with primary active surveillance
(AS) or 40% with watchful waiting (WW) following previous interventions, were randomised (2:1) to receive an oral capsule
containing a blend of pomegranate, green tea, broccoli and turmeric, or an identical placebo for 6 months.
RESULTS: The median rise in PSA in the food supplement group (FSG) was 14.7% (95% confidence intervals (CIs) 3.4–36.7%), as
opposed to 78.5% in the placebo group (PG) (95% CI 48.1–115.5%), difference 63.8% (P ¼ 0.0008). In all, 8.2% of men in the FSG and
27.7% in the PG opted to leave surveillance at the end of the intervention (w2 P ¼ 0.014). There were no significant differences
within the predetermined subgroups of age, Gleason grade, treatment category or body mass index. There were no differences in
cholesterol, blood pressure, blood sugar, C-reactive protein or adverse events.
CONCLUSIONS: This study found a significant short-term, favourable effect on the percentage rise in PSA in men managed with AS and
WW following ingestion of this well-tolerated, specific blend of concentrated foods. Its influence on decision-making suggests that this
intervention is clinically meaningful, but further trials will evaluate longer term clinical effects, and other makers of disease progression.

Prostate Cancer and Prostatic Disease (2014) 17, 180–186; doi:10.1038/pcan.2014.6; published online 11 March 2014
Keywords: nutrition; active surveillance; watchful waiting

INTRODUCTION Australian study linked a similar supplement intake with more


Diets deficient in polyphenols and other natural plant-based subsequent skin cancers.5 Long-term folate supplementation after
phytochemicals found in herbs, spices, fruit, teas, colourful myocardial infarct resulted in a higher cancer risk.21 A supplement
vegetables and other healthy plant-based foods, have been linked containing vitamin C, copper and manganese did not slow PSA
with higher risks of cancer particularly breast,1,2 pancreas,3 ovary,4 progression;15 the selenium and vitamin E cancer prevention trial
skin,5 prostate,6,7 bowel8 and oesophagus.9 (SELECT) study showed an increased prostate cancer incidence
The protective benefits of polyphenols, however, do not to stop following long-term intake of vitamin E and selenium22 as did men
after a diagnosis of cancer. Breast cancer survivors taking higher in the Health Professionals Study, who took zinc.23 Despite some
levels of fruit and vegetables had a lower recurrence10 and those initial encouragement from cohort and small prospective studies,
with a higher dietary intake of lignans, isoflavones, flavanones lycopene, saw palmetto or genistein extracts evaluated within
within soy-rich foods or green tea had a lower risk of breast cancer more scientifically robust analyses did not demonstrate a benefit
death.11–13 Individuals with skin cancer who had higher leafy, for either prostate cancer, benign prostatic hypertrophy or other
green vegetable intake had a lower rate of new cancer formation.5 malignancies.1,6,18,24–26
Men adopting healthy diets after prostate cancer were shown to As a consequence of these data, scientific attention has been
have slower PSA progression.14,15 turning towards the evaluation of concentrated polyphenol-rich
The potential benefits of concentrating foods into a pill has whole food supplements, rather than extracted chemicals, as
been the subject of extentive evaluation. Up to now, research has convenient ways to boost poor diets or further enhance already
focused on supplements containing specific, extracted chemicals adequate diets. There are some laboratory and phase II studies to
believed to be the anti-cancer candidates and although some support their further evaluation and hence the rationale for this
studies have shown benefits,16–18 most have not or were actually study. Men with prostate cancer, managed with active surveillance
linked to an increased risk of cancer. For example, the two vitamin (AS) or watchful waiting (WW) for a PSA relapse after radical
A and E studies increased the risk of lung cancer19,20 and an treatments, were selected as an ideal cohort to evaluate a lifestyle

1
The Primrose Research Unit, Bedford Hospital, Bedford, UK; 2Department of Oncology, Addenbrooke’s Cambridge University NHS Trust, Cambridge, UK and 3Department of
Postgraduate Medicine, Cranfield University, Cranfield, UK. Correspondence: Professor R Thomas, Bedford and Addenbrooke’s Cambridge University NHS Hospitals, c/o The
Research Unit, Primrose Oncology, Bedford Hospital, Bedford MK42 9DJ, UK.
E-mail: robert.thomas@bedfordhospital.nhs.uk
Received 13 October 2013; revised 14 January 2014; accepted 26 January 2014; published online 11 March 2014
Polyphenol rich food supplementation
R Thomas et al
181
intervention as they have a useful serum marker of their  green tea 5:1 extract (Camellia sinensis) 20 mg equivalent to 100 mg of
disease, PSA, and medical interventions are often not indicated green tea and
initially.15  bulking agent (di-calcium phosphate), anti-caking agents (modified maize-
based starch, maltodextrin and magnesium stearate) removed post trial.
Rationale for the ingredients of this interventional food
supplement The control group took a placebo containing identical bulking and anti-
caking agents with 10 mg of watercress extract to provide an identical
Pomegranate, rich in ellagic acid, has been shown in in vitro colour and substance.
studies to inhibit proliferation, markers of migration, induce All participants were men with an average age of 74 years (range 53–89
apoptosis and cell adhesion in breast and prostate cancer cell years), with histologically confirmed prostate cancer, 121 (60%) were being
lines.27 In humans, a phase II study reported a prolongation of PSA managed with AS, and 78 (40%) managed with WW following previous
doubling following pomegranate juice consumption and markers radical interventions and radical local salvage therapies had been excluded
of oxidative stress improved.28 A further phase II study, gave men (primary radiotherapy, 65; surgery followed by radiotherapy, 8; and
brachytherapy, 9). The baseline characteristics are summarised in Table 1.
pomegranate seed extract with similar effects.29 Its influence was
Following written informed consent, men were randomised by
not felt to be via hormonal route as it affected both androgen- externally generated, numerically sequenced, opaque, tamper-proof
sensitive and -resistant human prostate cancer cells, and one of envelopes to the food supplement group (FSG) or placebo group (PG).
the clinical studies showed no change in testosterone levels There was a 2:1 randomisation which resulted in 136 in the FSG and 67 in
following regular intake.25,30–34 the PG. Four men withdrew consent after initial randomisation and
Green tea, rich in epigallocatechin gallate, has been shown to proceeded to intervention before the 3-month consultation (two from
block ornithine decarboxylase, an enzyme which signals cells to each group), had no further relevant PSA and as such could not be
proliferate faster and bypass apoptosis.35–37 It has also been included in an intention to treat analysis (Figure 1). At baseline, 3 months
reported to reduce several growth factors which promote breast and 6 months post intervention, PSA, full blood count, urea and
and prostate cancer cell line growth, block de-differentiation and electrolytes, liver function profile, blood glucose, fasting cholesterol,
C-reactive protein, body weight, height and blood pressure were
angiogenesis.38 Men given an extract of tea illustrated a significant measured. Adverse and favourable events were recorded in the Case
reduction in the levels of several growth factors that promote Report Form according to the National Cancer Institute (NCI) common
cancer, as well as a beneficial effect on PSA.35 toxicity grading scale.
Broccoli, rich in isothiocyanate and its metabolite sulphora-
phane, has been shown to inhibit growth and promote apoptosis Certification and quality assurance
in cancer cells.39 In humans, a study found that regular broccoli
This trial was approved by the National Ethics Committee, was peer
intake downregulated cancer genes linked to cancer promotion reviewed by the National Cancer Research Institute (NCRI) Complementary
and up-regulated genes link to cancer suppression.5,7,39,40 Therapies Research Committee and formally adopted by the National
Curcumin, which gives turmeric its yellow colour, has been Cancer Research Network (NCRN). The Medicines and Health Regulatory
shown to slow prostate cancer cell growth, increase apoptosis, Agency (MHRA) confirmed that no MHRA licence was required as the
reduce markers of invasion and migration of cells.41–44 It has been intervention was not classified as a medicinal product. The randomisation
shown to inhibit tyrosine kinase activity of the epidermal growth process was outsourced and the trial methodology, collection and storage
factor receptor,45 have cyco-oxidase-I-mediated anti-inflammatory of data were verified and independently audited by an external agency to
properties46 and halt the growth of stem cells that give rise to ensure adherence to European Good Clinical Practice. At the end of the
breast cancer without influencing normal breast cells.46 trial, data were externally audited for a second time to ensure that there
were no data inconsistencies or deviation from the trial design, before the
Finally, all four ingredients also have some anti-oxidant proper- database was sealed and sent for blinded analysis by the statistician at
ties that are thought to protect the DNA against oxidative damage Cranfield University. The UK manufacturers of the food supplement (Power
from ingested or environmental carcinogens,36,37,47 although this Health Products, York, UK) adhered to good manufacturing practice
precise mechanism has not been confirmed clinically.48 guidelines and performed in-house analysis for authenticity and purity (the
The rationale and hypothesis for selecting the ingredients of manufacturing analysis certificates were presented to the reviewers of the
this supplement were that as they originate different food sources publication). The food supplement and placebo tablets were supplied to
(fruit, herb, vegetable and leaf), each with their unique profile and the trials unit in tamper-proof, sealed containers. A batch of the
concentration of polyphenols, their separate anti-cancer mechan- supplement has been securely stored by the trust secretary and can be
isms, summarised above, could be synergistic49,50 yet at the same sent to any regulatory body at request.
time their variable composition would avoid over-consumption of
one particular phytochemical.25,30,32–34,41–44 The trial committee Table 1. Summary of baseline characteristic in the randomly assigned
determined the concentration of each ingredient based on the groups
amounts safely used within previous clinical studies.25,30,32–34,41–44
Baseline characteristic FSG (134) PG (65)

Age (mean years) 71.8 76.4a


MATERIALS AND METHODS PSA (mean mg l  1) 6.5 6.5
This was a placebo-controlled, double-blind, randomised trial designed Gleason gradep7 127 (95%) 57 (88%)
with the aim of establishing whether supplementing the diet with a Gleason grade 47 7 (5%) 8 (12%)
polyphenol-rich, whole food supplement containing a blend of green tea, Gleason grade mean (mg l  1) 6.5 6.2
pomegranate, broccoli and curcumin (turmeric) influenced the rate of PSA BMI (mean kg m  2) 28.1 28.3
progression, compared with placebo among men with prostate cancer Cholesterol (mean mmol l  1) 4.87 4.72
either managed with primary AS or WW, following a PSA relapse post- BP (mean systolic/diastolic mm Hg) 146/83 150/82
radical treatments. The 203 participants originated from across the UK and Serum glucose (mean mmol l  1) 5.15 5.30
were all consented and randomised at The Primrose Oncology Unit, C-reactive protein (mean mg l  1) 1.51 1.74
Bedford Hospital, from a total of 208 reviewed for eligibility, between
November 2011 and July 2012. The intervention consisted of a tablet taken Abbreviations: BMI, body mass index; BP, blood pressure; FSG, food
three times a day containing: supplement group; PG, placebo group.
The mean age in the PG was older by 4.4 years (t-test P ¼ 0.013) so age was
included in the analysis of the percentage change in PSA as a covariate.
 broccoli powder (Brassica oleracea) 100 mg, a
Randomisation produced no statistical difference in the group character-
 turmeric powder (Curcuma longa) 100 mg,
istics except for age.
 pomegranate whole fruit powder (Punica granatum) 100 mg,

& 2014 Macmillan Publishers Limited Prostate Cancer and Prostatic Disease (2014), 180 – 186
Polyphenol rich food supplementation
R Thomas et al
182

Figure 1. Consort diagram highlighting the flow of patients through the National Cancer Research Network Pomi-T study.

Statistical considerations Median percentage PSA change - all men


The patient representatives on the NCRI Development Committee felt that Difference 63.8% ANCOVA p=0.0008
Percentage rise plus 95% CI

a 2:1 randomisation would be more acceptable to participants. This 120


appeared to be a correct assertion because all but five eligible men invited
to enter this study agreed to be randomised. The PSA value used for the 100
final analysis was pre-determined to be the 6-month value or the 3-month
All men n=199
80
value in the men who withdraw at this stage.
60
40
Statistical methods
The percentage change in PSA from baseline to final measurement was 20
evaluated using an analysis of covariance (ANCOVA) which assessed the 0
effect of the FSG versus PG, as well as the predetermined subgroups of
Gleason grade, body mass index and treatment category. Age was -20
FS group Placebo group
included as a covariate to adjust for any differences in age between
groups. The interactions between the effect of the FS and each of the other Key: FS = Food supplement, ANCOVA = Analysis of covariance,
categorical effects were also included in the analysis. To satisfy the = 95% confidence intervals
assumptions of this analysis, the change in PSA had to be transformed to
logarithms, P-values given in this section followed by ANCOVA are from Figure 2. Median percentage rise in PSA between men taking the
the F-value in this ANCOVA. All median values were back transformed from food supplement versus placebo.
this ANCOVA and therefore allow for the effect of any differences in other
subgroups or age.
The analysis of the number of men at the end of the trial with the same PG. This difference was statistically significant (w2 value with 1
or lower PSA was analysed using a w2 test with 1 degree of freedom. The
degree of freedom ¼ 19.58, P ¼ 0.000010).
differences in toxicity measures between the two groups were tested using
an appropriate t-test (that is, with equal or unequal variances), and
transforming the measure if required to satisfy the assumptions of a t-test. Decisions to remain on AS or WW
Twenty-five men opted to leave AS or WW at 3 months (11 in the
FSG and 14 PG), and 4 after 6 months, 11 of 134 in the FSG (8.2%)
RESULTS and 18 of 65 (27.7%) in the PG (this difference of 19.5% was
Primary end point significant w2 value with 1 degree of freedom, P ¼ 0.014). The
In the FSG, the mean PSA rose from 6.50 to 6.81 ug l  6 from reasons for opting out were multifactorial and at the discretion of
baseline to the end of the intervention, a median PSA percentage the physician and patient who were both blind to the intervention
rise of 14.7% (95% confidence interval (CI)  3.4% to 36.7%). In arm but all had a rising PSA.
the PG, the mean PSA increased from 6.50 to 10.98 ug l  1, a
median percentage rise of 78.5% (95% CI 48.1–115.5%). The Predetermined subgroup analysis
median percentage PSA increased at a significantly slower rate in
A separate analysis of the cohort of men managed with AS
the FSG group compared with the PG (difference 63.8% ANCOVA,
(n ¼ 121) revealed that in the FSG the mean PSA dropped by
P ¼ 0.0008).
0.14% (95% CI  7.57 to 7.95), whereas in the PG it rose by 46.98%
(95% CI 28.51–68.31); difference 47.12% (ANVOCA, P ¼ 0.001), see
Secondary end point Figure 2. A separate analysis of the cohort of men managed with
The number of men with a PSA lower or the same value at trial WW (n ¼ 78) revealed that in the FSG the mean PSA rose by 8.78%
completion was 61 (46%) in the FSG as opposed to 9 (14%) in the (95% CI  6.32 to 26.62), whereas in the PG it rose by 80.34%

Prostate Cancer and Prostatic Disease (2014), 180 – 186 & 2014 Macmillan Publishers Limited
Polyphenol rich food supplementation
R Thomas et al
183
Median percentage change in PSA (AS) Other measures
Difference 47.12% ANCOVA p=0.001 There were no significant differences at the beginning or the end
100 of the study between groups for cholesterol, blood pressure,
serum glucose or C-reactive protein. Compliance, measured by
80
counting remaining tablets in returned pots, was excellent and
Men on active surveillance n=121
60 similar at 98.4% in the PG and 96.5% in the FSG. Sex hormones
were not a predetermined analysis, as the ingredients were
40 specifically chosen not to have phytoestrogenic properties but
were measured in 64 men who had been taking the supplement
20 for at least 3 months or more. Three of these men (5.5%) had
testosterone levels below our laboratory normal range. The
0
average testosterone (13.4 nmol l  1) was within the normal range
-20 as were the other sex hormones: follicle stimulating hormone,
FS group Placebo group 9.2 iu l  1; luteinizing hormone, 7.4 iu l  1; sex hormone binding
globulin, 41.4 nmol l  1; and free androgen index, 24.3%. Magnetic
Key: FS = Food supplement, ANCOVA = Analysis of covariance, resonance images (MRI) of the prostate were also not included at
= 95% confidence intervals specific time points in the trial protocol but a total of 74 of the 121
Figure 3. Subgroup analysis: median percentage change in PSA for men on AS, were taken as part of their routine management
the 121 men managed with active surveillance (AS). clinical protocol. All these scans, in addition to their original report,
were scrutinised within our multidisciplinary team meetings.
Twelve (16%) men had radiologically progressive disease and in
these men the average PSA rose from 7.65 to 8.67 ug l  1. Eight
(11%) had radiological regression and in these the average PSA
dropped from 7.2 to 4.1 ug l  1. No man in the FSG had
radiological progression with a stable PSA but one man (1.3%)
Median percentage change in PSA (WW) not taking the FS had radiological progression with a falling PSA.
Difference 71.56% ANCOVA p=0.001 Although these figures were not subjected to statistical analysis as
110 MRI was not a predetermined end point but they do give some
reassurance that PSA change is linked to underlying disease
90 status.
Men on watchful waiting n=78
70
Adverse events
50 There were 34 (24%) men who recorded adverse events (any NCI
score) in the FSG group and 23 (34%) in the PG. These differences
30
were not statistically significant (w2 value with 1 degree of
10 freedom ¼ 2.2, P ¼ 0.14). There were no grade X3 toxicities, but
one man in the FSG group had grade 2 diarrhoea. Gastrointestinal
-10 events, considered separately, occurred in 21 (15.5%) in the FSG
FS group Placebo group
group as opposed to 5 (7.5%) in the PG, but this difference
Key: FS = Food supplement, ANCOVA = Analysis of covariance, was not statistically significant (w2 value with 1 degree of
= 95% confidence intervals freedom ¼ 2.24, P ¼ 0.11). No man in either group reported central
nervous system symptoms, such as agitation, insomnia or tremors,
Figure 4. Subgroup analysis: median percentage change in PSA for none of the 30 men on warfarin reported any unexpected change
the 78 men managed with watchful watching (WW; PSA relapse in the international normalized ratio, nor did the 43 men taking
following previous radiotherapy). ramipril report an unexpected change in their blood pressure.

Positive events
(95% CI 50.54–116.55); difference 71.56% (ANVOCA, P ¼ 0.001), see There were 16 (12%) men who recorded positive events (mainly
Figure 3. This difference between the median percentage change improved bowel and urinary function) in the FSG, and 3 (4.6%) in
in PSA in either the AS or WW cohorts was not statistically the PG. These differences were not statistically significant (w2 value
significant (P ¼ 0.805 ANCOVA) (Figure 4). with 1 degree of freedom ¼ 2.67, P ¼ 0.10) (Table 2).
There was no statistical difference between the median
percentage PSA rise between men in the FSG or the PG as to
whether they were overweight (X25 kg m  2) or not (P ¼ 0.564 DISCUSSION
ANCOVA). Although men with higher Gleason grade for the entire This study demonstrated a significant effect on the rate of PSA
cohort tended to progress at a faster rate, there was no difference progression among men with prostate cancer, randomised to take
between the median percentage change in PSA between men in this nutritional supplement compared with placebo. The differ-
the FSG or the PG whichever Gleason grade category they were ence was large, the patient characteristics were well-balanced and
(P ¼ 0.089 ANCOVA). There was a significant effect of age on the the trial had sufficient numbers to ensure adequate statistical
median percentage change in PSA for the entire cohort with older power. However, there are some caveats with the trial design to
men tending to progress at a slower rate (P ¼ 0.0272 ANCOVA; consider and discuss.
slope of log(MPC-PSA) ¼ 0.0098, P ¼ 0.0272). As there were more The first caveat was the relatively short, 6 months, duration of
older men randomised to the PG, age was included as a covariate the intervention which although enough time to detect an early
in the analysis so that the difference between the median difference, as men are often managed with surveillance for many
percentage change in PSA for FSG and PG was adjusted to the years, a longer design would exclude the possibility that this effect
mean age of the whole cohort, so that any effect on PSA between was short lived. On the other hand, a longer study would have
FSG and PG would have been removed. been beyond the funding constraints of this non-commercial

& 2014 Macmillan Publishers Limited Prostate Cancer and Prostatic Disease (2014), 180 – 186
Polyphenol rich food supplementation
R Thomas et al
184
Another issue with the trial design was that the cohort included
Table 2. Summary of the adverse and positive events
men both on primary AS and those experiencing a PSA relapse
FSG PG Difference % after radical treatments, deemed not eligible for salvage local
(% of 134) (% of 65) (significance) therapies. The design committee believe that this was not a
negative caveat as there were appropriate reasons for this choice.
Adverse gastro-intestinal events First, in our experience, this is precisely the group of men who are
Loose bowels 6 (4.5%) 0 (0%) 4.5 (ns) most interested in lifestyle strategies15 and a trial cohort should,
Diarrhoea (grade 1) 2 (1.5%) 0 (0%) 1.5 (ns) where possible, reflect the wider population considering the study
Diarrhoea (grade 2) 2 (1.5%) 1 (1.5%) 0 (ns) intervention. Both groups of men are similar as they have viable
Diarrhoea (grade 0 (0%) 0 (0%) 0 (ns)
prostate cancer, and are not receiving other systemic therapies.
X3)
Constipation 2 (1.5%) 0 (0%) 1.5 (ns) Both groups have serum PSA being monitored as part of their
Flatulence 5 (3.6%) 0 (0%) 3.6 (ns) routine clinical management. Previous nutritional studies have
Rectal bleeding 0 (0%) 1 (1.5%) 1.5 (ns) also included both groups.15,17,26 To support this rationale, in
Nausea 0 (0%) 1 (1.5%) 1.5 (ns) this study, there was no statistical difference in the effect on
Bloating 4 (3%) 2 (3%) 0 (ns) PSA between both cohorts, suggesting a similar effect of
All GI adverse 21 (15.5%) 5 (7.5%) 8 (ns) polyphenol-rich foods, regardless of whether patients had
events received radiotherapy or not.
Despite these caveats, although PSA has its short falls, men
Other adverse events
managed with AS or WW are greatly concerned with their serum
Gout exacerbation 2 (1.5%) 1 (1.5%) 0 (ns)
Worsening urinary 2 (1.5%) 2 (3%) 1.5 (ns) levels and a rise is often a trigger for a change in their
flow management.31 This was borne out in the trial, as 29 men opted
Worsening renal 2 (1.5%) 0 (0%) 1.5 (ns) for to leave AS or WW during or at the end of the study when their
function PSA rose, twice the percentage in the PG than the FSG. This
Weight loss 0 (0%) 2 (3%) 3 (ns) difference, as well as achieving strong statistical significance,
Non-specific 2 (1.5%) 4 (6%) 4.5 (ns) suggests a clinically meaningful effect. Notwithstanding avoidance
‘feeling unwell’ of the inevitable toxicities of androgen deprivation therapy, with
Miscellaneous 5 (3.6%) 9 (13.4%) 9.8 (ns) over a 10-fold difference in price between this food supplement
unrelated
and androgen deprivation therapy, a future trial should include a
All adverse events 34 (24%) 23 (34%) 10 (ns)
cost-effectiveness analysis in order to determine the magnitude of
Positive events potential savings for healthcare providers.15
Improved erectile 1 (0.75%) 0 (0%) 0.75 (ns) As well as price, other attractive features of this supplement
function were its tolerance and safety. There were no overall statistically
Improve urinary 4 (3%) 1 (1.5%) 1.5 (ns) significant difference in symptoms compared with placebo, although
flow more men in the supplement group experienced non-significant
Reduced prostatic 1 (0.75%) 0 (0%) 0.75 (ns) bloating or diarrhoea, but almost as many reported beneficial
discomfort effects including improved digestion and urinary symptoms. The
All positive 6 (4.5%) 1 (1.5%) 3 (ns)
trend towards better urinary symptoms is worthy of future scrutiny
prostatic symptoms
Improved bowel 8 (6%) 0 (0%) 6 (ns) in the next study, especially as previous studies have linked
function turmeric with improved symptoms of prostatitis presumably via its
Improved well 2 (1.5%) 2 (3%) 0 (ns) anti-inflammatory properties.41,46 One of the ingredients of the
being supplement, pomegranate, has been cited to be a weak inhibitor
All positive event 16 (12%) 3 (4.5%) 7.5 (ns) of cytochrome P450 but, reassuringly, the quantities used in this
study resulted in no unexpected changes in blood pressure or
Abbreviation: FSG, food supplement group; GI, gastrointestinal; ns, non-
significant difference; PG, placebo group.
international normalized ratio levels, which could have related to
interference of the metabolism of ramipril or warfarin.
Although, these ingredients have previously demonstrated
study. Furthermore, a trial designed to take a potential placebo for fundamental effects on cancer progression in laboratory studies
longer would have been a less attractive option for men, so the including markers of angiogenesis, metastasis, adhesion and
rapid recruitment achieved in this study would have been less apoptotis,25,30,32–34,41–44 our next trial will explore these mech-
likely. anisms in more detail building upon the translational designs
The second caveat was that the main end point relied on PSA instigated by previous researchers.14 Furthermore, although these
measurement, without other formal indicators of disease progres- ingredients were specifically selected not to have phytoestrogenic
sion including MRI or prostate biopsies which are now becoming properties and the average sex hormone levels were reassuringly
routine in the management of men on AS. Although biopsy would normal in the subgroup who had them measured, the next trial
have enhanced scientific competence, not all men currently will also explore the physiological effects on participants in more
consent a repeat intervention and hence this inclusion would have detail, including serum testosterone and markers of oxidative
reduced the rate of recruitment, increased complexity. The cost of stress.2,34
including MRI before and after the trial period would also have In conclusion, this statistically valid double-blind randomised
been prohibitive for the level of sponsorship available but controlled trial has demonstrated a significant short-term effect on
fortunately, images were taken as part of their routine manage- PSA and is food for thought for men living with prostate cancer,
ment in over half of the men in the AS group. It is of some 50–70% of whom are reported to have taken ‘over-the-counter’
reassurance, that PSA reflected underlying disease status as the supplements.47 The favourable effect on PSA progression was
percentage change in PSA was 10-fold lower in the men with significant both in men on primary AS and those experiencing a
disease shrinking on MRI compared with disease progression and PSA relapse after radiotherapy. This low cost food supplement was
no man taking the food supplement had disease progression with well-tolerated and also influenced clinically relevant decisions, as
a stable PSA. All men choosing to continue to supplement after to whether to switch to interventions with more toxicity. Although
the trial period will now receive annual MRI’s this data will subject these results do not prove a long-term effect, they have provided
to a future publication. significant encouragement to design a major study with more

Prostate Cancer and Prostatic Disease (2014), 180 – 186 & 2014 Macmillan Publishers Limited
Polyphenol rich food supplementation
R Thomas et al
185
comprehensive physiological, radiological and translational 19 Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A et al. Risk
laboratory end points, in order to get a deeper understanding factors for lung cancer and for intervention effects in CARET, the beta-carotene in
of the evidence of benefit and role for these complex and readily retinol efficacy trial. J Natl Cancer Inst 1996; 88: 1550–1559.
available foods. 20 Heinonen O, Albanes D, Virtamo J, Taylor PR, Huttunen JK, Hartman AM et al.
Prostate cancer and supplementation with alpha-tocopherol and beta-
carotene: incidence and mortality in a controlled trial. J Natl Cancer Inst 1998; 90:
440–446.
CONFLICT OF INTEREST 21 Figueiredo JC, Grau MV, Haile RW, Sandler RS, Summers RS, Bresalier RS et al. Folic
The authors declare no conflict of interest. acid and risk of prostate cancer: results from a randomized clinical trial. J Natl
Cancer Inst 2009; 101: 432–435.
22 Klein EA, Thompson IM, Tangen CM, Crowley JJ, Lucia MS, Goodman PJ et al.
ACKNOWLEDGEMENTS Vitamin E and the risk of prostate cancer: the selenium and vitamin E cancer
prevention Trial (SELECT). JAMA 2011; 306: 1549–1556.
The authors thank the charities Prostate Action and The Primrose Oncology Fund for
23 Leitzmann MF, Stampfer MJ, Wu K, Colditz GA, Willet WC, Giovannucci EL. Zinc
sponsoring this study and the enthusiasm and co-operation of the participants. The
supplementation and the risks of prostate cancer. J Natl Cancer Inst 2003; 95:
full trial protocol is available on cancernet.co.uk/pomi-t.htm. Peer reviewed
1004–1007.
sponsorship was received from the charity ‘Prostate Action’, and the study was also
24 Bent S, Kane C, Shinohara K, Neuhaus J, Hudes ES, Goldberg H et al. Saw Palmetto
supported by ‘The Primrose Oncology Fund’. No sponsorship was received from the
for benign prostatic hyperplasia. N Engl J Med 2006; 354: 557–566.
UK manufacturer of the investigational supplement used in this study.
25 Barber NJ, Zhang X, Zhu G, Pramanik R, Barber JA, Martin FL et al.
Lycopene inhibits DNA synthesis in primary prostate epithelial cells in vitro
and its administration is associated with a reduced prostate-specific
REFERENCES antigen velocity in a phase II clinical study. Prostate Cancer Prostatic Dis 2006; 9:
1 Hu F, Wang YB, Liang J, Yi B, Zhang W, Liang J et al. Carotenoids and breast cancer 407–413.
risk: a meta-analysis and meta-regression. Breast Cancer Res Treat 2012; 131: 26 Clark PE, Hall MC, Borden LS, Clark PE, Miller AA, Hu JJ et al. Phase I-II prospective
239–253. dose-escalating trial of lycopene in patients with biochemical relapse of prostate
2 Wu AH, Yu MC. Tea, hormone-related cancers and endogenous hormone levels. cancer after definitive local therapy. Urology 2006; 67: 1257–1261.
Mol Nutr Food Res 2006; 50: 160–169. 27 Wang L, Alcon A, Yuan H, Ho J, Li QJ, Martins-Green M. Cellular and molecular
3 Banim PJ, Luben R, McTaggart A, Welch A, Wareham N, Khaw KT et al. mechanisms of pomegranate juice-induced anti-metastatic effect on prostate
Dietary antioxidants and the aetiology of pancreatic cancer. Gut 2013; 62: cancer cells. Integr Biol (Camb) 2011; 3: 742–754.
1489–1496. 28 Pantuck AJ, Leppert JT, Zomorodian N, Seeram N, Seiler D, Liker H et al. Phase II
4 Tung K, Wilkens LR, Wu AH, McDuffie K, Hankin JH, Nomura AM et al. Association study of pomegranate juice for men with rising PSA following surgery or radiation
of dietary vitamin A, carotenoids and other antioxidants with the risk of ovarian for prostate cancer. J Urol 2005; 173: 225–226.
cancer. Cancer Epidemiol Biomarkers Prev 2005; 14: 669–676. 29 Paller CJ, Ye X, Wozniak PJ, Gillespie BK, Sieber PR, Greengold HR et al.
5 Heinen MM, Hughes MC, Ibiebele TI, Marks GC. Intake of antioxidant nutrients and A randomised phase II study of pomegranate extract for men with rising PSA
the risk of skin cancer. EJC 2007; 43: 2707–2716. following initial therapy for localized prostate cancer. Prostate Cancer Prostatic Dis
6 Giovannucci E, Rimm EB, Liu Y, Meir J, Stampfer MJ, Walter C et al. A prospective 2013; 16: 50–55.
study of tomato products, lycopene, and prostate cancer risk. J Natl Cancer Inst 30 Rettig MB, Heber D, An J, Seeram NP, Rao JY, Liu H et al. Pomegranate extract
2002; 94: 391–398. inhibits androgen-independent prostate cancer growth through a nuclear factor-
7 Joseph MA, Moysich KB, Freudenheim JL, Shields PG, Bowman ED, Zhang Y et al. kappaB-dependent mechanism. Mol Cancer Ther 2008; 7: 2662–2671.
Cruciferous vegetables, genetic polymorphisms and prostate cancer risk. Nutr 31 Lansky EP, Harrison G, Froom P, Jiang WG. Pomegranate (Punica granatum) pure
Cancer 2004; 50: 206–213. chemicals show possible synergistic inhibition of human PC-3 prostate cancer cell
8 Steward A, Gescher AJ. Curcumin in cancer management: recent results of invasion across Matrigel. Invest New Drugs 2005; 23: 121–122.
analogue design and clinical studies and desirable future research. Mol Nutr Food 32 Malik A, Mukhtar H. Prostate cancer prevention through pomegranate fruit. Cell
Res 2008; 52: 1005–2009. Cycle 2006; 5: 371–373.
9 Sun CL, Yuan JM, Koh WP, Lee HP, Yu MC. Green tea and black tea consumption 33 Khan N, Hadi N, Afaq F, Syed DN, Kweon MH, Mukhtar H. Pomegranate fruit
and colorectal cancer risk: The Singapore Chinese Health Study. Carcinogenesis extract inhibits prosurvival pathways in human A549 lung carcinoma cells and
2007; 28: 2143–2148. tumor growth in athymic nude mice. Carcinogenesis 2007; 28: 163–173.
10 Pierce JP, Natarajan L, Caan BJ, Parker BA, Greenberg ER, Flatt SW et al. Influence 34 Choi C, Kim JK, Choi SH, Jung HS, Kim HJ, Cho SY et al. Identification of steroid
of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis hormones in pomegranate (Punica granatum) using HPLC and GC–mass spec-
following treatment for breast cancer: The Women’s Healthy Eating and Living trometry. Food Chemistry 2006; 96: 562–571.
(WHEL) randomized trial. JAMA 2007; 298: 289–298. 35 Liao S, Kao YH, Hiipalla RA. Green tea: biochemical and biological basis for health
11 Buck K, Vrieling A, Zaineddin AK, Becker S, Hüsing A, Kaaks R et al. Serum benefits. Vitam Horm 2001; 62: 1–94.
enterolactone and prognosis of postmenopausal breast cancer. J Clin Oncol 2011; 36 Porrini M, Riso P. Factors influencing the bioavailability of antioxidants in foods: a
29: 3730–3738. critical appraisal. Nutr Metab Cardiovas 2008; 80: 353–361.
12 Boyapati SM, Shu XO, Ruan ZX, Dai Q, Cai Q, Gao YT et al. Soy food intake and 37 Liao J, Yang GY, Park ES, Meng X, Sun Y, Jia D et al. Inhibition of lung carcino-
breast cancer survival: a follow up of the Shanghai Breast Cancer Study. Breast genesis and effects on angiogenesis and apoptosis in mice given green tea. Nutr
Cancer Res Treat 2005; 92: 11–17. Cancer 2004; 48: 44–53.
13 Ogunleye AA, Xue F, Michels KB. Green tea and breast cancer risk of recurrence: a 38 Yang CS, Maliakal P, Meng X. Inhibition of carcinogenesis by tea. Annu Rev
meta-analysis. Breast Cancer Res Treat 2010; 119: 477. Pharmacol Toxicol 2002; 42: 25–54.
14 Ornish D, Magbanua MJM, Weidner G, Weinberg V, Kemp C, Green C et al. 39 Traka M, Gasper AV, Melchini A, Bacon JR, Needs PW, Frost V et al. The dietary
Intensive lifestyle changes may affect the progression of prostate cancer. J Urol isothiocyanate sulforaphane modulates gene expression and alternative gene
2005; 174: 1065–1070. splicing in a PTEN null preclinical murine model of prostate cancer. Mol Cancer
15 Thomas R, Oakes R, Gordon J, Russell S, Blades M, Williams M. A randomised, 2010; 9: doi:10.1186/1476-4598-9-189.
double-blind phase II study of lifestyle counselling and salicylate compounds in 40 Gasper AV, Al-Janobi A, Smith JA, Bacon RJ, Fortun P, Atherton C et al.
patients with progressive prostate cancer. Nutr Food Sci 2009; 39: 295–305. Glutathione S-transferase M1 polymorphism and metabolism of sulforaphane
16 Meyer F, Galan P, Douville P, Bairati I, Kegle P, Bertrais S et al. Antioxidant vitamin from standard and high-glucosinolate broccoli. Am J Clin Nutr 2005; 82:
and mineral supplementation and prostate cancer prevention in the SU.VI.MAX 1283–1291.
trial. Int J Cancer 2005; 116: 182–186. 41 Shah BH, Nawaz Z, Pertani SA, Roomi A, Mahmood H, Saeed SA et al. Inhibitory
17 Schröder FH, Roobol MJ, Boevé ER, de Mutsert R, Zuijdgeest-van Leeuwen SD, effect of curcumin, a food spice from turmeric, on platelet-activating factor-
Kersten I et al. Randomized, double-blind, placebo-controlled crossover study in and arachidonic acidmediated platelet aggregation through inhibition of
men with prostate cancer and rising PSA: effectiveness of a dietary supplement. thromboxane formation and Ca2 þ signaling. Biochem Pharmacol 1999; 58:
Eur Urol 2005; 48: 922–930. 1167–1172.
18 Brasky TM, Kristal AR, Navarro SL, Lampe JW, Peters U, Patterson RE et al. Specialty 42 Zhang HN, Yu CX, Chen WW, Jiang AL, Kong F, Deng DJ et al. Curcumin down
supplements and prostate cancer risk in the VITamins And Lifestyle (VITAL) regulates gene NKX3.1 in prostate cancer cell lines (LNcaP). Acta Pharmacol Sin
cohort. Nutr Cancer 2011; 63: 573–582. 2007; 28: 423–430.

& 2014 Macmillan Publishers Limited Prostate Cancer and Prostatic Disease (2014), 180 – 186
Polyphenol rich food supplementation
R Thomas et al
186
43 Iqbal M, Sharma SD, Okazaki Y, Fujisawa M, Okada S. Dietary supplementation of 48 Freedland SJ, Carducci M, Kroeger N, Partin A, Rao JY, Jin Y et al. A double blind
curcurmin enhances antioxidant phase II metabolosing enzymes in mice. Phar- randomised neoadjuvant study of the tissue effects of Pomx pills in men with
macol Toxicol 2003; 92: 33–38. prostate cancer before radical prostatectomy. Cancer Prev Res (Phila) 2013; 6:
44 Somasundaram S, Edmund NA, Moore DT, Small GW, Shhi YY, Orlowski RZ. 1120–1127.
Curcumin inhibits chemotherapy-induced apoptosis in models of cancer. Cancer 49 Parada J, Aguilera JM. Food combinations and microstructure affects the bioa-
Res 2002; 62: 3868–3875. vailability of several nutrients. J Food Sci 2007; 72: 21–23.
45 Dorai T, Gehani N, Buttyan R, Katz AE. Therapeutic potential of curcumin in human 50 Kakarla M, Brenner DE, Khorkya H, Cheng C, Tazi K, Ginestier C et al. Targeting
prostate cancer. Curcumin inhibits tyrosine kinase activity of the Epidermal breast stem cells with the preventive compounds curcumin and piperine. Breast
growth factor receptor. Mol Urol 2000; 4: 1–6. Cancer Res Treat 2010; 122: 777–785.
46 Handler N, Jaeger W, Puschacher H, Leisser K, Erker T. Synthesis of noval curcumin
analogues and their evaluation as selective cyclooxygenase-1 inhibitors. Chem
Pharm Bull 2007; 55: 64–71.
47 Bauer CM, Johnson EK, Beebe-Dimmer JL, Cooney KA. Prevalence and correlates This work is licensed under a Creative Commons Attribution 3.0
of vitamin and supplement usage amongst men with prostate cancer. Integr Unported License. To view a copy of this license, visit http://
Cancer Ther 2012; 11: 83–89. creativecommons.org/licenses/by/3.0/

Prostate Cancer and Prostatic Disease (2014), 180 – 186 & 2014 Macmillan Publishers Limited
PSA Testing for Prostate Cancer Screening Invited Commentary

Invited Commentary

Prostate-Specific Antigen Testing for Prostate Cancer Screening—


Is the Message Getting Through?
Freddie C. Hamdy, MD

Since the discovery of prostate-specific antigen (PSA) more than benefit in favor of early radical treatment, although ProtecT
40 years ago and its subsequent wide adoption for the detec- showed that early intervention reduced the rate of disease pro-
tion of prostate cancer, screening for the disease has been gression and metastasis in favor of radical interventions com-
plagued with continuing controversies that remain largely pared with active monitoring4 and recent reports from PIVOT
unresolved. There is a pro- suggest a benefit of early treatment for intermediate-risk
Related article
foundly engrained public per- disease.5 The most mature treatment effectiveness random-
ception embraced by many ized clinical trial is the Scandinavian Prostate Cancer Group-4
professionals that cancer detection as early as possible saves (SPCG-4) trial, which showed consistent benefits of radical
lives and therefore should be promoted at any cost, be it finan- treatments compared with watchful waiting in disease-
cial or in kind, such as the inevitable adverse effects caused by specific, overall survival and progression, but approximately
radical treatments. Although this may be true for some can- half of the SPCG-4 cohort already had nonorgan-confined dis-
cers, more than 3 decades of PSA testing among asymptomatic ease, and most were not detected via PSA test, which largely
men across the globe, particularly in the Western world, has explains the benefit patients received from early treatment.6
proved otherwise. Leapman et al1 illustrate this point in their Based on emerging evidence from these few global ran-
article on the changes in the rates of PSA testing associated domized clinical trials, the USPSTF made recommendations
with recent updates in the US Preventive Services Task Force in 2012 that PSA testing should not be offered to any man at
(USPSTF) recommendations on prostate cancer screening. any age with the purpose of prostate cancer screening (grade
Currently, the biggest challenges to making recommen- D recommendation). This recommendation has been associ-
dations on PSA testing as a public health screening policy for ated with a notable reduction in indiscriminate PSA testing in
prostate cancer are 3-fold: (1) the avoidance of overdetection the US but was deplored by many clinicians and institutions
of disease that would not otherwise manifest itself as clini- as in turn being associated with an increase in the number of
cally important during an individual’s lifetime, (2) the subse- men presenting with metastatic and advanced disease. As
quent inevitable overtreatment with potential unwanted ad- the trials matured, these recommendations were amended in
verse effects on quality of life, and (3) our inability to determine 2018, suggesting that men between 55 and 69 years of age can
accurately the lethal potential of prostate cancer at diagno- be offered a PSA test but that men aged 70 years or older should
sis, resulting in undertreatment. It has been clearly demon- not be tested.
strated that screening by PSA testing saves lives, but at the un- Leapman et al1 report the variations in PSA testing in a US
acceptable cost of overdetection and overtreatment, which has cohort of men aged 40 to 89 years during 2 distinct periods
prevented many policymakers and organizations from recom- associated with changes in the USPSTF recommendations in
mending PSA testing as a public health policy for asymptom- 2012 and 2018. The analysis was triggered because of the per-
atic men in the general community. ceived increase in the presentation of late-stage disease,
Guidelines usually rely on evidence to make recommen- claimed by some as being caused by the reduction in PSA test-
dations about any health intervention, but for prostate can- ing. The findings of the analysis are that there was a progres-
cer screening, evidence has been scarce over decades. Two sive and significant increase in PSA testing among men of all
large randomized clinical trials of screening by PSA testing in ages after the revision of the USPSTF grade C recommenda-
the US and Europe have been major sources of high-level data tion in 2018 for men aged 55 to 59 years, with particular con-
on interval screening by PSA, complemented by the more re- cerns about the increase in testing for men aged 40 to 55 years
cent Cluster Randomized Trial of PSA Testing for prostate can- and 70 years or older falling outside the age category in
cer (CAP)2 in the UK investigating the benefits of screening with the USPSTF recommendations and causing potential harm.
a single PSA test. The Prostate, Lung, Colon and Ovary (PLCO) The findings are valuable, and the authors should be com-
study in the US has ruled itself out as providing evidence be- mended for undertaking this informative analysis, which re-
cause of heavy contamination and other unresolvable issues. flects a flawed attitude toward PSA testing in the community.
The European Randomized Trial of Screening for Prostate Clinicians often advocate that the answer to the overde-
Cancer (ERSPC) in Europe showed a benefit in favor of screen- tection of prostate cancer is the adoption of safe active sur-
ing but at a high cost of overdetection,3 and the UK CAP study veillance protocols. This is the incorrect approach. Whenever
showed no survival benefit with a single PSA test but in- an asymptomatic man receives a diagnosis of prostate can-
creased detection of low-risk, low-volume prostate cancer.2 At cer, irrespective of its indolent nature, he is given a “new” iden-
the same time, trials of the effectiveness in screening for dis- tity of becoming a patient with cancer. A “cancer passport” is
ease, such as the ProtecT (Prostate Testing for Cancer and Treat- delivered, and its consequences may be far reaching. Men
ment) study in the UK4 and the Prostate Cancer Intervention should therefore be counseled properly regarding the ben-
vs Observation Trial (PIVOT)5 in the US, showed no survival efits and harms of screening before a decision is taken to mea-

jamaoncology.com (Reprinted) JAMA Oncology Published online November 11, 2021 E1

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Mexico | Access Provided by JAMA User on 11/21/2021


Invited Commentary PSA Testing for Prostate Cancer Screening

sure their PSA level. Quality of life in the SPCG-4 randomized tries but with slower uptake in the US. It is now evident that
clinical trial of watchful waiting vs radical prostatectomy the long-term practice of a PSA test followed by systematic
among a cohort of patients with prostate cancer was ana- biopsies of the prostate is antiquated. In the CAP trial, for in-
lyzed and compared with a matched cohort of men without stance, a single PSA test missed more than one-third of lethal
cancer. It was revealing that the harm caused to quality of life prostate cancers in men who underwent screening. 2 Al-
was likely due to the diagnosis of cancer rather than the treat- though genomic testing and the use of other emerging risk
ment administered.7 Similar findings of anxiety were re- calculators, such as polygenic hazard scores, have limited evi-
ported by the ERSPC, affecting men’s mental health after PSA dence to be adopted in a risk-stratification approach to screen-
testing. In the ProtecT study, patient-reported outcomes ing, it is likely that with imaging and targeted biopsies, the field
showed no evidence of a difference in anxiety and depres- will progress further, minimizing the risk of overdetection
sion levels between men who received active monitoring and and overtreatment and focusing on identifying early disease
men who underwent radical treatments.4 that warrants tailored treatments to improve outcomes and
The prostate cancer screening landscape continues to save lives. But when will the message get through to the pub-
evolve with the emergence of prebiopsy imaging using mul- lic, clinicians, and health care professionals that inappropri-
tiparametric magnetic resonance imaging of the prostate, ate PSA testing outside evidence-based recommendations
which has transformed the diagnostic pathway in many coun- should cease?

ARTICLE INFORMATION Trial (Cancer Research UK C11043/A4286, C18281/ surgery, or radiotherapy for localized prostate
Author Affiliation: Nuffield Department A8145, C18281/A11326, C18281/A15064). cancer. N Engl J Med. 2016;375(15):1415-1424.
of Surgical Sciences, University of Oxford, doi:10.1056/NEJMoa1606220
Oxford, United Kingdom. REFERENCES 5. Wilt TJ, Vo TN, Langsetmo L, et al. Radical
Corresponding Author: Freddie C. Hamdy, MD, 1. Leapman MS, Wang R, Park H, et al. Changes in prostatectomy or observation for clinically localized
Nuffield Department of Surgical Sciences, prostate-specific antigen testing relative to the prostate cancer: extended follow-up of the Prostate
University of Oxford, Old Road Campus revised US Preventive Services Task Force Cancer Intervention Versus Observation Trial
Research Bldg, Headington, Oxford OX3 7DQ, recommendation on prostate cancer screening. (PIVOT). Eur Urol. 2020;77(6):713-724. doi:10.1016/
United Kingdom (freddie.hamdy@nds.ox.ac.uk). JAMA Oncol. Published online November 11, 2021. j.eururo.2020.02.009
doi:10.1001/jamaoncol.2021.5143 6. Bill-Axelson A, Holmberg L, Garmo H, et al.
Published Online: November 11, 2021.
doi:10.1001/jamaoncol.2021.5129 2. Martin RM, Donovan JL, Turner EL, et al; Radical prostatectomy or watchful waiting in
CAP Trial Group. Effect of a low-intensity PSA-based prostate cancer—29-year follow-up. N Engl J Med.
Conflict of Interest Disclosures: Dr Hamdy screening intervention on prostate cancer 2018;379(24):2319-2329. doi:10.1056/
reported receiving grants from the National mortality: the CAP randomized clinical trial. JAMA. NEJMoa1807801
Institute for Health Research HTA Programme 2018;319(9):883-895. doi:10.1001/jama.2018.0154
and Cancer Research UK during the conduct of 7. Johansson E, Steineck G, Holmberg L, et al;
the study; and personal fees from Intuitive 3. Hugosson J, Roobol MJ, Månsson M, et al; SPCG-4 Investigators. Long-term quality-of-life
Surgical outside the submitted work; serving as ERSPC investigators. A 16-yr follow-up of the outcomes after radical prostatectomy or watchful
editor-in-chief, British Journal of Urology European Randomized Study of Screening for waiting: the Scandinavian Prostate Cancer Group-4
International; and being chief investigator of the Prostate Cancer. Eur Urol. 2019;76(1):43-51. randomised trial. Lancet Oncol. 2011;12(9):891-899.
National Institute for Health Research HTA ProtecT doi:10.1016/j.eururo.2019.02.009 doi:10.1016/S1470-2045(11)70162-0
trial (96/20/99) and coinvestigator of the CAP 4. Hamdy FC, Donovan JL, Lane JA, et al; ProtecT
Study Group. 10-Year outcomes after monitoring,

E2 JAMA Oncology Published online November 11, 2021 (Reprinted) jamaoncology.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Mexico | Access Provided by JAMA User on 11/21/2021


Gynecologic Oncology 117 (2010) 270–275

Contents lists available at ScienceDirect

Gynecologic Oncology
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / y g y n o

Impact of hydronephrosis on outcome of stage IIIB cervical cancer patients with


disease limited to the pelvis, treated with radiation and concurrent chemotherapy:
A Gynecologic Oncology Group study
Peter G. Rose a,⁎, Shamshad Ali b, Charles W. Whitney c, Rachelle Lanciano d, Frederick B. Stehman e
a
Cleveland Clinic Foundation and Case Western Reserve University, Cleveland, OH, USA
b
Gynecologic Oncology Group Statistical and Data Center, Roswell Park Cancer Institute, Buffalo, NY, USA
c
Christiana Gynecologic Oncology, LLC, Apex Medical Center, Newark, DE, USA
d
Delaware County Memorial Hospital, Drexel Hill, PA, USA
e
Indiana University School of Medicine, Indianapolis, IN, USA

a r t i c l e i n f o a b s t r a c t

Article history: Objectives. To estimate the significance of hydronephrosis and impact of ureteral obstruction relief on
Received 7 December 2009 outcome in patients with stage IIIB cervical cancer treated with radiation and concurrent chemotherapy.
Available online 24 February 2010 Methods. We retrospectively studied stage IIIB cervical cancer patients treated on GOG trials 56, 85, 120
and 165 evaluating radiation and concurrent chemotherapy. Eligible patient records were reviewed to assess
Keywords: the presence of hydronephrosis and treatment of ureteral obstruction. Patients were classified into three
Stage IIIB cervical cancer
groups; no hydronephrosis, hydronephrosis relieved from ureteral obstruction via stent or percutaneous
Hydronephrosis
Performance status
nephrostomy and hydronephrosis without treatment of ureteral obstruction.
Survival Results. 539 stage IIIB patients were studied. Hydronephrosis was present in 238 (44.2%). Patient age, race,
and tumor characteristics (size, histology and grade) were not significantly different between patients with or
without hydronephrosis. Patients with hydronephrosis received similar doses of radiation and cisplatin-based
chemotherapy. Both overall and progression-free survival were worse with hydronephrosis (log-rank test
p value = 0.0189 and 0.0186, respectively). Univariable analysis identified five prognostic factors; pelvic nodal
metastasis (p = 0.0001), tumor diameter (p = 0.0007), cisplatin-based concurrent chemoradiation
(p = 0.0031), hydronephrosis (p = 0.0189), and performance status (p = 0.0359). Hydronephrosis was
associated with worse performance status (p b 0.001). On multivariable analysis hydronephrosis was not a
significant prognostic factor. Ureteral obstruction relief occurred for 88% of patients and was associated with
improved survival.
Conclusion. In patients with stage IIIB cervical cancer restricted to the pelvis, hydronephrosis at
presentation is a significant but not independent prognostic factor associated with poor performance status
and poorer survival. Relief of ureteral obstruction is correlated with improved outcome.
© 2010 Published by Elsevier Inc.

Introduction without concurrent cisplatin-based chemotherapy. Stage IIIB is


defined as local extension of the cervical tumor to the pelvic sidewall
The prognosis for patients with advanced stage cervical cancer is or the presence of hydronephrosis. Hydronephrosis, particularly if
based on both the extent of local pelvic disease and the presence of bilateral, may result in varying degrees of renal insufficiency which
metastases. The Federation International Obstetrics and Gynecology may limit the ability to deliver concurrent cisplatin-based chemo-
(FIGO) staging system for cervical cancer describes various stages and therapy which has been recommended in a National Cancer Institute
substages of disease which, with the exception of the most advanced Clinical Alert [1]. Renal function may be improved or restored if the
stage (stage IVB), describe the extent of local/regional pelvic disease. renal obstruction is relieved by ureteral stents or percutaneous
Despite the use of relatively standard radiation protocols, survival nephrostomies. The impact of hydronephrosis and interventions to
worsens with increasing disease stage for patients treated with or correct it has not been extensively reported in advanced stage cervical
cancer patients treated with curative intent with radiation and
concurrent chemotherapy [2–6].
⁎ Corresponding author. Division of Gynecologic Oncology, Department of Obstetrics
and Gynecology, Cleveland Clinic Foundation, A81, 9500 Euclid Avenue, Cleveland,
The disease extent in patients with stage IIIB disease can vary
OH 44195, USA. Fax: +1 216 444 8551. depending on whether there is unilateral or bilateral parametrial
E-mail address: rosep@ccf.org (P.G. Rose). disease, whether there is unilateral or bilateral sidewall involvement,

0090-8258/$ – see front matter © 2010 Published by Elsevier Inc.


doi:10.1016/j.ygyno.2010.01.045
P.G. Rose et al. / Gynecologic Oncology 117 (2010) 270–275 271

whether hydronephrosis is present and if present is it unilateral or Table 1


bilateral, and overall tumor size. In addition to these local factors Patient factors by hydronephrosis.

patients with stage IIIB are at increased risk for extrapelvic metastasis. Characteristic Hydronephrosis statusa
Although basic radiologic imaging with plain radiographs is allowed by
No Hyn Hyn WR Hyn WOR
the FIGO staging system, more advanced techniques including
n (%) n (%) n (%)
lymphangiography, computerized tomography (CT), magnetic reso-
nance imaging (MRI) and more recently positive emission tomogra- Age group ≤25 2 (0.7) 2 (1.0) 0 (0.0)
26–35 31 (10.3) 25 (12.0) 3 (10.3)
phy (PET) which have been utilized to evaluate patients for extrapelvic
36–45 80 (26.6) 44 (21.1) 9 (31)
nodal or distant metastases are not accepted [7]. Furthermore, the 46–55 83 (27.6) 75 (35.9) 11 (37.9)
prognosis of patients with stage IIIB disease depends on how metas- 56–65 62 (20.6) 36 (17.2) 6 (20.7)
tases to the paraaortic nodes have been excluded, i.e. radiologically or ≥66 43 (14.3) 27 (12.9) 0 (0.0)
by surgical lymphadenectomy [8]. Surgical findings are not utilized in Race Black 60 (19.9) 37 (17.7) 8 (27.6)
White 154 (51.2) 111 (53.1) 14 (48.3)
the FIGO staging system and therefore disease identified by retroper-
Other 9 (3.0) 8 (3.8) 0 (0.0)
itoneal lymphadenectomy does not change the patient's disease stage. Unknown 78 (25.9) 53 (25.4) 7 (24.1)
The purpose of the current study was to estimate the significance GOG performance 0 179 (59.5) 115 (55.0) 19 (65.5)
of ureteral obstruction as a prognostic variable in stage IIIB cervical 1 107 (35.5) 78 (37.3) 9 (31)
cancer with disease limited to the pelvis. Additionally, we sought to 2 13 (4.3) 14 (6.7) 0 (0.0)
3 2 (0.7) 2 (1.0) 1 (3.4)
estimate the impact of relieving ureteral obstruction by ureteral stent Tumor size (cm) ≤2.0 2 (0.7) 0 (0.0) 0 (0.0)
or percutaneous nephrostomy in patients with stage IIIB cervical 2.1–6.0 127 (42.2) 84 (40.2) 10 (34.5)
cancer treated with chemoradiation. 6.1–10.0 152 (50.5) 112 (53.6) 17 (58.6)
≥10.1 17 (5.6) 11 (5.3) 2 (6.9)
Not reported 3 (1.0) 2 (1.0) 0 (0.0)
Materials and methods
Tumor grade 1 13 (4.3) 14 (6.7) 2 (6.9)
2 205 (68.1) 141 (67.5) 19 (65.5)
We retrospectively reviewed patients entered on four randomized 3 79 (26.2) 51 (24.4) 8 (27.6)
clinical trials performed by the Gynecologic Oncology Group (GOG) in Not graded 4 (1.3) 3 (1.4) 0 (0.0)
locally advanced stage disease (stages IIB–IVA) conducted from June Cell type Squamous cell carcinoma 277 (92.0) 195 (93.3) 28 (96.6)
Adeno-squamous 12 (4.0) 8 (3.8) 1 (3.4)
1981 to August 2000 [9–12]. These studies have all been reported carcinoma
previously and all utilized external and intracavitary radiation and Adenocarcinoma, NS 10 (3.3) 5 (2.4) 0 (0.0)
concurrent chemotherapy. It is important to note that eligibility for Other 2 (0.7) 1 (0.5) 0 (0.0)
these trials included a serum creatinine less than 2.0 mg/dl prior to Parametrial None 3 (1.0) 2 (1.0) 1 (3.4)
involvement Unilateral 148 (49.2) 109 (52.2) 14 (48.3)
enrollment. Information regarding the presence of unilateral or bilat-
Bilateral 142 (47.2) 95 (45.5) 12 (41.4)
eral parametrial disease, overall tumor size, whether hydronephrosis Not done 8 (2.7) 3 (1.4) 2 (6.9)
is present and if present is it unilateral or bilateral, and what inter- Pelvic node status Negative 163 (54.2) 94 (45) 16 (55.2)
ventions were utilized to correct it prior to initiation of therapy Positive 43 (14.3) 28 (13.4) 6 (20.7)
was abstracted from individual patient records. Additionally, data Not done 95 (31.6) 87 (41.6) 7 (24.1)
a
regarding the presence of pelvic nodal metastasis if sampled, maxi- Hyn = hydronephrosis, WR = with relief, WOR = without relief.
mum tumor diameter, use of cisplatin-containing chemotherapy
during radiation, the presence of hydronephrosis, and performance cumulative doses of radiation and cisplatin-based chemotherapy com-
status were abstracted and analyzed. The maximum tumor diameter pared to patients without hydronephrosis (median radiation dose to
(cm) was assessed as a continuous variable. Performance status was point A was 81 Gy for both groups and median number of cycles of
evaluated on an ordinal scale (0, 1, 2, and 3). Due to differences in cisplatin-based therapy was 5 for both groups). There was no signifi-
progression-free survival (PFS) and overall survival (OS) for cisplatin- cant difference in observed toxicity with cisplatin-containing therapy
containing chemotherapy regimens we examined adverse effects in the patients with or without hydronephrosis (Supplementary
of cisplatin-based chemotherapy in patients with relative to those Table 1). Both PFS and OS were significantly worse for patients with
without baseline hydronephrosis. hydronephrosis (p = 0.0189 and 0.0186, respectively) (Figs. 1 and 2).
Product-limit estimates were calculated according to Kaplan– Approximately 12% of patients presenting with hydronephrosis
Meier method and differences in PFS and OS were assessed utilizing had bilateral hydronephrosis. The median survivals were 18.6 and
the log-rank test [13,14]. The Cox model was used to adjust for prog- 31.7 months for patients with bilateral or unilateral hydronephrosis,
nostic factors (reported by Stehman et al.) and to estimate hazard respectively (Fig. 3). However, this difference was not statistically
ratios (and 95% confidence interval) of PFS and OS [15]. In this analysis, different (p value = 0.2537).
performance status of (2 or 3) was compared to (0 or 1). The incidence Univariable analysis identified five unfavorable prognostic factors;
of maximum adverse event grade among patients treated with plati- pelvic nodal metastasis (p = 0.0001), increasing maximum tumor
num based chemotherapy was summarized and tabulated by hydro- diameter (p = 0.0007), the lack of cisplatin-based concurrent chemor-
nephrosis status at baseline. Fisher's exact test was implemented to adiation (p = 0.0031), the presence of hydronephrosis (p = 0.0189),
determine associations between hydronephrosis status and baseline and performance status N1 (p = 0.0359) (Table 2A). Hydronephrosis
traits [16]. was associated with poorer GOG performance status (p b 0.001).
Adjusting for tumor size, pelvic node status, performance status and
Results treatment, hydronephrosis was not a significant prognostic factor
(Table 2B).
Five hundred and thirty-nine patients with stage IIIB cervical Ureteral obstruction relief by either stent or percutaneous
cancer from prior GOG protocols 56 (N = 100), 85 (N = 118), 120 nephrostomy occurred for 88% of patients presenting with hydrone-
(N = 220) and 165 (N = 101) were studied. The demographics of these phrosis. In particular, patients presenting without hydronephrosis
patients are presented in Table 1. Hydronephrosis was present in 238 showed the most favorable outcome (median PFS = 48 months,
stage IIIB patients (44.2%). Patient age, race, and tumor characteristics OS = 70 months) followed by those who got treatment for ureteral
(size, histology and grade) were not significantly different between obstruction (median PFS = 18 months and OS = 34 months). The
the groups. Patients presenting with hydronephrosis received similar group of patients presenting with hydronephrosis in whom ureteral
272 P.G. Rose et al. / Gynecologic Oncology 117 (2010) 270–275

Fig. 1. PFS by hydronephrosis among stage IIIB patients (log-rank test, p value = 0.0189).

obstruction was not invasively relieved before treatment had the basis of stage I substages (IA1–IB2) they are not utilized to assess stage
worst outcome (median PFS = 10 months and OS = 17 months). Both IIB–IVA disease. The presence of hydronephrosis among stage IIIB
PFS and OS were significantly different across groups (log-rank test p patients was associated with poorer PFS and OS in this study. This was
value = 0.0190 and 0.0078 respectively) (Figs. 4 and 5). seen despite the fact that patients with hydronephrosis received similar
doses of radiation and cisplatin-based chemotherapy. The presence of
Discussion hydronephrosis is not taken into consideration in the current clinical
staging system. Although not an independent prognostic variable on
The strengths of this study include the large number (539) of stage multivariable analysis, hydronephrosis is an easily determined factor
IIIB cervical cancer patients treated in four prospective randomized associated with poorer performance status and poorer OS in univariable
trials utilizing standardized radiation doses and schedules. Prognostic analysis.
factors identified included pelvic nodal metastasis, tumor diameter, Logsdon and Eifel reported the MD Anderson experience with stage
cisplatin-based concurrent chemoradiation, hydronephrosis, and IIIB cervical cancer from 1960 to 1993 [2]. Of 983 patients treated with
performance status. The FIGO staging system is a clinical staging curative intent fewer than 5% received concurrent chemotherapy.
system which is widely applicable but lacks recognition of any of the Thirty percent (291) had hydronephrosis and these patients had a
above identified prognostic factors. While tumor dimensions are the poorer disease specific survival than patients without hydronephrosis

Fig. 2. OS by hydronephrosis among IIIB patients (log-rank test, p value = 0.0186).


P.G. Rose et al. / Gynecologic Oncology 117 (2010) 270–275 273

Fig. 3. OS by hydronephrosis type (log-rank test, p value = 0.2537).

(28% vs. 40%). Improved survival was noted in recent years with the Ureteral obstruction from cervical cancer occurs most often in the
routine use of intracavitary radiation [2]. The outcome for patients cardinal ligament 3–6 cm proximal to the ureterovesical junction
with bilateral vs. unilateral hydronephrosis was not different. The [12]. A number of surgical procedures can be employed to relieve
management of patients with hydronephrosis was not addressed in this obstructive uropathy with endoscopic retrograde ureteral catheteri-
study. Chao et al. reported the Millinckrodt Institutes experience with zation, surgical nephrostomy, surgical ureterostomy, and percutane-
hydronephrosis in 297 patients with stage IIIB cervical cancer [3]. These ous nephrostomy most commonly employed. However, endoscopic
authors noted a poorer outcome and a higher distant failure rate for retrograde ureteral catheterization is not technically feasible in over
stage IIIB patients that did not reach the sidewall in whom ureteral 70% of patients with malignant ureteral obstruction [17]. Percutaneous
obstruction was presumably due to nodal metastasis. In contrast for nephrostomy is currently the preferred supravesical diversion because
patients with sidewall extension no difference in survival was noted for of its minimal morbidity and mortality [18,19].
patients with or without hydronephrosis. Taylor and Anderson studied 18 patients from two institutions
The poor survival of patients with primary cervical cancer and with untreated cervical cancer and obstructive uropathy treated with
obstructive uropathy (0–50%) has been documented in previous either medical (hemodialysis and peritoneal dialysis) or surgical
reports [2–6]. Many of these patients have been treated with palliative management and found no method clearly superior [5]. Horan et al.
intent [4]. Since the number of patients with primary cervical cancer reported their retrospective experience with 13 patients; five with
and obstructive uropathy treated with curative intent with radiation unilateral and eight with bilateral ureteral obstruction [6]. Eight
and concurrent chemotherapy in previous reports is small, optimal required ureteral stenting which was usually performed prior to
management is not established. However, the patient with ureteral radiation therapy. Ureteral stenting during radiation resulted in
obstruction is often a candidate for pelvic radiation therapy with significant delays in treatment. They also prospectively studied three
curative intent. The current series is unique in that all patients in- patients with unilateral ureteral obstruction whose renal function and
cluded in this study were enrolled in National Cancer Institute trials hydronephrosis during treatment remained stable or improved with
and treated with curative intent.
Table 2B
Multivariate analysis of prognostics factors by Cox proportional hazard regression.

Table 2A Factor OS PFS


Univariate analysis of prognostics factors by Cox proportional hazard regression. HR (95% CI) p HR (95% CI) p
Factor OS PFS Tumor size (cm) 1.079 (1.018–1.143) 0.0099 1.059 (1.000–1.122) 0.0459
Performance 2.380 (1.276–4.438) 0.0064 1.790 (0.965–3.320) 0.0647
HR (95% CI) p HR (95% CI) p
status
Tumor size (cm) 1.077 (1.032–1.125) 0.0007 1.069 (1.024–1.116) 0.0025 (2, 3 vs. 0, 1)
Performance status 1.560 (1.030–2.364) 0.0359 1.501 (0.999–2.254) 0.0506 Pelvic node status 1.857 (1.378–2.504) b 0.0001 1.726 (1.285–2.318) 0.0003
(2, 3 vs. 0, 1) (positive vs.
Pelvic node status 1.780 (1.326–2.390) 0.0001 1.671 (1.248–2.237) 0.0006 negative)
(positive vs. Hydronephrosis 1.244 (0.945–1.639) 0.1199 1.248 (0.955–1.629) 0.1045
negative) (yes vs. no)a
Hydronephrosis 1.291 (1.043–1.597) 0.0189 1.282 (1.041–1.579) 0.0193 Chemotherapy 0.747 (0.568–0.981) 0.0362 0.745 (0.570–0.973) 0.0305
(yes vs. no) (cisplatin vs.
Chemotherapy 0.722 (0.582–0.896) 0.0031 0.718 (0.582–0.887) 0.0021 other)
(cisplatin vs. a
In the multivariate analysis, when adjusting for tumor size, pelvic node status,
other)
performance status and treatment, hydronephrosis was no longer significant.
274 P.G. Rose et al. / Gynecologic Oncology 117 (2010) 270–275

Fig. 4. PFS by hydronephrosis relief status (log-rank test, p value = 0.0190).

time. Two of these three received concurrent cisplatin chemotherapy. tion doses were able to be given. Eligibility for the trials in this report
The authors recommended elective urinary diversion for patients included a serum creatinine less than 2.0 mg/dl prior to enrollment. Our
prior to radiation therapy for patients with bilateral obstruction and a analysis of the impact of hydronephrosis in this trial is limited to
creatinine clearance b50 ml/min. patients whose renal function was either normal or improved with a
Ureteral obstruction may compromise the ability to deliver poten- nephrostomy or ureteral stent prior to enrollment. Therefore, patients
tially nephrotoxic cisplatin chemotherapy concurrently with radiation whose renal function was more abnormal or failed to improve are not
therapy which has been demonstrated in randomized trials to be supe- represented in this study. Additional weaknesses of this study are
rior to radiation alone or radiation and non-cisplatin chemotherapy. variations in radiation therapy techniques in the most recent study
However, in the current study the presence of hydronephrosis did not (Protocol #165) including a slightly higher (5%) radiation dose, more
limit the doses of cisplatin-containing chemotherapy and similar radia- compacted treatment time and the use of high dose-rate brachytherapy.

Fig. 5. OS by hydronephrosis relief status (log-rank test, p value = 0.0078).


P.G. Rose et al. / Gynecologic Oncology 117 (2010) 270–275 275

Patients with FIGO stage IIIB cervical cancer and hydronephrosis [2] Logsdon MD, Eifel PJ. FIGO IIIB squamous cell carcinoma of the cervix: an analysis
of prognostic factors emphasizing the balance between external beam and
had a shorter PFS and OS than those whose stage was based only on intracavitary radiation therapy. Int J Radiat Oncol Biol Phys 1999;43:763–75.
palpation of cancer extension to the pelvic sidewall. Thus, the pres- [3] Chao KS, Leung WM, Grigsby PW, Mutch DG, Herzog T, Perez CA. The clinical
ence of hydronephrosis is a clinical surrogate of poorer performance implications of hydronephrosis and the level of ureteral obstruction in stage IIIB
cervical cancer. Int J Radiat Oncol Biol Phys 1998;40:1095–100.
status and poorer OS. [4] Krynicki R, Bidzinski M, Wierzba W, Panek G, Jonska J, Lindner B. Hydronephrosis
as a prognostic factor for patients with FIGO IIIB cervical cancer. Ginekol Pol 2003;74:
32–9.
Conflict of interest statement [5] Taylor Jr PT, Anderson WA. Untreated cervical cancer complicated by obstructive
The authors have no conflicts of interest to declare. uropathy and olioguric renal failure. Gynecol Oncol 1981;11:162–74.
[6] Horan G, McArdle O, Martin J, Collins CD, Faul C. Pelvic radiotherapy in patients
with hydronephrosis in stage IIIB cancer of the cervix: renal effects and the
Acknowledgments optimal timing of urinary diversion. Gynecol Oncol 2006;101:441–4.
[7] Grigsby PW. The prognostic value of PET and PET/CT in cervical cancer. Cancer
Imaging 2008;8:146–55.
This study was supported by National Cancer Institute grants to [8] Gold MA, Tian C, Whitney CW, Rose PG, Lanciano R. Surgical versus radiographic
the Gynecologic Oncology Group Administrative Office (CA 27469) determination of para-aortic lymph node metastases before chemoradiation for
and the Gynecologic Oncology Group Statistical Office (CA 37517). locally advanced cervical carcinoma: a Gynecologic Oncology Group study. Cancer
2008;112:1954–63.
The following Gynecologic Oncology Group member institutions
[9] Stehman FB, Bundy BN, Thomas G, Keys HM, d'Ablaing III G, Fowler Jr WC, et al.
participated in the primary treatment studies: University of Alabama Hydroxyurea versus misonidazole with radiation in cervical carcinoma: long-term
at Birmingham, Oregon Health Sciences University, Duke University follow-up of a Gynecologic Oncology Group trial. J Clin Oncol 1993;11:1523–8.
Medical Center, Abington Memorial Hospital, University of Rochester [10] Whitney CW, Sause W, Bundy BN, Malfetano JH, Hannigan EV, Fowler Jr WC, et al.
A randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an
Medical Center, Walter Reed Army Medical Center, University of adjunct to radiation therapy in stages IIB–IVA carcinoma of the cervix with nega-
Minnesota Medical School, University of Southern California at Los tive para-aortic lymph nodes. A Gynecologic Oncology Group and Southwest
Angeles, University of Mississippi Medical Center, Colorado Gyneco- Oncology Group study. J Clin Oncol 1999;17:1339–48.
[11] Rose PG, Bundy BN, Watkins EB, Thigpen JT, Deppe G, Maiman MA, et al. Concurrent
logic Oncology Group P.C., University of California at Los Angeles, cisplatin-based chemoradiation improves progression free and overall csurvival in
University of Miami School of Medicine, Milton S. Hershey Medical advanced cervical cancer: results of a randomized Gynecologic Oncology Group
Center, Georgetown University Hospital, University of Cincinnati, study. N Engl J Med 1999;340:1144–53.
[12] Lanciano R, Calkins A, Bundy B, Parham G, Lucci III JA, Moore DH, et al. Randomized
University of North Carolina School of Medicine, University of Iowa comparison of weekly cisplatin or protracted venous infusion of fluorouracil in
Hospitals and Clinics, Indiana University School of Medicine, Wake combination with pelvic radiation in advanced cervix cancer: a Gynecologic
Forest University School of Medicine, Albany Medical College, Oncology Group study. J Clin Oncol 2005;23:8289–95.
[13] Bland JM, Altman DG. The logrank test. BMJ 2004;328:1073.
University of California Medical Center at Irvine, Rush-Presbyterian- [14] Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am
St. Luke's Medical Center and Eastern Virginia Medical School. Stat Assoc 1958;53:457–81.
[15] Stehman FB, Bundy BN, DiSaia PJ, Keys HM, Larson JE, Fowler WC. Carcinoma of the
cervix treated with radiation therapy I: a multi-variate analysis of prognostic
Appendix A. Supplementary data variables in the Gynecologic Oncology Group. Cancer 1991;67:2776–85.
[16] Agresti A. A survey of exact inference for contingency tables. Stat Science 1992;7:
131–77.
Supplementary data associated with this article can be found, in [17] Kearney GP, Tumeh SS. Urinary tract involvement in gynecologic cancer. In: Knapp
the online version, at doi:10.1016/j.ygyno.2010.01.045. RC, Berkowitz RS, editors. Gynecologic Oncology. 2nd ed. New York: McGraw-Hill;
1993. p. 341.
[18] Zadra JA, Jewett MA, Keresteci AG, Rankin JT, St. Louis E, Grey RR, et al. Nonoperative
References urinary diversion for malignant ureteral obstruction. Cancer 1987;60:1353–7.
[19] Soper JT, Blaszczyk TM, Oke E, Clarke-Pearson D, Creasman WT. Percutaneous
[1] NCI Clinical Announcement, U.S. Dept. of Health and Human Services, Public nephrostomy in gynecologic oncology patients. Am J Obstet Gynecol 1988;158:
Health Service, National Institutes of Health, February 1999. 1126–31.
Clinical Review & Education

JAMA Clinical Guidelines Synopsis

Cervical Cancer Screening Guideline for Individuals at Average Risk


Julie Chor, MD, MPH; Andrew M. Davis, MD, MPH; Jennifer M. Rusiecki, MD

Evidence Base
GUIDELINE TITLE Cervical Cancer Screening for Individuals The 2020 ACS guideline updates the 2012 ACS guideline for individu-
at Average Risk: 2020 Guideline Update From the als at average risk, defined as those with a cervix in situ who are ini-
American Cancer Society tiating cervical cancer screening, have normal cervical cancer screen-
ing results in the past, or who have been returned to routine cervical
RELEASE DATE July 30, 2020 cancer screening based on standard follow-up recommendations.4
The recommendations do not apply to individuals at increased risk for
PRIOR VERSION 2012
cervical cancer due to solid organ or stem cell transplant, HIV infec-
tion, immunosuppression from other causes, or in utero exposure
DEVELOPER AND FUNDING SOURCE American Cancer
to diethylstilbestrol.4
Society (ACS)
The 2020 ACS guideline includes 3 important changes: (1) the
TARGET POPULATION Individuals with cervix in situ age of initial screening is increased to 25 years (previously 21 years);
(2) the interval for testing is extended to every 5 years (previously
MAJOR RECOMMENDATIONS 3 years); and (3) an updated recommendation for initial screening
• Begin screening at age 25 years regardless of sexual history approach deemphasizes the 2012 guideline’s use of cervical
or HPV vaccination status (strong recommendation) cytology.4 The 2020 guidelines recommend primary HPV testing
• Primary HPV testing every 5 years through age 65 years alone every 5 years, with the option of cotesting with cytology and
(strong recommendation) HPV testing every 5 years or cytology alone every 3 years when pri-
• If primary HPV testing is not available use cotesting mary HPV testing is not available.4 The reason that cotesting may
(HPV+ cytology) every 5 years or every 3 years if cytology be available when primary HPV testing is not available is that the spe-
only (strong recommendation) cific HPV testing performed in a given hospital laboratory may not
• Discontinue screening at age 65 years if no history of be approved for primary HPV testing. There is no change to the prior
cervical intraepithelial neoplasia grade 2 or more severe recommendation to stop screening at age 65 years.
diagnosis in last 25 years and adequate negative prior Importantly, these recommendations do not differ by HPV vacci-
screening in last 10 years (qualified recommendation) nation status despite data from Sweden that demonstrated a decrease
in cervical cancer rates from 94 cases/100 000 persons among unvac-
cinated people to 47/100 000 persons among a widely vaccinated
population.5 While specific risk reduction to US populations cannot be
Summary of the Clinical Problem directlyinferred,theguidelinestates,“futurerecommendationsforcer-
Even though screening has significantly reduced cervical cancer– vicalcancerscreeningwillneedtoincorporateHPVvaccinationstatus.”4
related deaths in the US, an estimated 14 480 individuals will be di-
The ACS 2020 guideline was informed primarily by 2 reports
agnosed with cervical cancer and an estimated nearly 4300 will die
commissioned by the US Preventive Services Task Force (USPSTF)
from this disease during 2021.1 Worldwide, cervical cancer is the
2018 cervical cancer screening update: a systematic review and a
fourth most common cancer in women and is the leading cause of mathematical disease-simulation model,6,7 which accompanied the
cancer-related deaths among women in Africa.2 In the US, Black
USPSTF recommendation statement.8 The decision to delay initia-
women experience significantly higher cervical cancer mortality rates
tion of screening reflects the low incidence and mortality of cervi-
than White women,3 and disruptions of care during the COVID pan-
cal cancer among individuals younger than 25 years, the high inci-
demic have affected cancer screening in general. Testing for cervi- dence of transient HPV infections among those younger than 25
cal infection with high-risk human papillomavirus (HPV) has much years, and the potential for treatment of cervical abnormalities
higher sensitivity than traditional cytology for the dysplastic cervi- to result in adverse obstetric events (eg, premature delivery).4
cal changes that precede cancer, making this approach an increas-
ingly important prediction tool for invasive cervical cancer. The 2020 Table. Guideline Rating
American Cancer Society (ACS) Cervical Cancer Screening Guide-
Standard Rating
line provides the latest recommendations, including when to initi-
Establishing transparency Good
ate screening, frequency of testing, and which modality to use.4 Management of conflict of interest in the guideline development group Good
Guideline development group composition Good
Characteristics of the Guideline Source Clinical practice guideline-systematic review intersection Good
A volunteer ACS guideline development group (GDG) of 18 individu- Establishing evidence foundations and rating strength for each of the Good
als with wide expertise including gynecology, oncology, and pri- guideline recommendations
mary care reviewed the evidence, supported modeling analyses, and Articulation of recommendations Good
received input from various groups, including many professional so- External review Good

cieties and patient-based organizations.4 Required disclosures in- Updating Good

cluded financial and nonfinancial relationships (Table). Implementation issues Fair

jama.com (Reprinted) JAMA Published online November 12, 2021 E1

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Mexico | Access Provided by JAMA User on 11/21/2021


Clinical Review & Education JAMA Clinical Guidelines Synopsis

The recommendation to use primary HPV testing for screening was Discussion
based on data that showed increased sensitivity and long-term nega- The2020ACSguidelines4 andthe2018USPSTFcervicalcancerscreen-
tive predictive value of HPV-based screening compared with cytol- ingguidelines8 differonageofinitiation,frequency,andmodality.Based
ogy alone, as well as the approval of primary HPV screening tests on additional analyses obtained through systematic review and deci-
by the US Food and Drug Administration.4 sion analysis, the 2020 ACS guidelines aim to incorporate technologi-
The ACS GDG commissioned a decision analysis from the model- cal advances, increase effectiveness, and decrease potential risks and
ing group that supported the 2018 USPSTF cervical cancer screening burdens of cervical cancer screening (discomfort, anxiety, and addi-
update.Modelinganalysescomparedseveralscreeningstrategies,vary- tional procedures) compared with the 2018 USPSTF guidelines.
ing age of initiation (21 or 25 years), interval (3, 4, or 5 years), and mo- Clinicians who wish to optimize implementation of the 2020 ACS
dality (cytology alone, cotesting, or primary HPV). The models com- guidelinesandreconciletheserecommendationswiththe2018USPSTF
pared outcomes based on effectiveness (ability to identify treatable guidelinesareencouragedtoengagepatientsinshareddecision-making
cervicalabnormalities)andefficiency(minimizingunnecessaryscreen- regarding medical considerations, risk of HPV exposure, HPV vacci-
ing and colposcopies per life-year gained and cancer averted). nation status, and prior abnormal cervical cancer screening results.
All strategies conclude screening at age 65 for individuals with- Ideally,thesediscussionswillexplorepatients’values,experiences,and
out a history of cervical intraepithelial neoplasia (CIN) 2 or worse in preferences regarding cervical cancer screening and their comfort with
the past 25 years, and with documented adequate screening in the pelvic examinations. Fostering strong patient-clinician relationships in
past 10 years, defined as 2 consecutive tests within the past 10 years, earlyadulthoodmaykeeppatientsengagedinpreconceptualreproduc-
with the most recent within the past 3 to 5 years, depending on the tive health care and also promote earlier prenatal care.
test used. This recommendation holds even if a new sexual partner Theseconsiderationsargueforplanningandpoliciestoengageado-
of any age is reported after age 65. Individuals of any age who have lescents and adults younger than 25 years in preventive reproductive
had surgical removal of the cervix (and no history of CIN 2 or more health care. This need is even more salient in vulnerable communities,
severe diagnosis) should not be screened with cytology or HPV test- givenlong-standingracialandethnicreproductivehealthdisparities,in-
ing for lower genital tract malignancies, such as vaginal cancer. cludinglowerratesofcontraceptiveuseandHPVvaccination,andhigher
rates of STIs and reproductive cancers. Such inequities and barriers to
Benefits and Harms care result in a greater risk of preterm and low-birth-weight deliveries
The GDG prioritized reduction in cervical cancer cases while maxi- and markedly higher maternal morbidity and mortality.10
mizing life-years gained and minimizing unnecessary screening and
colposcopies. Delaying the starting age for cervical cytology test- Areas in Need of Future Study or Ongoing Research
ing (ie, Papanicolaou testing) may lessen emotional harms associ- Additional research is needed to evaluate how guideline implemen-
ated with low-risk screening, including anxiety and distress from ex- tation affects key outcomes, including initiation and adherence with
aminations, and may reduce the economic and emotional harms that Pap testing, access to effective contraception and timely and trusted
may occur after interventions in response to less serious and tran- prenatal care, and STI rates in persons younger than 25 years. Evalu-
sient cytopathologic findings. ation should include assessment for potentially widening racial and
A potential concern is that the first Pap test brings many indi- ethnic and socioeconomic inequities in reproductive health. The psy-
viduals in for their first preventive reproductive health visit. Wait- chosocial value of delayed screening initiation and less frequent pel-
ing until age 25 years for initial screening may delay counseling and vic examinations, particularly in those with a history of painful ex-
screening for effective contraception and intimate partner vio- aminations or trauma, deserves study. Finally, because the new ACS
lence, as well as service delivery around healthy sexuality. Guide- guideline does not incorporate vaccination status in its recommen-
lines suggesting a later age of screening have been linked to re- dations, monitoring of cervical cancer detection in populations with
duced screening for sexually transmitted infections (STIs).9 varying degrees of HPV vaccine coverage will be important.

ARTICLE INFORMATION 2. Arbyn M, Weiderpass E, Bruni L, et al. Estimates papillomavirus testing: updated evidence report
Author Affiliations: Department of Obstetrics and of incidence and mortality of cervical cancer in and systematic review for the US Preventive
Gynecology, University of Chicago, Chicago, Illinois 2018. Lancet Glob Health. 2020;8(2):e191-e203. Services Task Force. JAMA. 2018;320(7):687-705.
(Chor); Section of General Internal Medicine, doi:10.1016/S2214-109X(19)30482-6 doi:10.1001/jama.2018.10400
Department of Medicine, University of Chicago, 3. Beavis AL, Gravitt PE, Rositch AF. 7. Kim JJ, Burger EA, Regan C, Sy S. Screening for
Chicago, Illinois (Davis, Rusiecki). Hysterectomy-corrected cervical cancer mortality cervical cancer in primary care: a decision analysis
Corresponding Author: Andrew M. Davis, MD, rates reveal a larger racial disparity in the United for the US Preventive Services Task Force. JAMA.
MPH, Section of General Internal Medicine, States. Cancer. 2017;123(6):1044-1050. doi:10. 2018;320(7):706-714. doi:10.1001/jama.2017.19872
Department of Medicine, University of Chicago, 1002/cncr.30507 8. US Preventive Services Task Force. Screening for
5841 S Maryland Ave, Chicago, IL 60637 4. Fontham ETH, Wolf AMD, Church TR, et al. cervical cancer: US Preventive Services Task Force
(amd@uchicago.edu). Cervical Cancer Screening for Individuals at Average Recommendation Statement. JAMA. 2018;320(7):
Published Online: November 12, 2021. Risk: 2020 guideline update from the American 674-686. doi:10.1001/jama.2018.10897
doi:10.1001/jama.2021.13448 Cancer Society. CA Cancer J Clin. 2020;70(5):321-346. 9. Ursu A, Sen A, Ruffin M. Impact of cervical
doi:10.3322/caac.21628 cancer screening guidelines on screening for
Conflict of Interest Disclosures: None reported.
5. Lei J, Ploner A, Elfström KM, et al. HPV chlamydia. Ann Fam Med. 2015;13(4):361-363.
REFERENCES vaccination and the risk of invasive cervical cancer. 10. Sutton MY, Anachebe NF, Lee R, Skanes H.
N Engl J Med. 2020;383(14):1340-1348. Racial and ethnic disparities in reproductive health
1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer
statistics, 2021. CA Cancer J Clin. 2021;71(1):7-33. 6. Melnikow J, Henderson JT, Burda BU, et al. services and outcomes, 2020. Obstet Gynecol.
doi:10.3322/caac.21654 Screening for cervical cancer with high-risk human 2021;137(2):225-233.

E2 JAMA Published online November 12, 2021 (Reprinted) jama.com

© 2021 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by a Mexico | Access Provided by JAMA User on 11/21/2021


European Journal of Cancer (2014) 50, 1628– 1637

Available at www.sciencedirect.com

ScienceDirect
journal homepage: www.ejcancer.com

Effects of hormone replacement therapy on the rate


of recurrence in endometrial cancer survivors: A
meta-analysis

Seung-Hyuk Shim, Sun Joo Lee, Soo-Nyung Kim ⇑

Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, Republic of Korea

Received 12 December 2013; received in revised form 3 March 2014; accepted 6 March 2014
Available online 28 March 2014

KEYWORDS Abstract Background: To quantify the effect of hormone replacement therapy (HRT) on the
Endometrial cancer recurrence in endometrial cancer (EC) survivors through a meta-analysis.
Hormone replacement Methods: A systematic literature review was conducted through October 2013 and included
therapy studies reporting estimates of effect size for the relationship between HRT use and the risk
Recurrence of EC recurrence. Study design features that may affect the selection of participants, the detec-
Meta-Analysis tion of EC recurrence and manuscript publication were assessed. If there was no significant
statistical heterogeneity across studies, then a fixed effects model was used to obtain pooled
estimates for the effect of HRT use on EC recurrence by combining study-specific estimates
of the odds ratio (OR).
Results: One randomised trial and five observational studies included 896 EC survivors who
used HRT and 1079 non-users. Over the combined study period, 19 of the 896 HRT users
experienced recurrence, whereas 64 of the 1079 controls did. The meta-analysis based on
the fixed effects model indicates no significant increase in the risk of recurrence in EC survi-
vors using HRT relative to the control group (OR: 0.53; 95% confidence interval: 0.30–0.96,
I2 = 49.0). This pattern was also observed in the subgroup analysis for the stage and type of
HRT. There was no evidence of any publication bias.
Conclusions: Although based mainly on observational studies, the literature does not provide
support for a positive relationship between HRT use and the risk of EC recurrence. Future
research should verify this relationship through randomised controlled trials over a longer term.
Ó 2014 Elsevier Ltd. All rights reserved.

⇑ Corresponding author: Address: Department of Obstetrics and Gynecology, Konkuk University School of Medicine, 1 Hwayang-dong,
Gwangjin-gu, Seoul 143-701, Korea. Tel.: +82 2 2030 7641; fax: +82 2 2030 7748.
E-mail address: snkim@kuh.ac.kr (S.-N. Kim).

http://dx.doi.org/10.1016/j.ejca.2014.03.006
0959-8049/Ó 2014 Elsevier Ltd. All rights reserved.
S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637 1629

1. Introduction All searches were performed independently by the two


authors (S.S. and S.L.).
Endometrial cancer (EC) is the most common gyne-
cologic malignancy in Western countries. In the United 2.2. Eligibility criteria
States (US), 47,130 cases were newly diagnosed in
2012, and 8010 women died of EC in the same year Studies were included if they met the following crite-
[1]. The number of EC patients has increased rapidly ria: (1) randomised controlled trials, a prospective or
in Korea, where it was third most common gyneco- retrospective cohort study, a nested case-control study,
logic malignancy with 1616 new estimated cases in or a population-based case-control study; (2) partici-
2010 [2]. pants of interest being patients receiving surgical treat-
Surgical staging procedures such as hysterectomy and ment for EC; (3) the intervention of interest being
bilateral salpingooophorectomy represent the corner- HRT exposure and (4) the outcome measure being EC
stone treatment option for EC. Although EC is gener- recurrence measured by the relative risk (RRs), the odds
ally a postmenopausal disease, about 25% of EC ratio (ORs) or the hazard ratio (HRs) estimated with the
patients are premenopausal [3]. Therefore, menopausal 95% confidence interval (CI) (or sufficient data for cal-
symptoms, particularly in reproductive women undergo- culating them). Single-arm cohort studies were not
ing surgical treatment for EC, are common. In addition, included in the meta-analysis.
menopausal symptoms caused by surgery tend to be
more severe than those resulting from normal meno- 2.3. Data extraction
pause [4]. Accordingly, relieving these symptoms is an
important issue in terms of the quality of life after treat- Three authors (S.S., S.L. and S.K.) independently
ment because a majority of EC patients show an early- extracted data, including the name of the first author,
stage disease and a favourable prognosis. the year of publication, the study design, the study loca-
Although no scientific data demonstrate the detri- tion, the study period, age, sample size (cases and con-
mental effect of hormonal replacement therapy (HRT) trols or cohort size), tumour stage, grade, exposure to
in EC survivors, many clinicians remain reluctant to pre- HRT use, disease-free interval (DFI) between the initial
scribe HRT for these patients because of the concern EC diagnosis and HRT administration, type of HRT,
over oestrogen use as an important aetiologic factor duration of HRT, follow-up after HRT administration,
for EC [5]. Without estimates of the effect of HRT on EC recurrence and covariates controlled for in the anal-
EC recurrence, it is difficult for EC survivors and their ysis. When not reported, the follow-up after the initia-
physicians to weigh the risk and benefits of HRT. In this tion of HRT was assumed to equal the duration of
regard, the present study provides a meta-analysis to HRT use. Each study was systematically reviewed for
estimate the effect of HRT on EC recurrence in patients features that could introduce some bias, including pro-
previously treated for EC by considering observational cedures for identifying participants, whether risk factors
and randomised studies. for recurrence were similar at diagnosis, and whether the
follow-up duration was similar between HRT users and
non-users. Discrepancies in data extraction were jointly
2. Materials and methods reviewed until a consensus was reached.

2.1. Literature search 2.4. Quality assessment

A systematic review was conducted using developed Study quality was evaluated independently by two
guidelines for reporting a systematic review [6,7]. MED- authors (S.S. and S.L.) by using the nine-star Newcas-
LINE and EMBASE databases and the Cochrane Cen- tle–Ottawa scale (NOS) [8]. The NOS assesses study
tral Register for Controlled Trials were searched with no quality in three categories: selection, comparability and
language limitations up to 15th October 2013. Papers exposure (case-control studies) or outcomes (cohort
ahead of their publication were also included. The fol- studies). Nine points on the NOS reflects the highest
lowing keywords were used for this search: ‘endometrial study quality. Based on quality assessment standards in
neoplasm or cancer or carcinoma or malignancy,’ ‘uter- previous meta-analyses [9], a study awarded five or more
ine neoplasm or cancer or carcinoma or malignancy,’ stars was defined to be of high quality in the present
‘oestrogen,’ ‘oestrogen replacement therapy,’ ‘hormone meta-analysis. Any disagreement was resolved after a
replacement therapy,’ and ‘estradiol.’ The titles and discussion and reevaluation with the third author (SK).
abstracts were checked to exclude any clearly unrelated
articles. The full text of remaining papers was evaluated 2.5. Data synthesis and analysis
to determine their relevancy. In addition, the references
cited in selected papers and published reviews were con- From the original study data, the OR and the 95% CI
sidered to include any additional studies of relevance. were calculated for each study for the recurrence rate for
1630 S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637

HRT users and non-users. Heterogeneity across studies one was a letter to the editor [23], two were case series
was examined using I2, which measures the percentage without control groups [24,25] and one was a study from
of the total variation across studies [10]. Here substan- duplication GOG data used in the study by Barakat
tial heterogeneity was defined as an I2 value greater than et al. [26]. These were excluded from further analysis.
50% [11]. In the absence of significant heterogeneity, a The remaining six reported on HRT exposure and the
fixed effects model was used. When there was statistical EC recurrence and thus were included in the meta-
heterogeneity, a random effects model was used to esti- analysis [16–21].
mate the combined OR for randomised and observa-
tional studies. Then a subgroup analysis was
conducted for the type of study design a [randomised 3.2. Study characteristics
control trial (RCT) or a non-randomised observational
study (NRS)], HRT (oestrogen only or oestrogen plus A total of six papers were published between 1986
progestin) and stage (stage I, stages I and II or all and 2006, including a total of 896 EC survivors with
stages). The subgroup analysis was planned a priori HRT and 1079 without. One was a double-blind RCT
before data were collected and analysed. [20] and five were NRSs [16–19,21]. The studies were
A sensitivity analysis was conducted to investigate the from the US [16–20] and Turkey [21] and assessed to
robustness of conclusions by eliminating each study in range from six to eight on the nine-star NOS (Supple-
the meta-analysis one at a time to determine its effect mental Table, online only). In the selection category,
on the pooled OR. Publication bias was evaluated using all the included studies were awarded three stars. How-
the fail-safe N test [12] and the Begg–Mazumdar rank ever, only half of the included studies [19–21] attempted
correlation test [13]. A funnel plot was built to assess to adjust confounding factors by restrictive inclusion cri-
this bias by using the standard error and the diagnostic teria. For the comparability category, five studies [16–
OR [14,15]. Comprehensive Meta-Analysis version 2.0 19,21] were awarded one star because age and/or stage
(Biostat, Englewood, NJ, US) was used for all statistical were controlled. One study [20] was awarded two stars
tests. P < 0.05 was considered significant for this meta- as they satisfied both comparability criteria. For the
analysis. Data from this meta-analysis are presented exposure assessment category, two studies [16,18] lost
according to the checklist based on the Meta-Analysis stars for not adequately presenting loss to follow-up.
of Observational Studies in Epidemiology [6]. Table 1 shows the study characteristics in detail.
In two studies, the type of HRT was oestrogen only
[16,20]. That of HRT was combined oestrogen plus pro-
3. Results gestin in another study [21]. In the other studies, that of
HRT was a mixture of oestrogen only and combined
3.1. Literature search oestrogen plus progestin [17–19]. Five studies reported
similar stages and grade distributions for HRT users
Fig. 1 presents a flow diagram showing how relevant and non-users at the beginning of the observation period
studies were identified. A total of 11 potentially relevant [16,18–21], although one reported a higher number of
papers were found by focusing on HRT use and EC high-risk (stage IC, grade 3) patients in the control
recurrence [16–26]. Among those, one was an incom- group (37% of non-users) [17]. Not all studies reported
plete study providing no exact recurrence data [22], the DFI before initiating HRT in the control group,

Fig. 1. Flow diagram of the procedure for the literature search.


Table 1
Characteristics of studies included in the meta-analysis.
Study/design Location/ Treatment Mean Stage Grade HRT type Oestrogen HRT route Mean Mean Mean Mean Recurrence Adjusted variables
study age alone duration follow- DFI follow-
period (yr) of HRT up before up
(Mo) period HRT period
(Mo) (Mo) after
HRT
Creasman et al. [16]/ US/ HRT 57* IA-30 1–29 Conjugated oestrogen 47 (100%) Oral-7 26* 60* 15* 32* 1 Stage, LN, grade, depth of
Retrospective 1975– (n = 47) IB-17 2–13 only (0.625 or Vaginal-34 myometrial invasion, cytology
case-control study 1980 3–5 1.25 mg/d) Both-6
No HRT 62* IA- 1–87 42* 26
(n = 174) 115 2–60

S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637


IB-57 3–27
Lee et al. [17]/ US/ HRT 55.7 IA-24 1, 2–44 Conjugated oestrogen 29 (66%) Oral-44 (100%) 64* 87* <12 Mo: 64*,  0 Stage
Retrospective 1975– (n = 44) IB-20 3–0 (0.625 or 1.25 mg/d) 57%
case-control study 1985 with or without 13–
progestin 24 Mo:
9%
25–
60 Mo:
21%
>60 Mo:
13%
No HRT 60.1 IA-24 1, 2–87 63* 8
(n = 99) IB-43 3–12
IC-31
Chapman et al. [18]/ US/ HRT 57.5 IA, B- 1–39 Conjugated oestrogen 29 (47%) Oral-57 39.5§ 57.1 8* 39.5 2 Preoperative HRT, radiation
Retrospective 1982– (n = 62) 54 2–17 (0.625 or 1.25 mg/d or Vagina-1 (<12 Mo:
case-control study 1994 IC-6 3–6 transdermal patch) Oral + Vaginal- 60%
II-2 with or without 4 13–
progesterone (oral, 24 Mo:
2.5 mg/d) 19.4%
>25 Mo:
21%)
No HRT 69.3 IA, B- 1–25 39.1 6
(n = 61) 43 2–26
IC-9 3–10
II-9
Suriano et al. [19]/ US/ HRT 59.7 IA-14 1–31 Conjugated equine 38 (51%) Oral-75 (100%) NR 83 <6 Mo: NR 2 Age, stage
Retrospective 1984– (n = 75) IB-44 2–29 oestrogen (oral, 43 (57%)
matched cohort 1998 IC-6 3–15 0.625 mg/d) with or 7–12 Mo:
study IIA-4 without 12 (16%)
IIB-3 medroxyprogesterone >12 Mo:
IIIA- acetate (oral, 2.5 mg/ 20 (27%)
3IIIC- d)
1
(continued on next page)

1631
1632
Table 1 (continued)
Study/design Location/ Treatment Mean Stage Grade HRT type Oestrogen HRT route Mean Mean Mean Mean Recurrence Adjusted variables
study age alone duration follow- DFI follow-
period (yr) of HRT up before up
(Mo) period HRT period
(Mo) (Mo) after
HRT
No HRT 61.1 IA-14 1–24 69 11
(n = 75) IB-43 2–32
IC-7 3–19
IIA-5
IIB-2
IIIA-

S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637


3
IIIC-1

Ayhan et al. [21]/ Turkey/ HRT 51.2 I-44 1–40 Conjugated equine 0 Oral-50 (100%) 49.1 49.1 <1–2 Mo: 49.1  0 Age, parity, systemic disease,
Prospective 1992– (n = 50) II-6 2–7 oestrogen (oral, 50 (100%) Preoperative HRT, stage,
matched cohort 2001 3–3 0.625 mg/d) plus grade, depth of myometiral
study medroxyprogesterone invasion, LVSI, tumour size,
acetate (oral, 2.5 mg/ resected LN number, mean
d) follow-up period
No HRT 54.3 I-46 1–38 53.2 1
(n = 52) II-6 2–11
3–3
Barakat et al. [20]/ US/ HRT 57* IA- 1–366 Oestrogen (oral, dose 618 Oral-618 36* 35.7* <5 Mo: 36*,  14 Age, stage, grade, histology,
Randomised 1997– (n = 618) 232 2–203 not otherwise (100%) (100%) 618 adjuvant therapy, type of
controlled trial 2003 IB- 3–45 specified) (100%) surgery
305
IC-48
IIA-
22
IIB-8
No HRT 57* IA- 1–367 35.7* 12
(n = 618) 238 2–198
IB- 3–48
302
IC-42
IIA-
17
IIB-
16
Non-randomised HRT 56.6à IA, 1,2– 143 Oral-198 44.0à 67.9à NA 45.4à 5
observational (n = 278) IB- 249
studies combined 156 (89.6%)
IC-12 3–29
II-18 (10.4%)
III-4
S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637 1633

Abbreviations: HRT, hormone replacement therapy; DFI, disease-free interval; LN, lymph node; LVSI, lymphovascular space invasion; NR, not reported; NA, not able to calculate; PET/CT, positron
but several reported matching control individuals
according to the DFI before HRT [20,21]. On average,
HRT users were more than two years younger than
non-users. The mean duration of HRT use was
38.0 months, with the mean duration of the follow-up
after the initiation of HRT at 38.3 months. The mean
duration of the follow-up was 45.7 months for HRT
users and 42.6 months for non-users.

3.3. Meta-analysis

Six studies included a total of 1975 patients with a


52

19

64

combined total of 83 recurrences (19 among 896 HRT


38.3à

users). Fig. 2A shows the ORs for the risk of EC recur-


rence for each study and all studies combined comparing
the use of HRT with no use of HRT. The pooled OR for
HRT use and the risk of EC recurrence was 0.534 (95%
NA

NA

NA

CI: 0.297–0.960). Heterogeneity was low across the stud-


ies (P = 0.081 and I2 = 49.0). Fig. 2B–D show the
51.8à

45.7à

42.6à

results of the subgroup analysis for EC survivors based


on the study design, type of HRT and stage. In the sen-
38.0à

sitivity analysis, the results based on the omission of one


study at a time and the calculation of the pooled OR for
the remaining studies show that no study had a signifi-
cant effect on the pooled OR (Supplemental Fig. 1,
Oral-816

online only).

3.4. Subgroup analysis by the type of study design

Fig. 2B shows the OR for each study and the pooled


761

OR for categories of NRS and RCT. Only one RCT was


conducted with a total of 1236 patients. Here the median
age was 57 years, the DFI was less than 20 weeks before
HRT initiation, the median duration of HRT use was
36 months and the median follow-up period after
HRT initiation was 36 months (Table 1). A total of 26
emission tomography/computed tomography; CI, confidence interval.

Assumed equal to the mean follow-up period after HRT initiation.

recurrences (14 among 618 HRT users) were observed


(84.6%)

(91.3%)

(88.5%)

(11.0%)

in this trial, and the OR for the RCT was 1.17 (95%
(8.3%)

3–119
(15.4)
3–71

3–74
1,2–

1,2–

1,2–

CI: 0.54–2.56). Five NRSs involved a total of 739


390

818

955

patients with the mean age of 59.7 years, the mean dura-
IC-47

IC-60

IC-89
II-22

II-48

II-55
III-4

III-4

III-4

tion of HRT use of 44 months, and the mean follow-up


385

784

925
IA,

IA,

IA,
IB-

IB-

IB-

period after HRT initiation of 45.4 months (Table 1). A


No HRT 61.5à

56.9à

No HRT 58.9à

combined total of 57 recurrences (five among 278 HRT


Assumed equal to the duration of HRT use.

users) were observed in these five studies. The pooled


(n = 1079)
(n = 461)

(n = 896)

OR for observational studies was 0.19 (95% CI: 0.08–


0.46; P = 0.943 and I2 = 0).
HRT

3.5. Subgroup analysis by the type of HRT

Fig. 2C shows the OR for each study and the


Weighted average.

pooled OR for categories of oestrogen only and com-


bined oestrogen plus progestin. Oestrogen only was
All combined

not associated with EC recurrence (OR: 0.35; 95%


Median.

CI: 0.06–2.10; P = 0.054 and I2 = 65.7). However, a


significant negative association (i.e. protective effect)
was observed in studies with combined oestrogen plus
*
 
à
§
1634 S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637

progestin (OR: 0.23; 95% CI: 0.08–0.66; P = 0.824 and administration of HRT to women started approxi-
I2 = 0). mately 15 months after initial treatment. Because most
EC recurrences occur within two years after the initial
3.6. Subgroup analysis by stage diagnosis, patient candidates for recurrence in NRSs
might have been eliminated in this period [27]. We also
Fig. 2D shows the OR for each study and the pooled found that the control groups in five NRSs have a
OR for categories of stage I, stages I and II and stages higher-than-expected recurrence rate (52 out of 461
I–III. HRT use in stage I EC survivors did not lead to patients = 11%) compared to the RCT (12 out of 618
a significant increase in the risk of EC recurrence (OR: patients = 2%). As seen in Table 1, baseline risk factors
0.12; 95% CI: 0.02–0.64; P = 0.99 and I2 = 0). HRT in of recurrence were different between the control
stages I and II was also not associated with EC recur- groups; the control groups of NRSs have more diseases
rence (OR: 0.87; 95% CI: 0.44–1.74; P = 0.295 and with grade 3 (15.4% versus 7.8%) and deep myometrial
I2 = 18.1). For stages I–III, only one study was reported invasion (10.2% versus 6.8%) compared to the RCT.
(OR: 0.16; 95% CI: 0.03–0.75) [19]. This feature from selection bias may lead to the high
rate of recurrence in the control group of the observa-
3.7. Publication bias tional studies.
Several studies added progesterone to HRT based on
The Begg-Mazumdar rank correlation test shows no findings that progesterone inhibits the stimulatory effect
evidence of publication bias (P = 0.85). The fail-safe N of oestrogen on normal and hyperplastic endometrium
test indicates that for the combined two-tailed P-value [18,19,21]. In the subgroup analysis by the type of
to be non-significant (P > 0.05), nine more studies with HRT, there was a protective effect of combined HRT
significant findings would be required. A funnel plot (OR: 0.23; 95% CI: 0.08–0.66) on EC recurrence, whereas
for publication bias was symmetric (Supplemental there was no such effect for oestrogen only (OR: 0.35;
Fig. 2, online only). 95% CI: 0.06–2.10). However, this should be interpreted
with caution because there was significant heterogeneity
4. Discussion (I2 = 65.7) across oestrogen-only group studies. In addi-
tion, in studies with combined HRT, HRT users were
According to published data, HRT use had no nega- approximately five years younger than non-users (56.7
tive effect on recurrence and overall survival after EC versus 61.9 years). Here it should be noted that only half
treatment. However, the individual sample size in most of the HRT users received progestin in all studies with
studies was not large enough to sufficiently explain the combined HRT [17–19] except for Ayhan et al. [21]. It
effect of HRT on EC recurrence, and no quantitative remains unclear whether progestin truly suppresses the
analysis has estimated the effect of HRT on EC recur- oestrogen-induced stimulation of incipient tumour cells.
rence. The results of the present meta-analysis indicate Nonetheless, adding progestin to the HRT regimen
that HRT use by women with a history of EC did not deserves a more informed discussion.
lead to a significant increase in the risk of EC recur- Most studies targeted patients with stage I or II EC
rence. This pattern was also observed in the subgroup for HRT use [16–18,20,21]. The analysis of EC survivors
analysis for the stage and type of HRT. with stage I or II EC reveals that HRT did not increase
The pooled OR for HRT use and the risk of EC the risk of recurrence in these patients. Although one
recurrence were calculated using a fixed effects model study included stage III EC patients [19], only four
because of low heterogeneity across studies. Nonethe- (5.3%) patients received HRT. Therefore, the evidence
less, the results of the subgroup analysis based on the provides no support for the argument that HRT can
study design show different patterns. More specifically, be safely given to patients with advanced EC. Residual
estimates from the NRS indicate that HRT prevented malignant cells after a suboptimal surgical effort for
EC recurrence, whereas those from the RCT do not. advanced EC may be restimulated by HRT, provoking
This result may be related to design features of those a recurrence.
NRSs that could have introduced selection bias, despite Although most studies used the standard oral dose of
the analytic technique used to remove their potential oestrogen (0.625 mg of conjugated oestrogen daily), the
effect. Given the caution against prescribing HRT to dosage was up to 1.25 mg of conjugated oestrogen daily
women with a history of EC, those women with a more in three studies [16–18]. However, because none of the
favourable prognosis might have been selected to studies described the exact number of patients who
receive HRT. Actually, in NRSs, HRT users were received this daily dose, it was not possible to conduct
approximately five years younger than non-users. In a subgroup analysis for the oestrogen dose. The admin-
addition, in NRSs, the average follow-up period for istration route was generally oral except for two studies
HRT users was longer than that for non-users (67.9 in which 34 (72.3%) [16] and 1 (1.6%) [18] received HRT
versus 51.8 months). This difference implies that the through only vagina. This suggests that, because topical
S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637 1635

oestrogen can significantly increase the level of systemic


estradiol [28], topical vaginal oestrogen was considered
to represent systemic use, as in oral administration
[18,19].
Among the included studies, the duration of HRT
ranged from 2 months [16,17] to 132 months [17]. How-
ever, the mean duration of HRT in all studies was longer
than 36 months except for Creasman et al. (26 months),
which was well beyond the interval for a majority of
recurrences. Therefore, no subgroup analysis was con-
ducted to determine whether this difference in the period
of hormone exposure would have differential effects on
EC recurrence.
The mean DFI before HRT administration varied
across studies. HRT started within 6 months after sur-
gery in only two studies [20,21], and the timing of
HRT initiation ranged from immediate postoperation
to a delay of up to 108 months [16–19]. Approximately
57–73% of the patients in three studies initiated HRT
within a year, whereas the remaining 27–43% had
HRT later [17–19]. These latter figures may represent
patients who were less likely to face EC recurrence for
the reason discussed earlier.
The RCT reported by Barakat et al. [20] overcomes
many of the shortcomings of the NRS and provides
the best available data on the effect of HRT in EC sur-
vivors. However, the study was closed prematurely after
the publication of the findings of the Women’s Health
Initiative [29]. In addition, patients primarily with early,
good prognosis tumours were enroled (60% < 60 years
old; 38% and 50% with stage IA and IB EC, respec-
tively). On the other hand, 8% had stage IC EC, and
only 7% had grade 3 EC. The insufficient enrolment of
high-risk EC patients likely led to an insufficient number
of recurrences and thus suboptimal power for the clini-
cal trial.
Fig. 2. (A) The odds ratio for the risk of EC recurrence for each study
According to preclinical data, in vitro treatment of
and all studies combined comparing hormone replacement therapy
Ishikawa endometrial cancer cells with oestrogen upreg- (HRT) use with no HRT use in a meta-analysis based on the fixed
ulates oestrogen receptor expression and augments the effects model. Heterogeneity was low across studies (P = 0.081,
growth of these cells [30,31]. However, no study has ver- I2 = 49.0). HRT use and the risk of EC recurrence in the subgroup
ified in vivo growth of residual microscopic endometrial meta-analysis by (B) study design, (C) the type of HRT and (D) the
stage of HRT. The size of each square is proportional to sample size
cancer in patients. Several laboratory studies have sug-
for each study, and the horizontal line through the square indicates the
gested that oestrogen exposure is associated with the 95% confidence interval for that study. For the pooled analysis, the
increased mitosis of normal endometrial cells, thereby diamond indicates the pooled value, and the right and left ends of the
placing them in a particular molecular configuration diamond indicate the 95% confidence interval for the analysis.
susceptible to DNA damage [32,33]. This suggests that
oestrogen can act as a tumour promoter for already and EC recurrence has yet to be clearly illustrated,
genetically altered cells. Therefore, oestrogen may not and therefore further molecular biology research is
have any further deleterious effect once the neoplastic necessary.
transformation takes place [19]. In other words, when The results of this study should be interpreted with
the source of endometrial tissue is removed through hys- caution because of several limitations. First, a majority
terectomy as part of primary cancer treatment, recur- of the studies were retrospective and a meta-analysis can-
rence then becomes a function of occult residual not address problems related to confounding factors
tumour cells, not a result of some de novo hormonal inherent in studies considered. All of the included studies
stimulation or neoplastic transformation [19]. The were awarded three stars on the NOS selection category.
mechanism underlying the relationship between HRT However, only three studies tried to control for selection
1636 S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637

bias by indication or other confounding factors. The abil- Acknowledgement


ity of this study to control for potential confounders was
limited by a small number of studies considered and a This work was supported by Konkuk University.
lack of comprehensive reporting by those studies.
Although the included studies adjusted for potential con- Appendix A. Supplementary data
founding factors, residual or unknown confounding fac-
tors could not be excluded as a potential explanation for Supplementary data associated with this article can
the observed findings. Second, the overall use of HRT be found, in the online version, at http://dx.doi.org/
varied across studies: 21.3% (47/221) in Ref. [16], 30.8% 10.1016/j.ejca.2014.03.006.
(44/143) in Ref. [17], 50.5% (62/123) in Ref. [18] and
52.2% (130/249) in Ref. [19]. Third, it was not possible References
to consider a further subgroup analysis for patient age
(less or greater than 50), dosage of HRT and tumour [1] Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA
grade because of a lack of data. In addition, it was not Cancer J Clin 2012;62:10–29.
[2] Lim MC, Moon E-K, Shin A, et al. Incidence of cervical,
possible to address the question of whether HRT is con- endometrial, and ovarian cancer in Korea, 1999–2010. J Gynecol
traindicated for patients with advanced-stage EC because Oncol 2013;24:298–302.
only one study included stage III EC patients [19]. [3] Darai E, Gompel A, Deval B, et al. Hormone replacement
Finally, although no major publication bias was found, therapy after endometrial or ovarian cancer. Gynecol Obstet
Fertil 2000;28:198–204.
its presence cannot be completely excluded because small
[4] Michaelson-Cohen R, Beller U. Managing menopausal symptoms
studies showing a negative effect of HRT on recurrence after gynecological cancer. Curr Opin Oncol 2009;21:407–11.
are not likely to be published (the so-called ‘small-study [5] Hancke K, Foeldi M, Zahradnik HP, Gitsch G, Gilbert L,
effect’) [34]. This potential bias can underestimate the Denschlag D. Estrogen replacement therapy after endometrial
true risk of recurrence in HRT users. The Egger regres- cancer: a survey of physicians’ prescribing practice. Climacteric
sion asymmetry test performs better than the rank corre- 2010;13:271–7.
[6] Stroup DF, Berlin JA, Morton SC, et al. Meta-analysis of
lation test in evaluating the small-study effect, but it lacks observational studies in epidemiology: a proposal for reporting.
power unless the number of included trials is greater than Meta-analysis Of Observational Studies in Epidemiology
or equal to 10 [34]. In this regard, the results of the pres- (MOOSE) group. JAMA 2000;283:2008–12.
ent meta-analysis should be interpreted accordingly. [7] Moher D, Liberati A, Tetzlaff J, Altman DG, Group P. Preferred
In sum, the results suggest that HRT use does not reporting items for systematic reviews and meta-analyses: the
PRISMA statement. PLoS Med 2009;6:e1000097.
increase the risk of recurrence in women with stage I [8] Wells G, Shea B, O’connell D, et al. The Newcastle-Ottawa Scale
and/or II EC, although they are based mainly on retro- (NOS) for assessing the quality of nonrandomised studies in meta-
spective, case, or cohort-controlled studies subject to a analyses. In: 3rd Symposium on systematic reviews: beyond the
variety of biases. The selection of healthier and younger basics; 2000. p. 3–5.
women to begin HRT and publication bias may explain [9] Aziz O, Constantinides V, Tekkis PP, et al. Laparoscopic versus
open surgery for rectal cancer: a meta-analysis. Ann Surg Oncol
the protective effect of HRT on recurrence in EC survi- 2006;13:413–24.
vors. Although the results do not completely exclude [10] Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-
the possibility of HRT increasing the risk of recurrence, analysis. Stat Med 2002;21:1539–58.
they suggest that the magnitude of such a risk is not large. [11] Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring
The positive effect of HRT on the quality of life seems to inconsistency in meta-analyses. BMJ 2003;327:557–60.
[12] Orwin RG. A fail-safe N for effect size in meta-analysis. J Educ.
outweigh the unfounded suspicion about an increased Stat. 1983;8:157.
risk of recurrence. In this regard, additional well-designed [13] Begg CB, Mazumdar M. Operating characteristics of a rank
RCTs considering all potential confounding factors, correlation test for publication bias. Biometrics
including cancer characteristics and treatment, different 1994;50:1088–101.
[14] Sterne JA, Egger M. Funnel plots for detecting bias in meta-
types of HRT, the DFI before HRT initiation and the
analysis: guidelines on choice of axis. J Clin Epidemiol
duration of HRT use, are needed to verify the results of 2001;54:1046–55.
this meta-analysis. In the meantime, EC survivors inter- [15] Sterne JA, Egger M, Smith GD. Systematic reviews in health care:
ested in HRT should be informed of available data and investigating and dealing with publication and other biases in
not be exposed only to unsubstantiated bias. meta-analysis. BMJ 2001;323:101–5.
[16] Creasman WT, Henderson D, Hinshaw W, Clarke-Pearson DL.
Estrogen replacement therapy in the patient treated for endome-
Source of funding trial cancer. Obstet Gynecol 1986;67:326–30.
[17] Lee RB, Burke TW, Park RC. Estrogen replacement therapy
This study received no financial support. following treatment for stage I endometrial carcinoma. Gynecol
Oncol 1990;36:189–91.
Conflict of interest statement [18] Chapman JA, DiSaia PJ, Osann K, Roth PD, Gillotte DL,
Berman ML. Estrogen replacement in surgical stage I and II
endometrial cancer survivors. Am J Obstet Gynecol
None declared. 1996;175:1195–200.
S.-H. Shim et al. / European Journal of Cancer 50 (2014) 1628–1637 1637

[19] Suriano KA, McHale M, McLaren CE, Li KT, Re A, DiSaia PJ. [27] Levgur M. Estrogen and combined hormone therapy for women
Estrogen replacement therapy in endometrial cancer patients: a after genital malignancies: a review. J Reprod Med
matched control study. Obstet Gynecol 2001;97:555–60. 2004;49:837–48.
[20] Barakat RR, Bundy BN, Spirtos NM, Bell J, Mannel RS, [28] Kendall A, Dowsett M, Folkerd E, Smith I. Caution: vaginal
Gynecologic Oncology Group S. Randomized double-blind trial estradiol appears to be contraindicated in postmenopausal
of estrogen replacement therapy versus placebo in stage I or II women on adjuvant aromatase inhibitors. Ann Oncol
endometrial cancer: a Gynecologic Oncology Group Study. J Clin 2006;17:584–7.
Oncol 2006;24:587–92. [29] Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits
[21] Ayhan A, Taskiran C, Simsek S, Sever A. Does immediate of estrogen plus progestin in healthy postmenopausal women:
hormone replacement therapy affect the oncologic outcome in principal results from the Women’s Health Initiative randomized
endometrial cancer survivors? Int J Gynecol Cancer controlled trial. JAMA 2002;288:321–33.
2006;16:805–8. [30] Holinka CF, Hata H, Kuramoto H, Gurpide E. Responses to
[22] Lauritzen C. Östrogensubstitution in der Postmenopause vor und estradiol in a human endometrial adenocarcinoma cell line
nach behandeltem Genital- und Mammakarzinom: Menopause: (Ishikawa). J Steroid Biochem 1986;24:85–9.
Hormonsubstitution heute. Basel: Aesopus Verlag; 1993, p. 76– [31] Farnell YZ, Ing NH. The effects of estradiol and selective estrogen
80. receptor modulators on gene expression and messenger RNA
[23] Bryant GW. Administration of estrogens to patients with a stability in immortalized sheep endometrial stromal cells and
previous diagnosis of endometrial adenocarcinoma. South Med J human endometrial adenocarcinoma cells. J Steroid Biochem Mol
1990;83:725–6. Biol 2003;84:453–61.
[24] Baker DP. Estrogen-replacement therapy in patients with previ- [32] Samsioe G. The endometrium: effects of estrogen and estrogen-
ous endometrial carcinoma. Compr Ther 1990;16:28–35. progestogen replacement therapy. Int J Fertil Menopausal Stud
[25] Gitsch G, Hanzal E, Jensen D, Hacker NF. Endometrial cancer in 1994;39(Suppl. 2):84–92.
premenopausal women 45 years and younger. Obstet Gynecol [33] Li SF, Shiozawa T, Nakayama K, Nikaido T, Fujii S. Stepwise
1995;85:504–8. abnormality of sex steroid hormone receptors, tumor suppressor
[26] Maxwell GL, Tian C, Risinger JI, Hamilton CA, Barakat RR, gene products (p53 and Rb), and cyclin E in uterine endometrioid
Gynecologic Oncology Group S. Racial disparities in recurrence carcinoma. Cancer 1996;77:321–9.
among patients with early-stage endometrial cancer: is recurrence [34] Sterne JA, Gavaghan D, Egger M. Publication and related bias in
increased in black patients who receive estrogen replacement meta-analysis: power of statistical tests and prevalence in the
therapy? Cancer 2008;113:1431–7. literature. J Clin Epidemiol 2000;53:1119–29.
The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Hematuria in Adults
Julie R. Ingelfinger, M.D.​​

V
isible (macroscopic) hematuria, documented since ancient This article was updated on July 13, 2021,
times,1,2 is striking, particularly when there is no prior event, such as trau- at NEJM.org.

ma, dysuria due to cystitis, or flank pain with passage of a kidney stone, to N Engl J Med 2021;385:153-63.
provide a clear explanation. In contrast, nonvisible (microscopic) hematuria, or DOI: 10.1056/NEJMra1604481
Copyright © 2021 Massachusetts Medical Society.
microhematuria, may go undetected for years.2 Many patients are found to have
microhematuria when a urinalysis is performed for other reasons.
The reported prevalence of microhematuria varies greatly, from a small percent-
age of patients at screening to more than 40% in some urology clinics, probably
owing to referral bias regarding patients and their signs and symptoms.3-5 The
American Urological Association (AUA) guidelines note a prevalence ranging from
2.1 to 31.4%.5 Thus, a discussion of epidemiology per se is less important than a
consideration of individual patient circumstances and geographic region. In certain
parts of the world — for example, in northern Africa, where Schistosoma haematobium
is endemic — microhematuria is common, reflecting prevalent bladder infestation.
In addition, the causes of hematuria differ between men and women, and the
evaluation, accordingly, should reflect that difference.6-8 Furthermore, the implica-
tions of hematuria vary greatly, depending on the underlying cause. Given the
association of both visible hematuria and microhematuria with bladder and kidney
cancer, the focus of evaluation has long been to rule out cancer. Cancer is more
likely to be identified in men than in women when they are evaluated for micro-
hematuria. Worldwide, estimates of the incidence of kidney cancer are 6.0 cases
per 100,000 person-years for men and 3.1 cases per 100,000 person-years for
women, and estimates of the incidence of bladder cancer are 9.0 cases per 100,000
person-years for men and 2.2 cases per 100,000 person-years for women, accord-
ing to GLOBOCAN, a registry of data on the global incidence of cancer,9 a result
also found in a 2020 meta-analysis.10 However, evaluation in women is often de-
layed, which may contribute to worse outcomes of bladder cancer among women.
Beyond cancer, hematuria is associated with protean symptoms and causes.
This review focuses on major causes, the evaluation, and the implications of he-
maturia. Since therapy depends on the cause, treatment is not addressed here.

De tec t ion of Hem at ur i a


Urine hue normally ranges from nearly colorless, when dilute, to dark amber,
when concentrated. Red, pink, “rusty,” or brown urine may suggest hematuria but
may be due to substances besides blood. Available dipstick tests detect red cells,
as well as hemoglobin and myoglobin, necessitating microscopic examination of
the urine if a dipstick test is heme-positive, followed by other tests to confirm
hematuria and, potentially, to determine its cause.11,12
Commonly used dipstick tests incorporate a benzidine compound reduced
with a buffered organic peroxide (3,3′,5,5′-tetramethylbenzidine and diisopropyl-
benzene dihydroperoxide). When the test strip is dipped in urine that includes an
oxidizing substrate, a color reaction ensues; for example, the pseudoperoxidase

n engl j med 385;2  nejm.org  July 8, 2021 153


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 1. Selected Nonheme Causes of Pigmenturia, as Compared with Red Cells or Free Hemoglobin.*

Variable Red Cells Hemoglobin Myoglobin Porphyria Bile Pigments Alkaptonuria


Urine color Red to rusty Pink to red Rusty Turns black, Brown Turns dark in
brown, or red in sunlight
sunlight
Heme test Positive Positive Positive Negative Negative Negative
Usual microscopic Red cells, No cells No red cells; Normal Normal Normal
findings casts casts may be
present
Plasma Normal Pink Normal Normal Icteric Normal

* Shown are selected endogenous causes of pigmenturia. A heme test for exogenous causes, including beets, rhubarb,
azo dye, sulfonamides, and phenolphthalein, is negative.

activity of hemoglobin will oxidize the benzi- essential.15 True microhematuria is most often
dine compound, turning the dipstick blue. Free defined as more than 2 or 3 red cells per high-
hemoglobin and myoglobin also react with the power field, and this finding should be con-
test strip, as would be expected because of their firmed on two or three separate urinalyses.16,17
molecular structure.13 Some experts suggest that if even a single urinaly-
Certain substances may lead to false positive sis is positive, a patient should have follow-up
dipstick reactions: sodium hypochlorite, peroxi- urinalyses for at least a year, so as to avoid miss-
dases from vegetable or bacterial sources, and se- ing an intermittent source of microhematuria
men. In contrast, very high ascorbic acid levels in that could signify a clinically important problem.
urine may produce false negative results. The list of Careful examination of the urinary sediment
substances that can be confused with hematuria is central to differentiating glomerular from
is lengthy (see the partial listing in Table 1). The other forms of hematuria.15 Although micro-
absence of red cells on microscopic examination of graphs of red cells crossing the glomerular base-
a “heme-positive” urine specimen suggests ei- ment membranes are as rare as hen’s teeth, red
ther that the red cells have all lysed or that iso- cells that have traversed the glomerular base-
lated hemoglobinuria or myoglobinuria may be ment membrane into the glomerular filtrate are
present. Hemoglobinuria is dipstick-positive, but much the worse for wear and appear with blebs
if it is the sole cause of the positive test, red cells and other irregularities.18 In contrast, lower uri-
will not be present in the urinary sediment. In nary tract hematuria is characterized by normal
some instances, both hematuria and hemoglobin- red cells (Fig. 1). If dysmorphic red cells are
uria are present and red cells will be identified. found, the diagnostic evaluation should first
Distinguishing blood from myoglobin in the focus on the possibility of a glomerulopathy (see
urine (which may signify rhabdomyolysis and the pragmatic algorithm in Fig. 2), particularly
may or may not be suspected, given a patient’s if casts, especially red-cell casts, are present.
history) is clinically important.14 Frequently, cen- Isolated hematuria is less likely to be associ-
trifugation is helpful, since red cells will sedi- ated with a glomerulopathy than is hematuria
ment, leaving a clear supernatant, whereas myo- with albuminuria or decreased kidney function,
globin will not. However, free hemoglobin also though IgA nephropathy and familial nephropa-
will not precipitate in a routine laboratory cen- thies may not always be characterized by albu-
trifuge. In all, the absence of red cells in the minuria.15 Bacteria in the unspun urine and in
sediment of a “heme-positive” specimen suggests the sediment suggests urinary tract infection.
that either isolated hemoglobinuria or myoglo- Crystals may suggest nephrolithiasis.
binuria may be present. Myoglobinuria can then If examination of the urinary sediment shows
be identified biochemically through ammonium normal-appearing red cells, then imaging and
sulfate precipitation, as well as through electro- urologic referral should be considered. Most
phoretic and immunologic tests. urologists will focus on imaging results and cys-
Thus, after a positive dipstick test, micro- toscopy. In patients with risk factors for cancer,
scopic examination of the urinary sediment is many urologists and guidelines would suggest

154 n engl j med 385;2  nejm.org  July 8, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Hematuria in Adults

A Nondysmorphic Red Cells B Dysmorphic Red Cells

Figure 1. Nondysmorphic and Dysmorphic Red Cells in Freshly Voided Urine.


Panel A shows nondysmorphic red cells, and Panel B shows dysmorphic red cells, which have characteristic blebs.
Micrographs are courtesy of the estate of the late Michael Linshaw, M.D.

either urethrocystoscopy and computed tomo- renal tubular cells subsequently engage in erythro-
graphic (CT) urography19 or ultrasonography and phagocytosis, which has been linked to further
cystoscopy.5 nephrotoxicity induced by iron and hemoglobin.26
Glomerular hematuria in the absence of pro-
teinuria or kidney dysfunction has largely been C onfir med V isibl e Hem at ur i a
considered to be an innocuous finding or per-
haps a marker of changes in the glomerular Visible hematuria often has an obvious explana-
filtration apparatus or of inflammation, yet some tion, which is congruent with the patient’s his-
data suggest that isolated glomerular hematuria tory, signs, and symptoms (e.g., the passage of
may not always be innocuous.20 Over time, even a kidney stone, acute hemorrhagic cystitis, or a
in conditions considered to have a good progno- sickle-cell crisis). However, many other condi-
sis, microhematuria has been associated with tions can cause visible hematuria (Fig. 3). Thus,
progressive kidney failure, and macrohematuria the evaluation of gross hematuria should be
has been associated with acute tubular dysfunc- focused on the basis of the patient’s symptoms
tion and acute kidney injury.20 For instance, epi- and concomitant clinical and laboratory find-
sodes of gross hematuria in persons with IgA ings.27 In adults over the age of 40 years (some
nephropathy may be accompanied by diminution sources suggest a cutoff age of 35 years), an
of the glomerular filtration rate or frank acute unexplained episode of visible hematuria may
kidney injury, but the patient generally recov- signify bladder or upper urinary tract cancer,
ers.21 Vivante et al.22 reported that microhematu- and most experts suggest urologic referral
ria among army recruits was associated with a along with imaging and cystoscopy to rule out
risk of end-stage kidney disease many years later. cancer.28-31
Furthermore, kidney donors who have had micro-
hematuria have been reported to have an increased Medical History and Physical Examination
risk of kidney dysfunction after donation.23 A thorough history is critical in focusing the
The mechanism of kidney injury may be due diagnostic evaluation of a patient with gross
to the effects of tubular inflammation and oxi- hematuria.27-31 Has the patient had trauma, and
dative stress induced by breakdown products of might there be a renal contusion, vascular com-
red cells.24,25 Whether glomerular hematuria it- promise, or infarction? A history suggestive of
self induces progression or is a surrogate for urinary tract infection or renal colic calls for an
inflammation is unclear. Moreno et al.24,25 postu- initial evaluation to confirm or rule out an infec-
late that red-cell casts induce damage to tubular tion or a kidney stone. If the patient has just
cells through obstruction, that hemoglobin and undergone surgery, is there a complication, even
heme are directly toxic to the tubules, and that if the surgical procedure did not directly involve

n engl j med 385;2 nejm.org July 8, 2021 155


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Hematuria

If negative, pseudohematuria
Dipstick test administered
diagnosis

If positive, proceed with urinalysis and


basic metabolic panel

Dysmorphic red cells, casts Nondysmorphic red cells

Glomerular bleeding Nonglomerular bleeding

Evaluate blood and urine for Urine culture and sensitivity


UTI symptoms Yes
possible glomerulopathies test; treatment for UTI
Consider kidney biopsy
No

Nephrolithiasis symptoms Yes CT and stone evaluation

No

Diagnosis
Evaluate cancer risk No
confirmed

Yes

Moderate-to-high risk Low risk

Cystoscopy and CT (if high risk)


Findings still positive Repeat urinalysis in 4–6 wk Treatment
Ultrasonography and cystoscopy
(if moderate risk)

No diagnosis; consider evaluation Resolved; perform follow-up


Diagnosis and treatment
for glomerular bleeding urinalysis for 1–2 yr

Figure 2. Algorithm Incorporating Risk Assessment in the Evaluation of Hematuria.


The algorithm is based on the current American Urological Association guidelines. UTI denotes urinary tract infection.

the kidneys? Is the patient receiving anticoagu- workup.27-31 Are vital signs stable, and is there
lants? Has the patient engaged in strenuous ac- any sign of vascular instability that would sug-
tivity, such as a long-distance run, which may gest bleeding or some other acute, potentially
occasionally be associated with visible hematu- life-threatening problem? Costovertebral angle
ria? Has the patient’s voiding pattern changed, or suprapubic tenderness may suggest a kidney
which could be suggestive of prostatic hypertro- stone or a urinary tract infection. In male pa-
phy or obstruction? Does the patient have flank tients, rectal examination to palpate the prostate
pain or suprapubic pain? may help assess the possibility of prostatitis or
The physical examination further focuses the prostate cancer. In female patients, it is impor-

156 n engl j med 385;2  nejm.org  July 8, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Hematuria in Adults

Glomerular Hematuria Nonglomerular Hematuria


IgA nephropathy Polycystic kidney disease
Alport’s syndrome Sickle-cell disease and papillary necrosis
Thin basement membrane disease Nephrolithiasis
C3 nephropathy Tumors
Postinfectious glomerulonephritis Leukemia
Trauma
Infection
Strenuous exercise
Kidney

Ureteral Hematuria Ureter


Stones Vesicular Hematuria
Infection
UTI
Trauma
Stones
Tumor
Infections
Bladder Irritation
Tumor
Urethra Foreign body

Sexual Dimorphism Urethral Hematuria


Prostatic bleeding Trauma
Vaginal bleeding Infection
Endometriosis of the urinary tract

Figure 3. Hematuria According to Location.


Categories of hematuria are shown according to the location in the urinary tract.

tant to be sure that the bleeding is from the a mean age of 65.7 years) with visible or micro-
urinary tract, not the reproductive tract. Signs of scopic hematuria, 10% of the cohort had can-
systemic disease should also be sought. cers, mainly bladder cancer (in 8.0% of all pa-
tients evaluated); cancer was more common
Imaging among those with visible hematuria.33 Renal
Imaging is generally considered necessary in parenchymal cancer was seen in 1.0% of the
adults with gross hematuria, given the possibil- patients, upper tract transitional-cell cancer in
ity of cancer, unless there is an obvious cause 0.7%, and prostate cancer in 0.3%. In this study,
(e.g., hemorrhagic cystitis). For many patients, cancer was most likely to be diagnosed in the
ultrasonography plus cystoscopy suffices. CT male patients, especially older men and men
urography is recommended by some sources with a history of smoking. The results of most
but is more costly than ultrasonography and trials, taken together, have suggested that any
usually includes the administration of contrast patient with visible blood in the urine should
material.27-31 The use of magnetic resonance im- undergo urologic investigation.5,33
aging is not generally recommended. It is said
that dysuria is present in approximately 80% of C onfir med Microhem at ur i a
patients with bladder cancer and that dysuria
doubles the likelihood of finding a bladder The most common causes of microhematuria
cancer.32 are nonmalignant5: glomerulopathies (e.g., IgA
In DETECT 1, a prospective, observational nephropathy or thin glomerular basement mem-
study involving 3556 adults (59% were men, with brane disease) and inflammatory conditions of

n engl j med 385;2 nejm.org July 8, 2021 157


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

the urethra, prostate, and bladder, as well as agents (e.g., cyclophosphamide); and prolonged
renal calculi and benign prostatic hypertrophy. exposure to foreign bodies (e.g., catheters).
The causes of microhematuria are many and In the above-mentioned DETECT 1 trial,33
disparate, as summarized in Figure 3. 10% of patients had malignant tumors, the ma-
Most sources state that confirmed micro­ jority of which were bladder cancers. In another
hematuria requires evaluation, since overlooking study, 234 male patients who had had a negative
a bladder cancer or kidney cancer would be po- evaluation for microhematuria were followed for
tentially fatal, but that viewpoint has been chal- 14 years; urinary tract cancer developed in 2 of
lenged in recent years.34,35 Urinary tract cancer is these patients (0.9%).38
rare in women, and recent data suggest that the Despite the low frequency of urinary tract
likelihood of cancer is less than 5% in a woman cancer, expert opinion favors evaluation.5,39-42
under 50 years of age who has never smoked and Nielsen and Qaseem, for example, recommend
who has asymptomatic microhematuria with that if a patient has persistent microhematuria,
fewer than 25 red cells per high-power field.8 evaluation should proceed, even if that patient is
Even for men, some guidelines suggest individ­ receiving antiplatelet or anticoagulant medica-
ualized evaluation. Such recommendations note tion.39 In contrast, the 2012 AUA guidelines,
that evaluation of microhematuria is time-con- based on a review of 191 evidence-based reports
suming and costly and may cause more harm and on expert opinion for areas and concepts
than good. In one study, up to 57% of patients lacking such data, recommended evaluation for
referred to a urology service for microhematuria all patients 35 years of age or older in whom
turned out to have pseudohematuria.12 Given all microhematuria does not have an obvious, identi-
these considerations, the decision to evaluate a fied benign cause, according to the history, physi-
patient may best be based on a mutual decision cal examination, and urinalysis.42 That workup,
arrived at through discussion between patient according to the AUA, should include cystoscopy
and clinician. and upper urinary tract imaging with CT.
There is debate about whether evaluation of
microhematuria should include invasive testing Medical History
such as cystoscopy, even though the annual inci- As with visible hematuria, it is crucial to obtain
dence of urinary tract cancers is substantial. For a good history in order to focus the evaluation of
example, in 2009, an estimated 70,980 bladder microhematuria.5,15 Is the patient symptomatic,
cancers and 57,760 renal cancers were diagnosed and if so, what are those symptoms?
in the United States. In 2020, the estimated The evaluation of microhematuria is focused
numbers of new bladder and renal cancer cases on ruling out diagnoses that are very important
were 81,400 and 73,750, respectively, according not to miss — mainly, urinary tract cancers.
to the American Cancer Society.36 Microhematuria is detected by dipstick testing
Although, as noted above, most patients with or examination of the urinary sediment. It is
urinary tract cancer present with hematuria, important to ascertain that the patient does not
asymptomatic microhematuria has a consistently have a urinary tract infection, is not menstruat-
poor performance as a trigger for diagnostic ing, and has not just exercised, engaged in sex-
investigations. For example, Jung et al.37 exam- ual activity, or had exposure to instrumentation
ined the medical records of 156,692 patients (e.g., through cystoscopy).
with microhematuria; the incidence of urinary A careful examination of the urinary sediment
tract cancer was 0.7% over a period of 3 years.37 is key. It is best to obtain a clean-catch mid-
An age of more than 40 years, the presence of stream urine sample — ideally, the first void of
more than 25 red cells per high-power field, and the day — and examine it within 1 to 2 hours.
male sex increase the risk of cancer. Additional The first void of the day has higher osmolality
risk factors include occupational exposure to and lower pH (both of which are helpful in pre-
carcinogens, particularly aromatic amines and serving the morphologic features of formed
hydrocarbons (involved in chimney maintenance sediment elements) than a sample obtained later
and dye work); analgesic abuse; pelvic irradia- in the day.5 There are standard ways to perform
tion; chemotherapy courses involving alkylating a urinalysis, generally spinning the sample at

158 n engl j med 385;2  nejm.org  July 8, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Hematuria in Adults

4000 rpm for 5 minutes after obtaining a dip- In a study by Mishriki et al.,49 in which 2278
stick reading.11,15 The normal number of red patients underwent urinary cytologic analysis,
cells in the urinary sediment is generally agreed only 2 were determined to have cancer on the
to be fewer than 2 or 3 per high-power field.5,11,15 basis of the cytologic findings. Cytologic analy-
The presence of red cells in one urinalysis sis in a study by Hofland et al.50 showed cancer
should be confirmed in one or two more uri- in 4 of 1000 urine samples, and in 2 of those
nalyses. If microhematuria is not confirmed in a 4 samples, cancer was detected only on the basis
second or third urinalysis, many sources suggest of cytologic findings. Current practice is to
repeated testing over a period of a year or so. If limit the use of cytologic analysis to cases of
microhematuria is confirmed, then it is impor- gross hematuria or symptomatic hematuria.5,52
tant next to consider whether the findings sug-
gest an upper or a lower urinary tract origin. Imaging
The morphologic characteristics of red cells,43-47 Adults with established microhematuria should
as well as a review of dipstick results, the undergo imaging, and as with visible hematuria,
formed elements in the urinary sediment, and the most cost-effective combination of studies
any other urinary abnormalities detected, may has been found to be ultrasonography plus cys-
suggest whether upper or lower urinary tract toscopy.5 Using decision analysis, Halpern et al.34
bleeding is more likely, with dysmorphic red looked at simulated cancer detection rates in
cells in the sediment suggesting an upper uri- persons with asymptomatic microhematuria and
nary tract (glomerular) source of bleeding.5 Ac- associated costs in terms of the incremental
cording to some sources, a finding that more costs of cancer detection. They examined four
than 25% of urinary red cells are dysmorphic imaging approaches: CT urography alone, cys-
points to glomerular disease.47 Urinary red-cell toscopy alone, CT urography plus cystoscopy,
mean corpuscular volume has also been used to and ultrasonography plus cystoscopy. Ultraso-
help differentiate between upper and lower uri- nography plus cystoscopy detected the most
nary tract disease.48 The sensitivity of both dys- cancers per incremental cost of cancer detection,
morphic and small red-cell findings has been and exchanging CT urography for ultrasonogra-
challenged. The presence of proteinuria should phy would have detected only one additional
impel the clinician to launch an investigation for cancer.
a major nephropathy.15 In sum, isolated hema-
turia with dysmorphic red cells or red cells with Cystoscopy
a decreased mean corpuscular volume, with or For patients who have gross hematuria without
without casts, should prompt an evaluation for an obvious cause, cystoscopy is generally recom-
glomerulopathy as well. The most commonly mended in order to rule out cancer, most often
associated glomerulopathies are IgA nephropa- bladder cancer. The 2012 AUA guidelines42 recom-
thy and Alport’s syndrome (familial nephritis). mended cystoscopy for all patients with micro-
But the list of potential glomerulopathies is hematuria who are older than 35 years of age,
long, and an evaluation of kidney function, but the 2020 AUA guidelines5 added risk levels
complement status, and autoimmune markers is and recommended immediate cystoscopy only
indicated, depending on the individual patient. for patients at increased risk.
Even if the blood in the urine appears to be due
to lower urinary tract disease, kidney function Molecular Biomarkers
and the estimated glomerular filtration rate The possibility that molecular markers might
should be checked. The specific studies will de- focus the workup of hematuria is attractive.53
pend on the possibilities that seem likely (Fig. 2). Biomarkers for various urinary tract cancers,54,55
for glomerular disease,56,57 and for genetic dis-
Cytologic Analysis eases58 are of great interest. Bladder cancer is
Urinary cytologic analysis49-52 has long been rec- heterogeneous, and if there were prospectively
ommended as a possible adjunct to cystoscopy validated markers, they might serve as important
and can serve as a way to find evidence of small adjuncts in clinical decision making.53 Whereas
carcinomas that are overlooked on cystoscopy. 90 to 95% of bladder cancers are urothelial

n engl j med 385;2  nejm.org  July 8, 2021 159


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

(transitional-cell) carcinomas, only 3 to 7% are the absence of a specific diagnosis, monitoring


squamous-cell carcinomas (usually in the renal at intervals is considered to be important — ini-
pelvis and ureters), and less than 3% are adeno- tially after an interval of a few months and then
carcinomas (most often in the trigonal region). once or twice yearly.
The relevant markers for distinct types of can-
cer appear to differ. Sathianathen et al.54 car- Guidance for Screening and Evaluation
ried out a meta-analysis in which 17 studies Given the wide differential diagnosis and the
met the specified criteria. The authors found disparate reports about the import of micro­
substantial heterogeneity among the studies. hematuria, current guidance varies. The Ameri-
Evaluating several biomarkers in combination can College of Physicians has stated (in 2016)39
(NMP22, UroVysion, and uCyt+) or alone (blad- that asymptomatic adults as a group should not
der tumor–associated antigen [BTA], NMP22, be screened by means of urinalysis for cancer
uCyt+, Cxbladder, and AssureMDx), they calcu- detection and that for those who are nonetheless
lated that the biomarkers had high sensitivity screened and found to have a positive dipstick
but insufficient specificity to eliminate the need test, the presence of blood should be confirmed
for cystoscopy. Wilson et al.55 noted that BTA by microscopic urinalysis. The U.S. Preventive
and NMP22, which are approved by the Food Services Task Force does not recommend screen-
and Drug Administration for use together with ing for microhematuria, citing inadequate evi-
cystoscopy for the diagnosis of bladder cancer, dence of benefit versus harm.61 Others have also
do not work well if used alone, since hematuria considered the issue of harms and benefits.62,63
in the absence of cancer, as well as infections
and stones, is associated with false positive test
results. To date, despite the interest in bio- Table 2. Risk Stratification for Bladder Cancer.
markers, the types of prospective studies need- Low risk (all criteria must be met)
ed to render biomarkers clinically useful are
<40 Yr of age for men, <50 yr of age for women
lacking.
Never smoked or <10 pack-yr of smoking
Nucleic acids detected in plasma, including
DNA (genomic, as well as mitochondrial and 3–10 Red cells per high-power field on one urinalysis
viral), RNA, and microRNA, have been recog- No risk factors for urothelial cancer
nized for some time as potential biomarkers. Intermediate risk (one criterion raises the risk to inter-
Circulating tumor DNAs (ctDNAs) are used as mediate)
biomarkers in multiple areas of oncology. The 40–59 Yr of age for men and women
detection of ctDNA in muscle-invasive bladder 10–30 Pack-yr of smoking
cancer is being piloted for use in diagnosis and 11–25 Red cells per high-power field on repeat urinalysis
for monitoring during treatment and follow-up
Additional risk factors for urothelial cancer
care.59 Cell-free DNA has been used similarly in
High risk (one criterion raises the risk to high)
renal-cell carcinoma.60
≥60 Yr of age for men and women
Microhematuria with No Identified Cause >30 Pack-yr of smoking
The specific cause of hematuria may be elusive. >25 Red cells per high-power field on single urinalysis
In some series, more than half of the patients History of gross hematuria
have microhematuria with no definable cause. Additional risk factors for urothelial cancer
Bolenz et al.32 suggest a “risk-adapted investiga-
Irritative lower urinary tract symptoms
tion,” connoting a personalized and risk–benefit
approach to evaluation that is based on the view Previous pelvic radiation therapy
that every patient with hematuria should be Previous chemotherapy with cyclophosphamide
or ifosfamide
evaluated, but not every possible test should be
used. Furthermore, if no cause is identified, at a Family history of urothelial cancer or Lynch syndrome
certain point, it makes sense to live with uncer- Occupational exposure to benzene or aromatic amines
tainty. Follow-up of the patient with microhema- Chronic indwelling foreign body in the urinary tract
turia depends on the history and evaluation. In

160 n engl j med 385;2  nejm.org  July 8, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Hematuria in Adults

In a 2018 review of guidelines and guidance19 ety of Urodynamics, Female Pelvic Medicine and
on microhematuria, the lack of consensus on Urogenital Reconstruction. The assessment now
several points was clear. However, most of the favored is based on risk categories (Table 2). The
guidelines state that the presence of 3 or more current guidelines comprise recommendations
red cells in one, two, or three urinalyses justifies that are intended to control costs and limit risks
the diagnosis of microhematuria, whereas the associated with “overevaluation” of persons with
Japanese Urological Association requires 5 or a low likelihood of having urinary tract cancer
more red cells. Some other guidelines require a while avoiding “underevaluation” of persons at
positive dipstick test. high risk. Not surprisingly, the guidelines sug-
Tan et al. recently developed a Haematuria gest a shared decision-making plan between
Cancer Risk Score (HCRS)64 to aid clinicians in patient and clinician. Many other guidelines and
considering evaluation. Using the DETECT 1 guidance documents67-72 are available, and they
cohort, the investigators created a test cohort of agree about the importance of not missing a
3539 patients in 40 hospitals in the United King- urinary tract cancer.
dom (ClinicalTrials.gov number, NCT02676180)
and a Swiss validation cohort of 656 persons. Sum m a r y
Use of the HCRS score, which is based on the
patient’s age and sex, type of hematuria, and Hematuria is an important sign that may con-
smoking history, appeared to detect more can- note serious disease, yet on many occasions no
cers than were found by following the United specific cause is identified. A high index of sus-
Kingdom’s National Institute for Health and picion and good communication between clini-
Care Excellence (NICE) guidelines65 or the AUA cian and patient can result in a reasoned and
guidelines.5 There is also another risk score, reasonable approach to evaluation and therapy.
which is not in general use.66
The AUA guidelines were updated in 2020,5 No potential conflict of interest relevant to this article was
reported.
through a panel created 2 years earlier by AUA Disclosure forms provided by the author are available with the
Education and Research (AUAER) and the Soci- full text of this article at NEJM.org.

References
1. Shokeir AA, Hussein MI. The urology wide claims-based investigation. Cancer bin and hemoglobin in biological fluids.
of Pharaonic Egypt. BJU Int 1999;​84:​755- 2014;​120:​555-61. Ann Clin Lab Sci 1980;​10:​493-9.
61. 8. Rabinowitz DE, Buford K, Wood AM, 15. Cohen RA, Brown RS. Microscopic
2. Armstrong JA. Urinalysis in Western et al. Incidence of significant findings of hematuria. N Engl J Med 2003;​ 348:​
culture: a brief history. Kidney Int 2007;​ microhematuria workup in women-what 2330-8.
71:​384-7. guidelines work best? Urology 2021;​151:​ 16. Grossfeld GD, Litwin MS, Wolf JS,
3. Mohr DN, Offord KP, Owen RA, 19-23. et al. Evaluation of asymptomatic micro-
Melton LJ III. Asymptomatic microhema- 9. Ferlay J, Soerjomataram I, Dikshit R, scopic hematuria in adults: the American
turia and urologic disease. A population- et al. Cancer incidence and mortality Urological Association best practice pol-
based study. JAMA 1986;​256:​224-9. worldwide: sources, methods and major icy — part I: definition, detection, preva-
4. Woolhandler S, Pels RJ, Bor DH, Him- patterns in GLOBOCAN 2012. Int J Can- lence, and etiology. Urology 2001;​57:​599-
melstein DU, Lawrence RS. Dipstick uri- cer 2015;​136(5):​E359-E386. 603.
nalysis screening of asymptomatic adults 10. Jubber I, Shariat SF, Conroy S, et al. 17. Sutton JM. Evaluation of hematuria in
for urinary tract disorders. I. Hematuria Non-visible haematuria for the detection adults. JAMA 1990;​263:​2475-80.
and proteinuria. JAMA 1989;​262:​1214-9. of bladder, upper tract, and kidney can- 18. Makino H, Nishimura S, Soda K,
5. Barocas DA, Boorjian SA, Alvarez RD, cer: an updated systematic review and Takaoka M, Kaneshige T, Ota Z. Mecha-
et al. Microhematuria: AUA/SUFU Guide- meta-analysis. Eur Urol 2020;​77:​583-98. nism of hematuria. I. Electron microscop-
line. J Urol 2020;​204:​778-86. 11. Simerville JA, Maxted WC, Pahira JJ. ic demonstration of the passage of a red
6. Ark JT, Alvarez JR, Koyama T, et al. Urinalysis: a comprehensive review. Am blood cell through a glomerular capillary
Variation in the diagnostic evaluation Fam Physician 2005;​71:​1153-62. wall in rat masugi nephritis. Virchows
among persons with hematuria: influ- 12. Rao PK, Gao T, Pohl M, Jones JS. Dip- Arch B Cell Pathol Incl Mol Pathol 1986;​
ence of gender, race and risk factors for stick pseudohematuria: unnecessary con- 50:​199-208.
bladder cancer. J Urol 2017;​198:​1033-8. sultation and evaluation. J Urol 2010;​183:​ 19. Linder BJ, Bass EJ, Mostafid H, Boor-
7. Cohn JA, Vekhter B, Lyttle C, Stein- 560-4. jian SA. Guideline of guidelines: asymp-
berg GD, Large MC. Sex disparities in di- 13. Litwin MS, Graham SD Jr. False-posi- tomatic microscopic haematuria. BJU Int
agnosis of bladder cancer after initial tive hematuria. JAMA 1985;​254:​1724. 2018;​121:​176-83.
presentation with hematuria: a nation- 14. Adams EC. Differentiation of myoglo- 20. Yuste C, Gutierrez E, Sevillano AM,

n engl j med 385;2  nejm.org  July 8, 2021 161


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

et al. Pathogenesis of glomerular haema- diagnostic algorithm. JAMA Intern Med A, Tanguay S, Kassouf W. Is the perfor-
turia. World J Nephrol 2015;​4:​185-95. 2017;​177:​808-9. mance of urinary cytology as high as re-
21. Praga M, Gutierrez-Millet V, Navas JJ, 36. American Cancer Society. Cancer sta- ported historically? A contemporary analy-
et al. Acute worsening of renal function tistics center (https://cancerstatisticscenter​ sis in the detection and surveillance of
during episodes of macroscopic hematu- .­cancer​.­org/​­?_ga=​­2​.­82720342​.­1930708337​ bladder cancer. Urol Oncol 2014;​ 32(1):​
ria in IgA nephropathy. Kidney Int 1985;​ .­1595527626​-­1692772379​.­1595527626#!/​­). 27.e1-6.
28:​69-74. 37. Jung H, Gleason JM, Loo RK, Patel 52. Kassouf W. The value of urine cytol-
22. Vivante A, Afek A, Frenkel-Nir Y, et al. HS, Slezak JM, Jacobsen SJ. Association of ogy in the workup of hematuria. Cancer
Persistent asymptomatic isolated micro- hematuria on microscopic urinalysis and Cytopathol 2016;​124:​303-4.
scopic hematuria in Israeli adolescents risk of urinary tract cancer. J Urol 2011;​ 53. Soria F, Krabbe L-M, Todenhöfer T,
and young adults and risk for end-stage 185:​1698-703. et al. Molecular markers in bladder can-
renal disease. JAMA 2011;​306:​729-36. 38. Madeb R, Golijanin D, Knopf J, et al. cer. World J Urol 2019;​37:​31-40.
23. Kido R, Shibagaki Y, Iwadoh K, et al. Long-term outcome of patients with a neg- 54. Sathianathen NJ, Butaney M, Weight
Persistent glomerular hematuria in living ative work-up for asymptomatic micro- CJ, Kumar R, Konety BR. Urinary bio-
kidney donors confers a risk of progres- hematuria. Urology 2010;​75:​20-5. markers in the evaluation of primary
sive kidney disease in donors after hemi- 39. Nielsen M, Qaseem A. Hematuria as hematuria: a systematic review and meta-
nephrectomy. Am J Transplant 2010;​ 10:​ a marker of occult urinary tract cancer: analysis. Bladder Cancer 2018;​4:​353-63.
1597-604. advice for high-value care from the Amer- 55. Wilson JL Jr, Antoniassi MP, Lopes PI,
24. Moreno JA, Sevillano Á, Gutiérrez E, ican College of Physicians. Ann Intern Azevedo H. Proteomic research and diag-
et al. Glomerular hematuria: cause or Med 2016;​164:​488-97. nosis in bladder cancer: state of the art
consequence of renal inflammation? Int 40. Murakami S, Igarashi T, Hara S, review. Int Braz J Urol 2021;​47:​503-14.
J Mol Sci 2019;​20:​2205. Shimazaki J. Strategies for asymptomatic 56. Suzuki H. Biomarkers for IgA ne-
25. Moreno JA, Martín-Cleary C, Gutiér- microscopic hematuria: a prospective phropathy on the basis of multi-hit patho-
rez E, et al. AKI associated with macro- study of 1,034 patients. J Urol 1990;​144:​ genesis. Clin Exp Nephrol 2019;​23:​26-31.
scopic glomerular hematuria: clinical and 99-101. 57. Coppo R. Biomarkers and targeted
pathophysiologic consequences. Clin J Am 41. Khadra MH, Pickard RS, Charlton M, new therapies for IgA nephropathy. Pedi-
Soc Nephrol 2012;​7:​175-84. Powell PH, Neal DE. A prospective analy- atr Nephrol 2017;​32:​725-31.
26. Sheerin NS, Sacks SH, Fogazzi GB. In sis of 1,930 patients with hematuria to 58. Benonisdottir S, Kristjansson RP,
vitro erythrophagocytosis by renal tubu- evaluate current diagnostic practice. J Urol Oddsson A, et al. Sequence variants as-
lar cells and tubular toxicity by haemoglo- 2000;​163:​524-7. sociating with urinary biomarkers. Hum
bin and iron. Nephrol Dial Transplant 42. Davis R, Jones JS, Barocas DA, et al. Mol Genet 2019;​28:​1199-211.
1999;​14:​1391-7. Diagnosis, evaluation and follow-up of 59. Tan MP, Attard G, Huddart RA. Circu-
27. Willis GC, Tewelde SZ. The approach asymptomatic microhematuria (AMH) in lating tumour DNA in muscle-invasive
to the patient with hematuria. Emerg Med adults: AUA guideline. J Urol 2012;​188:​ bladder cancer. Int J Mol Sci 2018;​ 19:​
Clin North Am 2019;​37:​755-69. 2473-81. 2568.
28. Mishriki SF, Vint R, Somani BK. Half 43. Birch DF, Fairley KF. Haematuria: glo- 60. Nuzzo PV, Berchuck JE, Korthauer K,
of visible and half of recurrent visible he- merular or non-glomerular? Lancet 1979;​ et al. Detection of renal cell carcinoma
maturia cases have underlying pathology: 2:​845-6. using plasma and urine cell-free DNA
prospective large cohort study with long- 44. Pollock C, Liu PL, Györy AZ, et al. methylomes. Nat Med 2020;​26:​1041-3.
term followup. J Urol 2012;​187:​1561-5. Dysmorphism of urinary red blood cells 61. Moyer VA. Screening for bladder can-
29. Mishriki SF, Grimsley SJS, Nabi G. In- — value in diagnosis. Kidney Int 1989;​36:​ cer: U.S. Preventive Services Task Force
cidence of recurrent frank hematuria and 1045-9. recommendation statement. Ann Intern
urological cancers: prospective 6.9 years 45. Köhler H, Wandel E, Brunck B. Acan- Med 2011;​155:​246-51.
of followup. J Urol 2009;​182:​1294-8. thocyturia — a characteristic marker for 62. Bauer SR, Carroll PR, Grady D. Hema-
30. Expert Panel on Urological Imaging, glomerular bleeding. Kidney Int 1991;​40:​ turia practice guidelines that explicitly
Wolfman DJ, Marko J, et al. ACR Appro- 115-20. consider harms and costs. JAMA Intern
priateness Criteria® hematuria. J Am Coll 46. Kitamoto Y, Tomita M, Akamine M, Med 2019;​179:​1362-4.
Radiol 2020;​17(5S):​S138-S147. et al. Differentiation of hematuria using 63. Georgieva MV, Wheeler SB, Erim D,
31. Wood DP Jr. Is it time to revise our a uniquely shaped red cell. Nephron 1993;​ et al. Comparison of the harms, advan-
evaluation of hematuria? J Urol 2009;​182:​ 64:​32-6. tages, and costs associated with alterna-
1253-4. 47. Hamadah AM, Gharaibeh K, Mara tive guidelines for the evaluation of hema-
32. Bolenz C, Schröppel B, Eisenhardt A, KC, et al. Urinalysis for the diagnosis of turia. JAMA Intern Med 2019;​179:​1352-62.
Schmitz-Dräger BJ, Grimm M-O. The in- glomerulonephritis: role of dysmorphic 64. Tan WS, Ahmad A, Feber A, et al. De-
vestigation of hematuria. Dtsch Arztebl red blood cells. Nephrol Dial Transplant velopment and validation of a haematuria
Int 2018;​115:​801-7. 2018;​33:​1397-403. cancer risk score to identify patients at
33. Tan WS, Feber A, Sarpong R, et al. 48. Shichiri M, Hosoda K, Nishio Y, et al. risk of harbouring cancer. J Intern Med
Who should be investigated for haematu- Red-cell-volume distribution curves in di- 2019;​285:​436-45.
ria? Results of a contemporary prospec- agnosis of glomerular and non-glomeru- 65. Suspected cancer:​ recognition and
tive observational study of 3556 patients. lar haematuria. Lancet 1988;​1:​908-11. referral. NICE guideline. London:​Na-
Eur Urol 2018;​74:​10-4. 49. Mishriki SF, Aboumarzouk O, Vint R, tional Institute for Health and Care Ex-
34. Halpern JA, Chughtai B, Ghomrawi Grimsley SJ, Lam T, Somani B. Routine cellence, 2015 (https://www​.­n ice​.­org​.­u k/​
H. Cost-effectiveness of common diagnos- urine cytology has no role in hematuria ­g uidance/​­ng12/​­resources/​­suspected​
tic approaches for evaluation of asymp- investigations. J Urol 2013;​189:​1255-8. -­c ancer​-­recognition​-­a nd​-­referral​-­pdf​
tomatic microscopic hematuria. JAMA 50. Hof land CA, Mariani AJ. Is cytology -­1837268071621).
Intern Med 2017;​177:​800-7. required for a hematuria evaluation? 66. Wu X, Lin J, Grossman HB, et al. Pro-
35. Subak LL, Grady D. Asymptomatic J Urol 2004;​171:​324-6. jecting individualized probabilities of
microscopic hematuria — rethinking the 51. Yafi FA, Brimo F, Auger M, Aprikian developing bladder cancer in white indi-

162 n engl j med 385;2  nejm.org  July 8, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Hematuria in Adults

viduals. J Clin Oncol 2007;​25:​4974-81. 69. Wollin T, Laroche B, Psooy K. Cana- uation of the new American Urological
67. van der Molen AJ, Hovius MC. Hema- dian guidelines for the management of Association guidelines risk classification
turia: a problem-based imaging algo- asymptomatic microscopic hematuria in for hematuria. J Urol 2021;​205:​1387-93.
rithm illustrating the recent Dutch guide- adults. Can Urol Assoc J 2009;​3:​77-80. 72. Skaggs AW, Loehfelm TW, Fanana-
lines on hematuria. AJR Am J Roentgenol 70. Anderson J, Fawcett D, Feehally J, pazir G, Dall’Era M, Corwin MT. Utiliza-
2012;​198:​1256-65. Goldberg L, Kelly J, MacTier R. Joint con- tion and yield of CT urography: are the
68. Assmus MA, Beyer DB, Hanks J, et al. sensus statement on the initial assess- American Urological Association guide-
Quality and cost assessment of Canadian ment of haematuria. July 2008 (https:// lines for imaging of patients with asymp-
Urological Association microscopic hema- www​.­baus​.­org​.­u k/​­_userfiles/​­pages/​­f iles/​ tomatic microscopic hematuria being fol-
turia guidelines in clinical practice: turn- ­News/​­haematuria_consensus_guidelines lowed? AJR Am J Roentgenol 2021;​216:​
ing urine into gold. Can Urol Assoc J _July_2008​.­pdf). 106-10.
2019;​13:​406-11. 71. Woldu SL, Ng CK, Loo RK, et al. Eval- Copyright © 2021 Massachusetts Medical Society.

images in clinical medicine


The Journal welcomes consideration of new submissions for Images in Clinical
Medicine. Instructions for authors and procedures for submissions can be found
on the Journal’s website at NEJM.org. At the discretion of the editor, images that
are accepted for publication may appear in the print version of the Journal,
the electronic version, or both.

n engl j med 385;2  nejm.org  July 8, 2021 163


The New England Journal of Medicine
Downloaded from nejm.org by Salvador Macias-Díaz on August 6, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The New England Journal of Medicine
Downloaded from nejm.org at The University Of Illinois on November 26, 2015. For personal use only. No other uses without permission.
From the NEJM Archive. Copyright © 2009 Massachusetts Medical Society. All rights reserved.
The New England Journal of Medicine
Downloaded from nejm.org at The University Of Illinois on November 26, 2015. For personal use only. No other uses without permission.
From the NEJM Archive. Copyright © 2009 Massachusetts Medical Society. All rights reserved.
MARIJUANA USE DOES NOT SPURIOUSLY ELEVATE
SERUM HUMAN CHORIONIC GONADOTROPIN LEVELS

GLENN D. BRAUNSTEIN, M.D.


RONALD THOMPSON, M.D.
STANLEY GROSS, M.D.
JAMES R. SOARES, PH.D.

From the Departments of Medicine and Radiation Therapy,


Cedars-Sinai Medical Center-UCLA School of Medicine,
Los Angeles, California

ABSTRACT-Marijuana use has been reported to spuriously elevate immunoreactive human cho-
rionic gonadotropin (HCG) in the serum of patients with testicular germ cell tumors. To reinvesti-
gate this finding, we measured serum HCG and AQ-tetrahydrocannabinol (A”-THC) levels in 16
men known or suspected to be marijuana users. Eight of the serum samples had measurable levels of
AQ-THC, while eight did not. None contained immunoreactive HCG. The addition of Ae-THC
directly to pooled male serum had no effect on the HCG radioimmunoassay. These results indicate
that marijuana does not artificially elevate serum HCG concentrations.

It has been well established that human cho- separated and stored at - 20°C until assayed.
rionic gonadotropin (HCG) measurements are Serum AQ-tetrahydrocannabinol (Ag-THC) was
useful for monitoring disease activity in patients measured by a specific radioimmunoassay. l5
with germ cell tumors of the testes.’ Spuriously HCG measurements were performed by our
elevated HCG concentrations have been noted previously described double antibody beta sub-
in some assay systems because of cross-reaction unit radioimmunoassay method using reagents
with human pituitary luteinizing hormone supplied by the National Hormone and Pi-
(HLH) ,2-5 lipid interference,s presence of tuitary Program, Baltimore, MD.le
heterophilic antibodies,7,8 or HCG binding pro- To investigate the possibility of cross-reaction
teins.Q-12 In addition, Garnick13 suggested that of marijuana in the HCG radioimmunoassay,
marijuana abuse may result in an elevation of we added the active ingredient of marijuana,
HCG in the serum of patients with germ cell A9-tetrahydrocannabinol, directly to normal
tumors of the testes. However, we were unable pooled male serum and measured the pool in
to demonstrate an alteration by marijuana on HCG radioimmunoassay (RIA).
HCG concentrations during pregnancy. l4
Therefore, we examined directly the effects of Results
marijuana on HCG measurements to determine
whether or not cannabinoids interfere with Eight of the 16 patients had measurable
quantities of A9-THC (range 5.6-34.2 ng/ml) in
HCG determinations.
their sera, while 8 had no detectable AQ-THC.
Material and Methods None of the 16 samples had measurable levels of
immunoreactive HCG.
Blood was obtained from 16 men suspected No apparent HCG activity was present in the
or known to be marijuana users. The serum was pooled male serum samples after the addition

UROLOGY i JUNE 1985 i VOLUMEXXV,NUMBER6 605


from 5 to 500 pg of Ag-THC to the samples prior Los Angeles, California 90048
to the assay. (DR. BRAUNSTEIN)

ACKNOWLEDGMENTS. To Ms. Helene Zauderer for cleri-


Comment cal help, and the support of the Linda Simon Cancer Re-
search Fund.
Our results indicate that neither Ag-THC nor
other cannabinoid metabolites present in the References
serum of individuals who smoke the drug inter-
1. Braunstein GD: HCG expression in trophoblastic and non-
feres in the HCG radioimmunoassay or leads to trophoblastic tumors, in Fishman WH (Ed): Oncodevelopmental
the apparent, but spurious, presence of HCG in Markers, Biologic, Diagnostic and Monitoring Aspects, New York,
blood. This finding is supported by the recent Academic Press, 1983, p 351.
2. Catalona WJ, Vaitukaitis JL, and Fair WR: Falsely positive
report by Hogan and co-workers” who found specific human chorionic gonadotropin assays in patients with tes-
no effect of the synthetic cannabinoid nabilone titular tumors: conversion to negative with testosterone adminis-
on serum HCG concentrations in men receiving tration, J Urol 122: 126 (1981).
3. Ketchum C, et al: False increases of beta-subunit cho-
combination chemotherapy for testicular can- riogonadotropin in commercial kit results because of cross reactiv-
cer. In addition, we previously had shown no ity with lutropin, Clin Chem 28: 1998 (1982).
difference in the serum concentrations of three 4. Javadpour N, and Soares T: False-positive and false-nega-
tive alphafetoprotein and human chorionic gonadotropin assays
trophoblastic proteins (HCG, pregnancy-spe- in testicular cancer: a double blind studv. Cancer 48: 2279 (1981).
cific-betal-glycoprotein [SPr], and placenta 5. Fowler JE, et al: Commercial radioimmunoassay for beta
lactogen) at any time during pregnancy in 13 subunit of human chorionic gonadotropin: false positive deter-
minations due to elevated serum luteinizing hormone, ibid 49:
women who used marijuana regularly through- 136 (1982).
out pregnancy in comparison with 13 pregnant 6. Fritz TJ, Bunker DM, and Lawlor J: Hyperlipidemia inter-
control women who did not use marijuana.14 ference in radioimmunoassavs. Clin Chem 28: 2325 (1982).
7. Baulieu JL, et al: Falsely elevated results of radidimmunoas-
Thus, it is unlikely that even if a patient with a says using double antibody method: arguments for a third anti-
testicular tumor containing trophoblastic ele- rabbit IgG antibody present in certain human sera, Eur J Nucl
ments which produced HCG smoked mari- Med 7: 121 (1982).
8. Vladutiu AO, et al: Heterophilic antibodies interfering with
juana heavily, A9-THC or one of its derivatives radioimmunoassay. A false-positive pregnancy test, JAMA 248:
would stimulate the tumor to secrete increased 2489 (1982).
quantities of the hormone. 9. Chan DW, Bill MJ, and Rosenshein N: Specific binding pro-
teins for human chorionic gonadotrophin in a patient with
It should be stressed that our beta HCG ra- trophoblastic disease, Am J Clin Path01 76: 214 (1981).
dioimmunoassay utilizes reagents supplied by 10. Braunstein GD, et al: Characterization of antihuman cho-
the National Hormone and Pituitary Program, rionic gonadotropin serum antibody appearing after ovulation in-
duction, J Clin Endocrinol Metab 57: 1164 (1983).
Baltimore, MD. Since the antisera in the com- 11. Musch K, Wolf AS, and Lauritzen C: Antibodies to human
mercial beta HCG RIA kits differ in their reac- chorionic gonadotropin in humans, Clin Chim Acta 113: 95
tivity to HCG and HLH,18 our results may not (1981).
12. Nieschlag E, Bernitz S, and Topert M: Antigenicity of hu-
be found with other HCG assays. Several man chorionic gonadotropin preparations in men, Clin Endo-
authors have noted that high concentrations of crinol 16: 483 (1982).
HLH may result in false positive elevations of 13. Garnick MB: Spurious rise in human chorionic gonadotro-
pin induced by marihuana in patients with testicular cancer, N
HCG with some HCG RIA kits in patients with Engl J Med 303: 1177 (1980).
testicular tumors.2-5 This problem can be cir- 14. Braunstein GD, et al: Pregnancy hormone concentrations
cumvented by using more specific reagents or in marijuana users, Life Sci 33: 195 (1983).
15. Yeager ER et al: A8-tetrahydrocannabinol by GLC-MS val-
by measuring the HCG concentration following idated radioimmunoassays of hemolyzed blood or serum, J Anal
suppression of HLH by testosterone administra- Toxic01 5: 81 (1981).
16. Braunstein GD, et al: First trimester chorionic gonadotro-
tion.2.3
pin measurements as an aid in the diagnosis of early pregnancy
A rise in serum HCG concentration measured disorders, Am J Obstet Gynecol 131: 25 (1978).
by a reliable, specific assay in a patient with a 17. Hogan P, et al: Cannabinoids and hCG levels in patients
testicular tumor should be considered evidence with testicular cancer, Lancet 2: 1144 (1983).
18. Rasor JL, Farber S, and Braunstein GD: An evaluation of
of relapse and not be attributed to the use of 10 kits for determination of human choriogonadotropin in serum,
cannabis derivatives. Clin Chem 29: 1828 (1983).

606 UROLOGY / JUNE 1985 / VOLUME XXV, NUMBER 6


VOLUME 33 䡠 NUMBER 8 䡠 MARCH 10 2015

JOURNAL OF CLINICAL ONCOLOGY O R I G I N A L R E P O R T

Randomized Controlled Trial of the Prophylactic Effect of


Urea-Based Cream on Sorafenib-Associated Hand-Foot
Skin Reactions in Patients With Advanced
Hepatocellular Carcinoma
ZhengGang Ren, KangShun Zhu, HaiYan Kang, MinQiang Lu, ZengQiang Qu, LiGong Lu, TianQiang Song,
WeiPing Zhou, Hui Wang, WeiZhu Yang, Xuan Wang, YongPing Yang, LeHua Shi, YuXian Bai,
XiaoFeng Guo, and Sheng-Long Ye
ZhengGang Ren and Sheng-Long Ye,
Liver Cancer Institute, Zhongshan A B S T R A C T
Hospital, Fudan University, and Key
Laboratory of Carcinogenesis and Purpose
Cancer Invasion, Ministry of Education; To assess whether urea-based cream (UBC) has prophylactic benefits on sorafenib-induced
ZengQiang Qu, WeiPing Zhou, and hand-foot skin reaction (HFSR) in patients with advanced hepatocellular carcinoma (HCC).
LeHua Shi, Eastern Hepatobiliary
Surgery Hospital of the Second Military Patients and Methods
Medical University, Shanghai; In this randomized, open-label trial, 871 patients with advanced HCC throughout China were
KangShun Zhu and MinQiang Lu, The treated with 10% UBC three times per day plus best supportive care (BSC; n ⫽ 439) or BSC alone
Third Affiliated Hospital of Sun Yat-sen
excluding all creams (n ⫽ 432), starting on day 1 of sorafenib treatment, for up to 12 weeks. HFSR
University; LiGong Lu, Guangdong
Provincial People’s Hospital, Guang-
was assessed every 2 weeks and at 14 weeks for patients completing the study. Once HFSR
dong; HaiYan Kang, 301 Military Hospi- occurred, patients were allowed any cream, including a UBC.
tal; YongPing Yang, 302 Military
Hospital; XiaoFeng Guo, Chinese Anti- Results
Cancer Association, Beijing; TianQiang The 12-week incidence of any grade HFSR was significantly lower in the UBC group versus the
Song, Tianjin Cancer Hospital, Tianjin; BSC-alone group (56.0% v 73.6%, respectively; odds ratio [OR], 0.457; 95% CI, 0.344 to 0.608;
Hui Wang, Jilin Provincial Tumor Hospi- P ⬍ .001), as was the incidence of grade ⱖ 2 HFSR (20.7% v 29.2%, respectively; OR, 0.635; 95%
tal, Jilin; WeiZhu Yang, Union Hospital CI, 0.466 to 0.866; P ⫽ .004). Median time to first occurrence of HFSR was significantly longer in
of Fujian Medical University, Fujian;
the UBC group than the BSC-alone group (84 v 34 days, respectively; hazard ratio, 0.658; 95% CI,
Xuan Wang, The 81 Hospital of the
Chinese People’s Liberation Army,
0.541 to 0.799; P ⬍ .001). Elevated AST was associated with increased risk of HFSR but did not
Nanjing; and YuXian Bai, Heilongjiang alter the treatment effect of UBC. UBC plus BSC, compared with BSC alone, did not affect the
Provincial Cancer Hospital, Heilongjiang, sorafenib dose reduction or interruption rate (9.1% v 11.8%, respectively; P ⫽ .1937), response
People’s Republic of China. rate (11.1% v 10.1%, respectively; P ⫽ .6674), or disease control rate (98.8% v 98.2%,
Published online ahead of print at respectively; P ⫽ .5350) at week 12.
www.jco.org on February 9, 2015.
Conclusion
Sponsored by the Chinese Anti-Cancer UBC prophylaxis in patients with advanced HCC starting sorafenib reduced HFSR rates, extended
Association and Bayer HealthCare.
the time to first occurrence of HFSR, and improved patient quality of life compared with BSC.
Authors’ disclosures of potential Blinded, randomized, placebo-controlled trials to determine the role of UBC on the incidence and
conflicts of interest and author severity of HFSR are warranted.
contributions are found at the end of
this article.
J Clin Oncol 33:894-900. © 2015 by American Society of Clinical Oncology
Clinical trial information: NCT01934829.

Corresponding author: Sheng-Long Ye,


mal edema, parakeratosis, and hyperkeratosis.12-15 A
MD, Liver Cancer Institute, Zhongshan INTRODUCTION
Hospital, Fudan University, 180 Fenglin meta-analysis of 24 clinical trials including more
Rd, Shanghai 200032, People’s Repub- The multikinase inhibitor sorafenib1-3 was shown to than 6,000 patients with various malignancies found
lic of China; e-mail: slye@shmu.edu.cn. that the rates of any grade and grade 3 HFSR in
be effective in two phase III clinical trials in patients
© 2015 by American Society of Clinical with advanced hepatocellular carcinoma (HCC)4,5 sorafenib-treated patients were 39% and 9%, re-
Oncology
and has become standard systemic therapy for HCC spectively.16 The two phase III trials of sorafenib in
0732-183X/15/3308w-894w/$20.00
worldwide, including in China.6-11 Dermatologic patients with HCC reported any grade HFSR rates of
DOI: 10.1200/JCO.2013.52.9651 adverse events (AEs), however, are frequent, includ- 21% and 45% and grade 3 HFSR rates of 8% and 11%,
ing hand-foot skin reaction (HFSR; palmar and/or withHFSRbeingoneofthemostcommonAEsleading
plantar erythrodysesthesia), rash, and alopecia. to dose reduction.4,5 High rates of any grade HFSR
HFSR associated with tyrosine kinase inhibitors (72% to 82%) have been reported during sorafenib
(TKIs) is characterized by keratinocytic necrosis, der- treatment of Asian patients with HCC.17,18

894 © 2015 by American Society of Clinical Oncology

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Prophylactic Urea-Based Cream for Sorafenib HFSR in Advanced HCC

Screened
(N = 901)

Excluded (n = 30)

Randomly assigned
(n = 871)

Assigned prophylactic Assigned BSC only


urea-based cream + BSC excluding all creams
(n = 439) (n = 432)

Evaluable for incidence of HFSR Evaluable for incidence of HFSR


Fig 1. Study design. BSC, best support-
(n = 439) (n = 432)
ive care; HFSR, hand-foot skin reaction.

Nonevaluable (n = 85) Nonevaluable patients (n = 87)


patients Missing informed (n = 5)
Missing informed (n = 4) consent date
consent date Calculated time (n = 6)
Calculated time (n = 14) to first event
to first event outside of limit
outside of limit Missing date or (n = 76)
Missing date or (n = 67) grade of HFSR
grade of HFSR

Evaluable for time to first HFSR Evaluable for time to first HFSR
(n = 354) (n = 345)

Despite prolonging survival in patients with HCC, the advan- treatment, willingness and ability (with assistance if necessary) to fill out
tages of sorafenib are reduced by its toxicities, including patient-reported questionnaires, discontinuation of cancer therapy at least 3
HFSR.12,13,19,20 Although not life threatening, HFSR can severely af- weeks before study entry, and life expectancy of ⱖ 3 months. Exclusion criteria
fect health-related quality of life (HRQoL), including physical, psy- included previous treatment with sorafenib, ongoing concomitant anticancer
therapy, or concurrent enrollment in another clinical trial. Each patient pro-
chological, and social well-being,12,19,21 resulting in sorafenib dose
vided written informed consent, and the study protocol was approved by the
reductions and/or discontinuations and potentially altering survival investigative review and/or ethics board of each participating institution.
outcomes. Appropriate HFSR prophylaxis and management are nec-
essary to ensure proper sorafenib dosing and enhance HRQoL.
Study Design
Mild hyperkeratosis, an early sign of HFSR, may be the only
All patients receiving sorafenib were randomly assigned 1:1 to either
manifestation of sorafenib-associated HFSR. Creams containing urea UBC (10% urea; Eucerin, Beiersdorf, Hamburg, Germany) three times per day
are widely used to treat hyperkeratotic conditions, including psoria- plus BSC (n ⫽ 439) for AEs other than HFSR or BSC alone (n ⫽ 432)
sis,22 and have been recommended for TKI-related HFSR.23 These excluding use of all creams (Fig 1). HFSR was evaluated using a modified
creams are generally inexpensive, readily available, and well tolerated. grading system specific to HFSR symptoms (Appendix Table A2, online only).
At present, however, there is no consensus on best supportive care All other AEs were evaluated by the treating physician using National Cancer
(BSC) for management of TKI-associated HFSR, and to our knowl- Institute Common Terminology Criteria for Adverse Events (version 3). Once
edge, no randomized controlled trials have evaluated methods to HFSR occurred, patients were allowed any cream, per the treating physician.
Patients were examined every 2 weeks for 12 weeks and followed up at week 14.
prevent or palliate sorafenib-associated HFSR.23,24 This large random-
During each visit, concomitant medications, laboratory values (hematology
ized controlled trial evaluated the prophylactic effects of a urea-based and chemistry), and 26 preselected/precoded AEs were recorded, including
cream (UBC) on the incidence of sorafenib-associated HFSR in pa- AEs commonly associated with sorafenib treatment. Patients returned any
tients with advanced HCC. unused urea cream at each visit. Patients underwent abdominal computed
tomography or magnetic resonance imaging scans to measure and evaluate
tumors at screening, at week 6 or 8, and at the final (week 12) or follow-up
PATIENTS AND METHODS (week 14) visit.
Sorafenib dose modifications (interruptions and reductions) for derma-
Patients tologic toxicities, including HFSR, were allowed as predefined in the study
This investigator-initiated, randomized, open-label trial assessed protocol. Dosing was uninterrupted for grade 1 toxicity. Doses were reduced
whether a topical UBC could reduce the incidence and severity of HFSR in for grade 2 toxicities until resolution to grade 0 to 1, with resumption at the
patients with advanced HCC starting sorafenib at 64 centers throughout China next cycle of full-dose sorafenib (400 mg twice per day) after the first incident,
(Appendix Table A1, online only). Other inclusion criteria included willing- reduction of sorafenib dose to 400 mg once every day after the second incident,
ness and ability to report for clinic visits every 2 weeks for the first 3 months of and reduction of sorafenib dose to 400 mg every 2 days after subsequent

www.jco.org © 2015 by American Society of Clinical Oncology 895

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ren et al

incidents. Sorafenib doses were interrupted for grade 3 toxicities until resolu- Response rates, rates of common AEs, and rates of dose reductions and
tion to grade 0 to 1, with reduction at the next cycle to sorafenib 400 mg once discontinuations as a result of HFSR were compared using ␹2 or Fisher’s exact
every day after the first incident and to sorafenib 400 mg every 2 days after test. Mean HFSR symptom score and HF-QoL daily activity score were com-
subsequent incidents. pared using the Wilcoxon rank sum test.

Study End Points


The primary end point was the incidence of any grade HFSR within 12 RESULTS
weeks of starting sorafenib. Secondary end points included incidence of grade
ⱖ 2 HFSR within 12 weeks, time to first occurrence of HFSR, duration of
Of the 901 screened patients with HCC, 871 were randomly as-
HFSR, percentages of patients with sorafenib dose reductions/interruptions
and discontinuations, HFSR-associated HRQoL using the Hand-Foot Reac- signed to UBC plus BSC (n ⫽ 439) or BSC alone (n ⫽ 432) when
tion Quality of Life (HF-QoL) questionnaire, and tumor response according starting sorafenib 400 mg twice per day (Fig 1). At baseline, the two
to RECIST criteria. The population evaluable for HFSR consisted of all patients groups were generally well matched demographically and clini-
who received at least one dose of sorafenib. The efficacy population consisted cally, including in the etiology of HCC, ECOG PS, previous anti-
of all patients who received at least one dose of sorafenib and for whom cancer therapy, and history of disease (Table 1), except that
computed tomography scans were available for tumor assessment. transarterial embolizartion was significantly more frequent in the
The HF-QoL questionnaire (Anderson et al, manuscript in preparation)
UBC plus BSC group (10.1% v 6.3% for BSC alone; P ⫽ .0409).
was adapted from the validated Dermatology-Specific QoL questionnaire, a
multidimensional questionnaire assessing HRQoL across a variety of skin Analysis of history of dermatosis showed that one patient in the
diseases.25 The HF-QoL includes 48 items covering HFSR symptoms on the UBC plus BSC group had a skin eruption, and one patient each in
feet, hands and fingers, and other body areas; the impact of these symptoms on the BSC group had psoriasis and an unidentified dermatosis.
a patient’s ability to sleep and perform daily activities; and the effects of HFSR
on a patient’s social life and feelings or mood. The HF-QoL questionnaire
includes the following three global items: a visual scale measuring overall
health, a question on overall rating of HFSR severity, and a global rating of
change in HFSR. Table 1. Baseline Demographics and Clinical Characteristics of the
For HF-QoL parameters, the minimal clinically important difference, or Evaluable Population
the smallest change indicative of a clinically important improvement or de- UBC ⫹ BSC BSC Alone
cline, was estimated from change in scores corresponding to a unit change in (n ⫽ 439) (n ⫽ 432)
toxicity from grade 0 to grade 1, change in self-rated severity of 1 (better), and Demographic or Clinical No. of No. of
change from no to mild symptoms. Thus, differences of ⱖ 4 and ⱖ 6 units in Characteristic Patients % Patients %
HF-QoL Daily Activities and Symptoms, respectively, were considered clini-
Age, years
cally meaningful.
Median 51.8 52.0
Range 20.4-86.4 20.2-84.3
Statistical Analysis Male 377 85.9 368 85.2
We estimated that 45% of patients with HCC would develop HFSR Weight, kg
during the first 12 weeks of sorafenib treatment and that the UBC would Median 67.0 66.5
reduce the relative risk by 25%, resulting in a nominal HFSR incidence of Range 40.5-88.0 40.5-101.0
33.75%. With a one-sided ␣ ⫽ .025 and a power of 90%, we calculated a HCC etiology
requirement for 412 patients per group, or a total of 824 patients. To allow for Hepatitis B 339 77.2 332 76.9
a drop-out rate of 10%, 900 patients were screened. Hepatitis C 14 3.2 11 2.5
The primary end point of any grade HFSR within 12 weeks of starting ECOG performance score
sorafenib and the incidence of HFSR at each study visit were compared in the 0 209 47.7 201 46.5
two groups using the ␹2 or Fisher’s exact test. Correlations between treatment 1 211 48.2 215 49.8
effects and severity of HFSR were assessed using the Mantel-Haenszel test, 2 17 3.9 16 3.7
comparing grade 0 versus grade 1 versus grade 2 or 3. An exploratory logistic Previous therapyⴱ
regression analysis was used to identify demographic and clinical factors pre- Any 301 68.7 293 67.8
dictive of HFSR within 12 weeks of starting sorafenib. To identify potential Hepatectomy 107 24.4 100 23.1
predictors of HFSR, a multivariable logistic regression model was used with a TACE 201 45.8 221 51.3
stepwise variable selection process.26 Covariates and factors investigated in- TAE 44 10.1† 27 6.3
cluded age (⬍ v ⱖ 65 years), sex, Eastern Cooperative Oncology Group Disease history
(ECOG) performance score (PS; 0 v ⱖ 1), body mass index (BMI; ⬍ v ⱖ 23.1 Alcohol/drug addiction 3 0.7 4 0.9
kg/m2), and ALT and AST concentrations (ⱖ 4 and ⱕ 40 v ⬎ 40 U/L for both). Allergy 4 0.9 2 0.5
Inclusion and exclusion criteria of P ⬍ .1 were used for stepwise selection. If the Sleep disorder 19 4.4 12 2.8
identified model contained predictive covariates other than treatment, inter- Arthritis 18 4.1 16 3.7
action terms were included to test for differential treatment effects. Hypertension 23 5.3 19 4.4
Time to first HFSR was defined as the time from date of random assign- Diabetes 11 2.5 14 3.3
ment to date of last visit or first report of HFSR; patients not experiencing Dermatosis‡ 1 0.2 2 0.5
HFSR were censored at the date of last visit. Patients who missed or with
Abbreviations: BSC, best supportive care; ECOG, Eastern Cooperative On-
invalid (eg, before random assignment) visits or who did not provide valid
cology Group; HCC, hepatocellular carcinoma; TACE, transarterial chemoem-
signed informed consent were excluded from analysis. Time to first HFSR was bolization; TAE, transarterial embolization; UBC, urea-based cream.

determined using the Kaplan-Meier method and compared using log-rank Patients may have received more than one therapy.
tests. Hazard ratio (HR) and 95% CIs were estimated by proportional hazards †P ⫽ .0409 compared with the BSC-alone group.
‡Including one patient with skin eruption in the UBC plus BSC group and
regression. Using a Cox regression model and prespecified covariates (age, sex,
one patient each with psoriasis and unidentified dermatosis in the
ECOG PS, BMI, and liver enzyme concentrations), an exploratory analysis was BSC-alone group.
performed to identify significant predictors of time to HFSR.

896 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Prophylactic Urea-Based Cream for Sorafenib HFSR in Advanced HCC

Table 2. Incidence of Worst-Grade Treatment-Emergent HFSR in the Evaluable Population Within 12 Weeks
UBC ⫹ BSC (n ⫽ 439) BSC Alone (n ⫽ 432)

Grade of HFSR No. of Patients % No. of Patients % Odds Ratio 95% CI Pⴱ


Model 1
None 193 44.0† 114 26.4† Reference group ⬍ .001
Grade 1 155 35.3 192 44.4 0.477 0.349 to 0.653
Grade 2 72 16.4 98 22.7 0.434 0.296 to 0.636
Grade 3 19 4.3 28 6.5 0.401 0.214 to 0.750
Model 2
None 193 44.0† 114 26.4† Reference group ⬍ .001
Grade 1 155 35.3 192 44.4 0.477 0.349 to 0.653
Grade 2/3 91 20.7 126 29.2 0.427 0.299 to 0.609
Model 3
None 193 44.0† 114 26.4† Reference group ⬍ .001
Grade 1/2/3 246 56.0 318 73.6 0.457 0.344 to 0.608
Model 4
None/grade 1 348 79.3 306 70.8 Reference group ⬍ .004
Grade 2/3 91 20.7 126 29.2 0.635 0.466 to 0.866

Abbreviations: BSC, best supportive care; HFSR, hand-foot skin reaction; UBC, urea-based cream.

Mantel-Haenszel test.
†Includes all patients without recorded incidence of HFSR.

The incidence of any grade treatment-emergent HFSR within 12 PS, BMI, and ALT were not predictive of increased risk, abnormally
weeks of starting sorafenib was significantly lower in the UBC plus high AST levels were associated with an OR of 2.123 (95% CI, 1.521 to
BSC group than in the BSC-alone group (56.0% v 73.6%, respectively; 2.962). A logistic regression model testing for the interaction between
odds ratio [OR], 0.457; 95% CI, 0.344 to 0.608; P ⬍ .001; Table 2). treatment and AST found a similar treatment effect in patients with
Rates of moderate (grade 2) and severe (grade 3) HFSR were also normal and abnormal AST (P ⫽ .2814). A univariable logistic regres-
significantly lower in the UBC plus BSC group than the BSC-alone sion model, using only treatment, yielded results consistent with the
group (20.7% v 29.2%, respectively; OR, 0.635; 95% CI, 0.466 to 0.866; multivariable model using treatment and AST (OR, 0.457; 95% CI,
P ⫽ .004). The similarity of the ORs suggested that treatment effect 0.344 to 0.608). Application of the same univariable model to the
was not associated with HFSR severity and was similar across all grades subsets of patients with normal (OR, 0.424; 95% CI, 0.295 to 0.609)
of HFSR. The incidence of any grade HFSR at each study visit was and abnormal (OR, 0.594; 95% CI, 0.362 to 0.974) AST yielded results
lower in the UBC plus BSC group than in the BSC-alone group (Fig 2), similar to the multivariable model.
as were the rates of any grade HFSR for the 12-week observation The median time to first occurrence of HFSR was significantly
period (56.0% v 73.6%, respectively; P ⬍ .001) and the entire study longer in the UBC plus BSC group than in the BSC-alone group (84 v
period (53.8% v 70.4%, respectively; P ⬍ .001; Table 3).
A stepwise exploratory logistic regression analysis evaluated vari-
ables associated with increased risk of HFSR. Although age, sex, ECOG Table 3. Any-Grade Drug-Related Adverse Events Associated With
Sorafenib Occurring in ⬎ 1.0% of the Evaluable Populations of Both Groups

UBC ⫹ BSC BSC Alone


(n ⫽ 439) (n ⫽ 432)
60
Urea cream + BSC
Adverse No. of No. of
BSC
50 Event Patients % Patients % P
HFSR 236 53.8 304 70.4 ⬍ .001ⴱ
40 Diarrhea 46 10.5 44 10.2 .8870ⴱ
Percent

Rash 27 6.2 22 5.1 .4982ⴱ


30 Fatigue 13 3.0 19 4.4 .2597ⴱ
Alopecia 13 3.0 12 2.8 .8712ⴱ
20 Anorexia 8 1.8 10 2.3 .6095ⴱ
Hypertension 7 1.6 7 1.6 .9758ⴱ
10 Headache 6 1.4 12 2.8 .1433ⴱ
Dry skin 5 1.1 13 3.0 .0524ⴱ
0 Erythema 4 0.9 6 1.4 .5435†
0 2 4 6 8 10 12 14
NOTE. Adverse events recorded according to Common Terminology Criteria
Time (weeks) for Adverse Events (version 3).
P .1445 .0081 < .001 < .001 < .001 < .001 .0445 Abbreviations: BSC, best supportive care; HFSR, hand-foot skin reaction;
UBC, urea-based cream.
ⴱ 2
␹ test.
Fig 2. Prevalence of any-grade hand-foot skin reaction at each study visit. †Fisher’s exact test.
Statistical analysis was conducted using the ␹2 test. BSC, best supportive care.

www.jco.org © 2015 by American Society of Clinical Oncology 897

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ren et al

1.0 A 15
Urea cream + BSC
BSC
HFSR-Free Probability

12.4
0.8 11.5 11.1
10 8.9
9.3 9

Score
8.3
0.6 7.5 7.1
6.4
5.2 5.1
0.4 Urea cream + BSC 5 4.8 4.9
Median 84 days
95% CI: 45 to 93 days
HR: 0.658
0.2 BSC 95% CI: 0.541, 0.799 P .2681 .0171 < .001 < .001 .0036 .0041 .0255
Median 34 days P < .001
95% CI: 29 to 43 days 0 2 4 6 8 10 12 14

0 10 20 30 40 50 60 70 80 90 100 Time (weeks)


Time (days)
No. at risk
Urea cream 354 297 237 217 203 186 173 156 139 129
B 15 Urea cream + BSC
BSC
BSC 345 290 209 186 160 140 116 99 77 4

Fig 3. Kaplan-Meier analysis of time to first hand-foot skin reaction (HFSR) 10 9.6

Score
event in patients who received urea-based cream plus best supportive care (BSC) 8.3 7.6
and patients who received BSC alone. HR, hazard ratio. 7.2 6.9 6.4 6.2
6.0 5.3
5 4.9
4.0 4.9
3.9
3.8

34 days, respectively; HR, 0.658; 95% CI, 0.541 to 0.799; P ⬍ .001; Fig P .8630 .2406 .0015 .0365 .3876 .4937 .7796
3), with a significant reduction in HFSR occurrence by week 12 of 0 2 4 6 8 10 12 14
treatment. Exploratory Cox regression analysis showed that treatment
(HR, 0.621; 95% CI, 0.503 to 0.766; P ⬍ .001) and abnormal AST
Time (weeks)
No. at risk
concentration (HR, 1.949; 95% CI, 1.581 to 2.403; P ⫽ .001) were Urea 439 424 418 413 373 345 336 256
significant predictors of increased risk of HFSR over time. A Cox cream
BSC 432 415 414 408 372 341 332 249
model testing the interaction between treatment and AST on risk of
HFSR over time found similar treatment effects in patients with nor-
Fig 4. Hand-Foot Reaction Quality of Life outcomes. (A) Hand-foot skin
mal and abnormal AST (P ⫽ .0732). Of the 871 patients, 172 patients reaction (HFSR) symptoms and (B) HFSR daily activity over time in patients who
(19.7%), 85 in the UBC plus BSC group and 87 in the BSC-alone received urea-based cream plus best supportive care (BSC) and in patients who
group, could not be assessed for time to HFSR as a result of missing or received BSC alone. Scores expressed as mean ⫾ SE. P values were calculated
using the Wilcoxon rank sum test.
incorrect visits or lack of valid informed consent.
Treatment with UBC was associated with significant reductions
in risk of HFSR during the first 12 weeks of treatment (OR, 0.457; 95% Analysis of responses showed that the HFSR symptom score was
CI, 0.344 to 0.608; P ⬍ .001) and during the entire study period (OR, lower at each study visit from week 4 to week 14 in patients treated
0.490; 95% CI, 0.370 to 0.647; P ⬍ .001). These findings are consistent with UBC plus BSC versus BSC alone, indicating a lower symptom
with the significant reduction in risk of HFSR per unit time (HR, burden in the former group. In addition, the HFSR daily activity score
0.658; 95% CI, 0.541 to 0.799; P ⬍ .001). was lower at weeks 6 and 8 in patients receiving UBC plus BSC,
Twenty-six preselected/precoded AEs commonly associated indicating that HFSR had a reduced impact on these patients’ daily
with sorafenib were recorded at each visit. Except for HFSR, there activities (Fig 4). Analysis by RECIST criteria of the 670 patients
were no significant between-group differences in the incidence of evaluable for tumor assessment (333 patients in the UBC plus BSC
drug-related AEs (Table 3). Only HFSR and diarrhea occurred in group and 337 patients in the BSC-alone group) showed similar over-
more than 10% of patients. all response rates (11.1% v 10.1%, respectively; P ⫽ .6674) and disease
Assessment of the effects of UBC on treatment with sorafenib control rates (98.8% v 98.2%, respectively; P ⫽ .5350) at week 12.
showed that, in the UBC plus BSC and BSC-alone groups, the rates of
sorafenib dose reductions/interruptions as a result of HFSR (9.1% v
11.8%, respectively; P ⫽ .1937) and sorafenib discontinuations (1.1% DISCUSSION
v 0.7%, respectively; P ⫽ .4893) were similar.
At the initial visit, 424 (96.6%) of 439 patients in the UBC HFSR usually develops 2 to 4 weeks after initiation of treatment with a
plus BSC group and 415 (96.1%) of 432 patients in the BSC- TKI, including sorafenib, with the risk being highest at 5 weeks fol-
alone group completed HF-QoL questionnaires, making the rates lowed by a gradual decline.23,27 HFSR may be severe enough to require
of missing questionnaires at this visit 3.4% and 3.9%, respectively. TKI dose reduction and to seriously impair activities of daily liv-
At visits 2, 3, 4, 5, and 6, 418 (95.2%), 413 (94.1%), 374 (85.2%), ing.12,28,29 Despite relatively high HFSR rates, the management of
345 (78.6%), and 336 patients (76.5%), respectively, in the UBC HFSR has been reactive rather than proactive. Recent consensus rec-
plus BSC group and 413 (95.6%), 408 (94.4%), 372 (86.1%), 341 ommendations by an international, interdisciplinary expert panel30
(78.9%), and 332 patients (76.9%), respectively, in the BSC-alone included the use of over-the-counter and prescription-strength
group returned questionnaires. creams and moisturizers during treatment with TKIs.23 Anecdotal

898 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Prophylactic Urea-Based Cream for Sorafenib HFSR in Advanced HCC

evidence has suggested that several prescription-strength topical Although prophylactic UBC reduced the incidence and severity
agents, including those containing 40% urea, 0.1% tazarotene, and 5% of HFSR, patients in the control arm were not allowed use of creams
fluorouracil, benefit patients with HFSR and other dermatologic con- before development of HFSR, preventing a determination of whether
ditions.22,31,32 To our knowledge, however, no previous trial has sys- the benefits of UBC were a result of the urea or of the cream itself.
tematically tested the ability of a topical agent, started at the initiation Other limitations included the nonblinded nature of the study and the
of TKI treatment, to prevent and/or treat TKI-associated HFSR. Thus, evaluation of HFSR by treating physicians and not by central review of
this randomized, open-label trial tested whether treatment with UBC photographs of the affected areas. Concurrent central review was not
for 12 weeks had a prophylactic effect in a study of 871 patients with possible because the symptoms were acute and required immediate
advanced HCC starting sorafenib at 64 centers throughout China. attention.20 Longer follow-up may determine whether prophylactic
The incidence of sorafenib-related HFSR has been found to vary use of UBC reduces HFSR rates and intensity, increasing drug
in clinical trials of patients with advanced HCC. These differences may exposure and improving therapeutic outcomes. Further studies in
be a result of differences in tumor type and starting dose33 and sex and a blinded, placebo-controlled, randomized setting are warranted.
PS.27 The incidence of any grade HFSR in this study was higher than in In conclusion, prophylactic UBC in patients starting sorafenib for
most other studies of sorafenib4,5,34 but was not highest overall.18 The advanced HCC reduced the incidence and severity of HFSR, de-
disparities between this trial and others may also have been a result of layed the time to its first occurrence, and improved HFSR-
the use of different HFSR grading systems and of increased awareness associated quality of life.
of HFSR by clinicians and patients during treatment with sorafenib.
Importantly, however, the rates of any grade and grade ⱖ 2 HFSR were
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
significantly lower in the UBC plus BSC group than in the BSC-alone OF INTEREST
group, with similar reductions in each grade of HFSR severity. More-
over, UBC significantly extended the median time to first occurrence Although all authors completed the disclosure declaration, the following
of HFSR, decreasing its risk through week 12 by 34%. author(s) and/or an author’s immediate family member(s) indicated a
Although the mechanism underlying sorafenib-induced HFSR is financial or other interest that is relevant to the subject matter under
unknown, factors such as age, sex, ECOG PS, and laboratory values at consideration in this article. Certain relationships marked with a “U” are
baseline have been assessed.27 Multivariable regression analyses indi- those for which no compensation was received; those relationships marked
with a “C” were compensated. For a detailed description of the disclosure
cated that prophylactic use of UBC reduced HFSR incidence and
categories, or for more information about ASCO’s conflict of interest policy,
extended the time to first occurrence. In addition, these analyses please refer to the Author Disclosure Declaration and the Disclosures of
identified patients with abnormal AST as being at higher risk for Potential Conflicts of Interest section in Information for Contributors.
HFSR. Abnormal AST may reflect underlying liver disease in these Employment or Leadership Position: None Consultant or Advisory
patients, which may interfere with liver metabolism of sorafenib and Role: None Stock Ownership: None Honoraria: None Research
enhance its plasma concentration. Although plasma concentrations of Funding: Sheng-Long Ye, Bayer Expert Testimony: None Patents,
sorafenib were not measured in this study, the benefits of prophylactic Royalties, and Licenses: None Other Remuneration: None
UBC were similar in patients with higher (elevated AST) and lower
(normal AST) risk of HFSR. AUTHOR CONTRIBUTIONS
However, UBC had little effect on sorafenib dose reductions,
interruptions, and discontinuations; overall response rate; and disease Conception and design: ZhengGang Ren, WeiPing Zhou,
control rate. This may have been a result of the short duration of the Sheng-Long Ye
trial or of the ability of patients in the BSC-alone group to switch to Provision of study materials or patients: WeiZhu Yang, Sheng-Long Ye
UBC after the development of HFSR. The latter may have masked the Collection and assembly of data: KangShun Zhu, HaiYan Kang,
effects of UBC on sorafenib dose reductions, interruptions, and dis- MinQiang Lu, ZengQiang Qu, LiGong Lu, TianQiang Song, WeiPing
Zhou, Hui Wang, WeiZhu Yang, Xuan Wang
continuations, but not on the rates of any grade and grade ⱖ 2 HFSR Data analysis and interpretation: YongPing Yang, LeHua Shi, YuXian
or time to first occurrence of HFSR. Moreover, UBC improved patient Bai, XiaoFeng Guo
HFSR-associated quality of life during sorafenib treatment, reducing Manuscript writing: All authors
HFSR symptoms and their impact on daily activities. Final approval of manuscript: All authors

tocellular carcinoma model PLC/PRF/5. Cancer Res Treatment of Cancer: EASL-EORTC clinical practice
REFERENCES 66:11851-11858, 2006 guidelines: Management of hepatocellular carci-
4. Llovet JM, Ricci S, Mazzaferro V, et al: noma. J Hepatol 56:908-943, 2012
1. Wilhelm SM, Carter C, Tang L, et al: BAY 43-9006 Sorafenib in advanced hepatocellular carcinoma. 8. Verslype C, Rosmorduc O, Rougier P, et al:
exhibits broad spectrum oral antitumour activity and N Engl J Med 359:378-390, 2008 Hepatocellular carcinoma: ESMO-ESDO clinical
targets the RAF/MEK/ERK pathway and receptor ty- 5. Cheng AL, Kang YK, Chen Z, et al: Efficacy and practice guidelines for diagnosis, treatment and
rosine kinases involved in tumor progression and angio- safety of sorafenib in patients in the Asia-Pacific region follow-up. Ann Oncol 56:vii41-vii48, 2012 (suppl 7)
genesis. Cancer Res 64:7099-7109, 2004 with advanced hepatocellular carcinoma: A phase III 9. Chinese Anti-Cancer Association Society of Liver
2. Wilhelm SM, Adnane L, Newell P, et al: randomised, double-blind, placebo-controlled trial. Lancet Cancer, Chinese Society of Clinical Oncology, Chinese
Preclinical overview of sorafenib, a multikinase in- Oncol 10:25-34, 2009 Society of Hepatology Liver Cancer Study Group: The
hibitor that targets both Raf and VEGF and PDGF 6. Bruix J, Sherman M, American Association expert consensus on the treatment standards for hepa-
receptor tyrosine kinase signaling. Mol Cancer Ther for the Study of Liver Diseases: Management of tocellular carcinoma. Dig Dis Endosc 3:40-51, 2009
7:3129-3140, 2008 hepatocellular carcinoma: An update. Hepatology 10. Ministry of Health of the People’s Republic of
3. Liu L, Cao Y, Chen C, et al: Sorafenib blocks 53:1020-1022, 2011 China: The diagnosis and treatment guideline of
the RAF/MEK/ERK pathway, inhibits tumor angio- 7. European Association for the Study of the primary liver cancer (version 2011). Chin Clin Oncol
genesis, and induces tumor cell apoptosis in hepa- Liver, European Organisation for Research and 6:929-946, 2011

www.jco.org © 2015 by American Society of Clinical Oncology 899

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ren et al

11. Song P, Tobe RG, Inagaki Y, et al: The man- 19. Porta C, Paglino C, Imarisio I, et al: Uncovering 27. Dranitsaris G, Vincent MD, Yu J, et al: Devel-
agement of hepatocellular carcinoma around the Pandora’s vase: The growing problem of new toxic- opment and validation of a prediction index for
world: A comparison of guidelines from 2001 to ities from novel anticancer agents—The case of hand-foot skin reaction in cancer patients receiving
2011. Liver Int 32:1053-1063, 2012 sorafenib and sunitinib. Clin Exp Med 7:127-134, sorafenib. Ann Oncol 23:2103-2108, 2012
12. Robert C, Soria JC, Spatz A, et al: Cutaneous 2007 28. Manchen E, Robert C, Porta C: Management
side-effects of kinase inhibitors and blocking anti- 20. Autier J, Escudier B, Wechsler J, et al: Pro- of tyrosine kinase inhibitor-induced hand-foot skin
bodies. Lancet Oncol 6:491-500, 2005 spective study of the cutaneous adverse effects of reaction: Viewpoints from the medical oncologist,
13. Beldner M, Jacobson M, Burges GE, et al: sorafenib, a novel multikinase inhibitor. Arch Derma- dermatologist, and oncology nurse. J Support Oncol
Localized palmar-plantar epidermal hyperplasia: A tol 144:886-892, 2008 9:13-23, 20113
previously undefined dermatologic toxicity to 21. Nardone B, Hensley JR, Kulik L, et al: The 29. Robert C, Mateus C, Spatz A, et al: Dermatologic
sorafenib. Oncologist 12:1178-1182, 2007 effect of hand-foot skin reaction associated with the symptoms associated with the multikinase inhibitor
14. Lacouture ME, Reilly LM, Gerami P, et al: multikinase inhibitors sorafenib and sunitinib on sorafenib. J Am Acad Dermatol 50:299-305, 2009
health-related quality of life. J Drugs Dermatol 11:
Hand foot skin reaction in cancer patients treated 30. Wood LS, Lemont H, Jatoi A, et al: Practical
e61-e65, 2012
with the multikinase inhibitors sorafenib and considerations in the management of hand-foot skin
22. Hagemann I, Proksch E: Topical treatment by
sunitinib. Ann Oncol 19:1955-1961, 2008 reaction caused by multikinase inhibitors. Commu-
urea reduces epidermal hyperproliferation and in-
15. Yang CH, Lin WC, Chuang CK, et al: Hand-foot nity Oncol 7:23-29, 2010
duces differentiation in psoriasis. Acta Derm Vene-
skin reaction in patients treated with sorafenib: A 31. Duvic M, Asano AT, Hager C, et al: The
reol 76:353-356, 1996
clinicopathological study of cutaneous manifesta- pathogenesis of psoriasis and the mechanism of
23. Lacouture ME, Wu S, Robert C, et al: Evolving
tions due to multitargeted kinase inhibitor therapy. action of tazarotene. J Am Acad Dermatol 39:S129-
strategies for the management of hand-foot skin
Br J Dermatol 158:592-596, 2008 reaction associated with the multitargeted kinase S133, 1998
16. Zhang L, Zhou Q, Ma L, et al: Meta-analysis of inhibitors sorafenib and sunitinib. Oncologist 13: 32. Scheithauer W, McKendrick J, Begbie S, et al:
dermatological toxicities associated with sorafenib. 1001-1011, 2008 Oral capecitabine as an alternative to i.v. 5-fluorouracil-
Clin Exp Dermatol 36:344-350, 2011 24. Anderson R, Jatoi A, Robert C, et al: Search based adjuvant therapy for colon cancer: Safety results of
17. Ogasawara S, Kanai F, Obi S, et al: Safety and for evidence-based approaches for the prevention a randomized, phase III trial. Ann Oncol 14:1735-1743,
tolerance of sorafenib in Japanese patients with and palliation of hand-foot skin reaction (HFSR) 2003
advanced hepatocellular carcinoma. Hepatol Int caused by the multikinase inhibitors (MKIs). Oncol- 33. Chu D, Lacouture ME, Fillos T, et al: Risk of
5:850-856, 2011 ogist 14:291-302, 2009 hand-foot skin reaction with sorafenib: A systematic
18. Kudo M, Imanaka K, Chida N, et al: Phase III 25. Anderson RT, Rajagopalan R: Development and review and meta-analysis. Acta Oncol 47:176-186, 2008
study of sorafenib after transarterial chemoemboli- validation of a quality of life instrument for cutaneous 34. Abou-Alfa GK, Schwartz L, Ricci S, et al:
sation in Japanese and Korean patients with unre- diseases. J Am Acad Dermatol 37:41-50, 1997 Phase II study of sorafenib in patients with ad-
sectable hepatocellular carcinoma. Eur J Cancer 26. SAS Institute: SAS/STAT(R) User’s Guide, vanced hepatocellular carcinoma. J Clin Oncol 24:
47:2117-2127, 2011 Version 8. Cary, NC, SAS Institute, 1999 4293-4300, 2006

■ ■ ■

900 © 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Prophylactic Urea-Based Cream for Sorafenib HFSR in Advanced HCC

Acknowledgment
We acknowledge the contributions of all the other investigators (Appendix Table A1) and the patients in this trial and of BelMed Professional
Resources (New Rochelle, NY) for editorial support.

Appendix

Table A1. Investigators and Institutions Enrolling Patients Onto This Trial

Name of Investigator Institution


YuXian Bai Heilongjiang Provincial Cancer Hospital
JianQiang Cai Cancer Institute and Hospital Chinese Academy of Medical Sciences
LiPing Cao The Second Affiliated Hospital of Zhejiang University School of Medical
Hong Chen The Hospital of Armed Police Force
HuanWei Chen The First People’s Hospital of Fushan
Hui Chen Beijing Cancer Hospital
Jun Chen Jiangsu Cancer Hospital
PeiFeng Chen Zhejiang Provincial Hospital of Traditional Chinese Medicine
YanLing Chen Fujian Medical University Union Hospital
YuTang Chen Zhejiang Cancer Hospital
DianYuan Cui First Affiliated Hospital of Dalian Medical University
Sheng Dong ShangHai Changzheng Hospital
YongChong Dou Shengzhen People’s Hospital
Wen Fang Zhongda Hospital Southeast University
GuoSheng Feng GuangXi People’s Hospital
YuJia Gao Liaoning Cancer Hospital and Institute
Sheng Guan The First Affiliated Hospital of Zhengzhou University
WuHua Guo The Second Affiliated Hospital to Nanchang University
Zhi Guo Tianjin Medical University Cancer Institute and Hospital
ChunYi Hao Beijing Cancer Hospital
XiangDong Hua Liaoning Cancer Hospital and Institute
XingYu Huang Shanghai Sixth People’s Hospital
Feng Huo General Hospital of Guanzhou Military Command of People’s Liberation Army
ZhiXiang Jian Guangdong General Hospital
JiaJi Jiang The First Affiliated Hospital of Fujian Medical University
TianJun Jiang People’s Liberation Army 302 Hospital
HaiYan Kang People’s Liberation Army 301 Hospital
Bing Li The Second Affiliated Hospital of Zhejiang University School of Medical
Bing Li The First Affiliated Hospital Xiamen University
Bo Li West China Hospital Sichuan University
HaiLiang Li Henan Provincial Cancer Hospital
Huai Li Cancer Institute and Hospital Chinese Academy of Medical Sciences
WenTao Li Fudan University Shanghai Cancer Center
XiaoWu Li Southwest Hospital
Jun Liang The Affiliated Hospital of Qingdao University
LiJian Liang The First Affiliated Hospital Sun Yat-sen University
ZhengYin Liao West China Hospital Sichuan University
HaiLan Lin Fujian Provincial Cancer Hospital
FengYong Liu People’s Liberation Army 301 Hospital
JingFeng Liu The First Affiliated Hospital of Fujian Medical University
ZhaoYu Liu Shengjing Hospital of China Medical University
Zhe Liu People’s Liberation Army 301 Hospital
ZhenWen Liu People’s Liberation Army 302 Hospital
GuoLiang Lou Shanghai Changhai Hospital
LiGong Lu Guangdong General Hospital
MinQiang Lu The Third Affiliated Hospital Sun Yat-sen University
AiYing Ma People’s Liberation Army 455 Hospital
Rui Ma Liaoning Cancer Hospital and Institute
YiLong Ma Guangxi Medical School Affiliated Cancer Hospital
XiongYing Miao The Second Xiangya Hospital of Central South University
Wei Mou Southwest Hospital
(continued on following page)

www.jco.org © 2015 by American Society of Clinical Oncology

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
Ren et al

Table A1. Investigators and Institutions Enrolling Patients Onto This Trial (continued)

Name of Investigator Institution


Lei Nie Hubei Cancer Hospital
YuDong Qiu Nanjing Drum Tower Hospital
ZenQiang Qu Eastern Hepatobiliary Surgery Hospital
JiaLing Shen Renji Hospital Affiliated to ShangHai Jiaotong University School of Medicine
LiJun Sheng Shangdong Cancer Hospital
LeHua Shi Eastern Hepatobiliary Surgery Hospital
Zheng Shi The First Affiliated Hospital of Fujian Medical University
GuangMing Shu Peking University People’s Hospital
YiJin Shu Zhejiang Provincial Hospital of Traditional Chinese Medicine
HongLi Song Tianjing First Central Hospital
TianQiang Song Tianjin Medical University Cancer Institute and Hospital
LiYing Sun Tianjing First Central Hospital
Tao Sun Liaoning Cancer Hospital and Institute
ZhenHua Tu The First Affiliated Hospital of Zhejiang University School of Medical
Hui Wang Jilin Provincial Cancer Hospital
TongShan Wang Jiangsu Province Hospital
Xuan Wang People’s Liberation Army 81 Hospital
YaoDong Wang Fujiang Provincial Hospital
JinShu Wu Hunan Provincial People’s Hospital
BaoCai Xing Beijing Cancer Hospital
AiBing Xu Nantong Tumor Hospital
GuoHui Xu The Second People’s Hospital of Sichuan
JianMing Xu People’s Liberation Army 307 Hospital
LinFeng Xu The Second Affiliated Hospital Sun Yat-sen University
WeiZhu Yang Fujian Medical University Union Hospital
WuWei Yang People’s Liberation Army 307 Hospital
YongPing Yang People’s Liberation Army 302 Hospital
Hua Ye The Second Affiliated Hospital Sun Yat-sen University
ShengLong Ye Zhongshan Hospital Fudan University
Tao Yin Hubei Cancer Hospital
JiaSheng Zheng Beijing YouAn Hospital Capital Medical University
SuiLiang Zhang Eastern Hepatobiliary Surgery Hospital
XiPing Zhang Zhejiang Cancer Hospital
GuangWen Zhou Ruijin Hospital Affiliated to ShangHai Jiaotong University School of Medicine
Shi Zhou Affiliated Hospital of Guiyang Medical College
WeiPing Zhou Eastern Hepatobiliary Surgery Hospital
XinWen Zhou Jiangxi Cancer Hospital
Yun Zhou Henan Provincial People’s Hospital
KangShun Zhu The Third Affiliated Hospital Sun Yat-sen University
Li Zhuang The First Affiliated Hospital of Zhejiang University School of Medical

Table A2. Grading of Hand-Foot Skin Reaction


Grade Description
1 Numbness, dysesthesia/paresthesia, tingling, painless swelling or erythema of the hands and/or feet and/or discomfort, which does not
disrupt normal activities
2 Painful erythema and swelling of the hands and/or feet and/or discomfort affecting the patient’s activities
3 Moist desquamation, ulceration, blistering, or severe pain of the hands and/or feet and/or severe discomfort that causes the patient to
be unable to work or perform activities of daily living

© 2015 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY

Downloaded from ascopubs.org by 200.92.202.106 on August 19, 2022 from 200.092.202.106


Copyright © 2022 American Society of Clinical Oncology. All rights reserved.
ORIGINAL CONTRIBUTION

Effect of Duloxetine on Pain, Function,


and Quality of Life Among Patients
With Chemotherapy-Induced
Painful Peripheral Neuropathy
A Randomized Clinical Trial
Ellen M. Lavoie Smith, PhD Importance There are no known effective treatments for painful chemotherapy-
Herbert Pang, PhD induced peripheral neuropathy.
Constance Cirrincione, MS Objective To determine the effect of duloxetine, 60 mg daily, on average pain severity.
Stewart Fleishman, MD Design, Setting, and Patients Randomized, double-blind, placebo-controlled cross-
Electra D. Paskett, PhD over trial at 8 National Cancer Institute (NCI)–funded cooperative research networks
that enrolled 231 patients who were 25 years or older being treated at community
Tim Ahles, PhD and academic settings between April 2008 and March 2011. Study follow-up was com-
Linda R. Bressler, PharmD pleted July 2012. Stratified by chemotherapeutic drug and comorbid pain risk, pa-
tients were randomized to receive either duloxetine followed by placebo or placebo
Camilo E. Fadul, MD followed by duloxetine. Eligibility required that patients have grade 1 or higher sen-
Chetaye Knox, BS sory neuropathy according to the NCI Common Terminology Criteria for Adverse Events
Nguyet Le-Lindqwister, MD and at least 4 on a scale of 0 to 10, representing average chemotherapy-induced pain,
after paclitaxel, other taxane, or oxaliplatin treatment.
Paul B. Gilman, MD
Interventions The initial treatment consisted of taking 1 capsule daily of either 30
Charles L. Shapiro, MD mg of duloxetine or placebo for the first week and 2 capsules of either 30 mg of du-
for the Alliance for Clinical Trials in loxetine or placebo daily for 4 additional weeks.
Oncology Main Outcome Measures The primary hypothesis was that duloxetine would be more
effective than placebo in decreasing chemotherapy-induced peripheral neuropathic pain.

A
PPROXIMATELY 20% TO 40% OF Pain severity was assessed using the Brief Pain Inventory-Short Form “average pain” item
patients with cancer who re- with 0 representing no pain and 10 representing as bad as can be imagined.
ceiveneurotoxicchemotherapy Results Individuals receiving duloxetine as their initial 5-week treatment reported a mean
(eg, taxanes, platinums, vinca decrease in average pain of 1.06 (95% CI, 0.72-1.40) vs 0.34 (95% CI, 0.01-0.66) among
alkaloids, bortezomib) will develop pain- those who received placebo (P=.003; effect size, 0.513). The observed mean difference
ful chemotherapy-induced peripheral in the average pain score between duloxetine and placebo was 0.73 (95% CI, 0.26-
neuropathy.1-3 Painful chemotherapy- 1.20). Fifty-nine percent of those initially receiving duloxetine vs 38% of those initially
induced neuropathy can persist from receiving placebo reported decreased pain of any amount.
months to years beyond chemotherapy Conclusion and Relevance Among patients with painful chemotherapy-induced
completion,causingsignificantchallenges peripheral neuropathy, the use of duloxetine compared with placebo for 5 weeks re-
for cancer survivors due to its negative in- sulted in a greater reduction in pain.
fluence on function and quality of life Trial Registration clinicaltrials.gov Identifier: NCT00489411
(QOL).4-8 Chemotherapy–induced pe- JAMA. 2013;309(13):1359-1367 www.jama.com
ripheral neuropathy is difficult to man-
age,andmostrandomizedcontrolledtrials testing a variety of drugs with diverse serotonin and norepinephrine are key
mechanisms of action revealed no effec- neurotransmitters that suppress trans-
CME available online at tive treatment.9 mission of painful peripheral stimuli by
www.jamanetworkcme.com There is mounting evidence that
and questions on p 1412.
serotonin and norepinephrine dual Author Affiliations are listed at the end of this article.
Author Video Interview available at reuptake inhibitors are effective in treat- Corresponding Author: Ellen M. Lavoie Smith, PhD,
www.jama.com. University of Michigan School of Nursing, Room 2151,
ing neuropathy-related pain.10 Both Ann Arbor, MI 48109 (ellenls@umich.edu).

©2013 American Medical Association. All rights reserved. JAMA, April 3, 2013—Vol 309, No. 13 1359

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

inhibiting input to the spinal dorsal scale, and reported 4 on a 10-point Intervention
horn neurons.11 Several phase 3 stud- scale, average neuropathic pain, for 3 Eligible patients were randomized
ies show that duloxetine is an effec- or more months after completing che- using a 1:1 allocation ratio to either
tive treatment for painful diabetic neu- motherapy. Patients with any cancer di- group A or group B. In this crossover
ropathy.12-15 Based on these trials, our agnosis or stage were potentially eli- design, group A received 60 mg of
hypothesis was that duloxetine would gible. To diminish the likelihood that duloxetine daily during the initial
ameliorate chemotherapy-induced pe- symptoms would spontaneously re- treatment period and placebo at
ripheral neuropathic pain as well. A ran- solve over the course of the study, crossover period. Group B received
domized phase 3 trial was conducted efficacy data were obtained over 5 placebo as initial treatment and
to test this hypothesis. weeks. duloxetine as the crossover treat-
Initially, patients who had received ment. Randomization, provided by
METHODS paclitaxel or oxaliplatin could partici- the CALGB/Alliance Statistical Cen-
STUDY DESIGN pate, but eligibility was later ex- ter, was stratified by neurotoxic drug
The Cancer and Leukemia Group B panded to allow prior treatment with class (taxanes vs platinums) and by
(CALGB/Alliance) conducted a ran- single-agent docetaxel, nanoparticle al- pain risk (high risk vs no risk). A
domized phase 3 double-blind, placebo- bumin–bound paclitaxel, or cisplatin. computer-generated kit number was
controlled crossover trial to assess Prior or ongoing treatment with other used to order the blinded study
whether 60 mg of duloxetine taken neurotoxic chemotherapeutic agents drug from a distribution center. Drug
orally once daily decreases the sever- was not allowed. Participants with a labels were applied to the capsule
ity of chemotherapy-induced periph- documented medical history of neu- bottles at the distribution center
eral neuropathy (CALGB-170601, ropathy from any type of nerve com- before being mailed to study sites;
NCT00489411). The primary hypoth- pression (eg, carpal or tarsal tunnel syn- thus, all patients and personnel were
esis was that duloxetine would be more drome, radiculopathy, spinal stenosis, blinded to the treatment assignment.
effective than placebo in decreasing the brachial plexopathy), leptomeningeal The initial (weeks 1-5) and cross-
average pain score after a 5-week treat- carcinomatosis, severe depression, sui- over (weeks 8-12) treatment periods
ment period. Secondary aims were to cidal ideation, bipolar disease, alcohol each consisted of receiving 1 capsule of
assess duloxetine’s effect on QOL and abuse, a major eating disorder, and either placebo or 30 mg of duloxetine
function and on adverse events. The markedly abnormal renal or liver func- for the first week and 2 capsules of
study was approved by each site’s in- tion tests were ineligible. either placebo or 30 mg of duloxetine
stitutional review board, and partici- Despite scant evidence supporting for 4 weeks. The initial 5-week period
pants provided signed informed con- the association between certain comor- was followed by a 2-week washout pe-
sent. Enrollment occurred between bid illnesses and the risk of develop- riod for a total study duration of 14
April 2008 and March 2011. Study fol- ing severe pain,9,16 patients with diabe- weeks (FIGURE 1).
low-up was completed July 2012. tes mellitus and peripheral vascular
disease, whose pain was thought to be Data Collection and Instruments
Patients from chemotherapy-induced periph- Patient-reported pain severity and func-
Using the National Cancer Institute’s eral neuropathy, were eligible but were tional interference was assessed weekly
(NCI’s) Clinical Trials Support Unit defined as high risk. We controlled for using the well-validated Brief Pain In-
mechanism to facilitate accrual, par- comorbid illness as a potential con- ventory Short Form (BPI-SF).17-19 The
ticipants were recruited from 8 multi- founder by assigning equal numbers of BPI-SF contains 4 items assessing av-
site NCI-funded cooperative research high-risk patients to each treatment erage, worst, least, and immediate pain
networks, resulting in a geographi- group. Concurrent use of other drugs severity in the last 24 hours. Pain se-
cally diverse population of patients dis- known to influence serotonin levels was verity items are scored using an 11-
tributed throughout the United States. not allowed. Concomitant use of se- point numeric rating scale (0, no pain;
The diagnosis of chemotherapy- lected analgesics was allowed (eg, opi- 10, pain as bad as you can imagine). The
induced peripheral neuropathy was de- oids, acetaminophen, aspirin, nonste- BPI-SF worse pain severity item has
termined based on symptom history, roidal anti-inflammatory drugs), but been shown to be reliable and valid for
loss of deep tendon reflexes, or the pres- only patients receiving stable doses in use as a single item.20 However, we
ence of symmetrical stocking-glove the 2 weeks before randomization could chose average pain severity as our
numbness or paresthesias beginning af- participate: (1) no new analgesics were primary outcome measure based on
ter neurotoxic chemotherapy. Eligible added, (2) no analgesics were discon- recommendations from the Initiative
patients were 25 years or older, had at tinued, and (3) the weekly 24-hour total on Methods, Measurement, and
least grade 1 sensory pain based on the analgesic dose did not fluctuate up or Pain Assessment in Clinical Trials
NCI Common Terminology Criteria for down by more than 10% in the 2 weeks (IMMPACT).21 In addition, average pain
Adverse Events version 3.0 grading before study registration. has been defined as the primary out-
1360 JAMA, April 3, 2013—Vol 309, No. 13 ©2013 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

come measure in numerous dulox-


Figure 1. Patient Flow Chart of Initial and Crossover Periods a
etine phase 3 studies involving pa-
tients with peripheral and central 231 Patients enrolled
neuropathic pain conditions due to dia-
betic- and oxaliplatin-induced neu-
231 Randomized
ropathy, fibromyalgia, and osteoarthri-
tis13-15,22-30 and thereby was chosen to
facilitate comparison of our findings Group A Group B
115 Randomized to receive duloxetine followed by 116 Randomized to receive placebo followed by
across similar studies. The minimal clini- crossover to placebo crossover to duloxetine
cally important difference in pain sever- 109 Received duloxetine as randomized
6 Withdrew consent prior to receiving the
111 Received placebo as randomized
5 Withdrew consent prior to receiving the
ity was defined for the current study as intervention intervention
a 0.98 difference in mean average pain
severity between the duloxetine and 88 Completed initial intervention 99 Completed initial intervention
21 Discontinued duloxetine early 12 Discontinued placebo early
placebo groups. Using an accepted 12 Toxic effects 1 Toxic effects
5 Withdrew consent 6 Withdrew consent
method for assessing the influence of 1 Other 2 Disease progression
pain on function,13-15,26-29 7 BPI-SF items 3 Unknown 3 Unknown

were used to quantify the degree to


which pain interfered with daily activi- 87 Included in primary analysis
1 Excluded (incomplete data)
94 Included in primary analysis
5 Excluded (incomplete data)
ties or function (0, does not interfere;
10, completely interferes). The 7 items 85 Crossed over and received placebo 93 Crossed over and received duloxetine
were summed to obtain a total inter-
ference score. Patient-reported QOL 74 Completed crossover intervention 81 Completed crossover intervention
11 Discontinued intervention early 12 Discontinued intervention early
was assessed using the Functional As- 2 Toxic effects 5 Toxic effects
sessment of Cancer Treatment, Gyne- 4 Withdrew consent
1 Received alternate treatment
2 Withdrew consent
2 Disease progression
cologic Oncology Group Neurotoxic- 4 Other 3 Other

ity (FACT/GOG-Ntx) subscale on day


1 of weeks 1, 6, 8, and 13. The assess- 67 Included in crossover analysis
7 Excluded (incomplete data)
74 Included in crossover analysis
7 Excluded (incomplete data)
ment’s strong psychometric proper-
ties have been previously demon- a The number screened and the number offered participation but declined was not captured, nor were the

strated.31-33 The instrument contains 11 reasons captured for why patients did not participate in the crossover treatment period.
questions assessing numbness, tin-
gling, and discomfort in the hands or weakness and sensory alterations in- diabetic research.30 We calculated the
feet; difficulty hearing; tinnitus; joint terfering with function; 3, weakness and proportion of patients experiencing any
pain or muscle cramps; weakness; or sensory alterations interfering with ac- decrease in pain. Also, using another ac-
trouble walking, buttoning buttons, or tivities of daily living or requiring brac- cepted approach for assessing clinical
feeling small shapes when placed in the ing or assistive devices; and 4, life significance, 13-15,22-24,26,39-41 we con-
hand. Items are scored from 0 to 4 (0, threatening, paralysis, or disabling). ducted an exploratory responder analy-
not at all; 4, very much) and summed sis based on the proportion of patients
(total score range, 0-44).33 Because there Statistical Analyses in both groups who experienced a 30%
are no published data defining a cut Statistical analyses were performed by (or 50%) decrease in pain severity. Rela-
point for determining a clinically im- CALGB/Alliance statisticians. The pri- tive risk-benefit was the proportion of
portant change in the score, we de- mary study end point was the change patients with 30% (or 50%) pain re-
fined a 2- to 3-point change as a clini- from start to end of the initial treat- duction in the duloxetine group rela-
cally meaningful improvement in QOL ment period (week 1 to week 5, mea- tive to that in the placebo group. It was
per published recommendations spe- sured on day 1 of weeks 1 and 6) in av- calculated from contingency tables. We
cific to similar measures.34-37 erage pain based on the BPI-SF average also conducted an exploratory sub-
Using the NCI CTCAE version 3.0, pain severity item. The comparison of group analyses based on chemo-
adverse events were reported weekly interest was the difference between the therapy class.
and graded on a 0 to 4 scale (0, nor- 2 treatment groups in pain change. Secondary end points were change in
mal; 4, life-threatening) 38 as was base- With 232 patients (assuming 20% at- chemotherapy-induced peripheral neu-
line sensory chemotherapy-induced pe- trition), the study had 90% power (2- ropathy–related QOL, measured by the
ripheral neuropathy (0, normal; 1, sided ␣ of .05) to detect a 0.98-point total score of the FACT/GOG-Ntx, and
asymptomatic, weakness on physical change between the 2 groups assum- degree of pain-related functional inter-
examination, loss of reflexes, or pares- ing a standard error of 0.31. The tar- ference based on the BPI-SF interfer-
thesias not interfering with function; 2, get difference of 0.98 is consistent with ence score. Changes attributable to ini-
©2013 American Medical Association. All rights reserved. JAMA, April 3, 2013—Vol 309, No. 13 1361

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

sion 9.2 (SAS Institute Inc). Data qual-


Table. Patient Demographics and Baseline Features
ity was ensured by review of data by the
No. (%)
of Participants CALGB/Alliance Statistical Center and
by the study chairperson. The study un-
Group A Group B Total P
Characteristics (n = 109) (n = 111) (n = 220) Value derwent standard biannual monitor-
Demographics ing by the CALGB/Alliance data and
Age, y safety monitoring board with 1 formal
30-39 2 (2) 1 (1) 3 (1)
40-49 16 (15) 23 (21) 39 (18)
interim efficacy analysis resulting in
50-59 43 (39) 39 (35) 82 (37) study continuation.
.70 a
60-69 30 (28) 29 (26) 59 (27)
RESULTS
ⱖ70 18 (17) 19 (17) 37 (17)
Mean (SD) 60 (10.4) 59 (10.6) 59 (10.5) Patients
Sex Patient disposition for the initial treat-
Men 38 (35) 44 (40) 82 (37)
.46 ment period is illustrated in Figure 1.
Women 71 (65) 67 (60) 138 (63)
Of the 231 patients recruited to the
Race
White 91 (83) 87 (78) 178 (81) study, 115 were allocated to group A
Black 14 (13) 17 (15) 31 (14) .50 (duloxetine first, placebo second), and
Other 4 (4) 5 (5) 9 (4) 116 to group B (placebo first, dulox-
Not reported 0 (0) 2 (2) 2 (1) etine second). Eleven patients never re-
Stratification factors ceived treatment, leaving 220 treated
Neurotoxic agent
Paclitaxel 44 (40) 43 (39) 87 (40)
patients. The dropout rate due to ad-
Oxaliplatin 63 (58) 66 (59) 129 (59) N/A
verse events in the duloxetine-first
Other taxane 2 (2) 2 (2) 4 (2) group was 11% vs 1% in the placebo-
Other platinum 0 (0) 0 (0) 0 (0) first group (P⬍.001). Despite using an
High risk of CIPN intent-to-treat analysis approach, 6 pa-
No 46 (42) 51 (46) 97 (44) tients (1 in group A; 5 in group B) dur-
N/A
Yes 63 (58) 60 (54) 123 (56)
ing the initial treatment period were ex-
Disease-related features
Primary disease cluded from the primary analysis
Breast 41 (38) 42 (38) 83 (38) because they provided no data at all.
GI 62 (57) 62 (56) 124 (56) This resulted in a 19% dropout rate.
Both breast & GI tract 0 (0) 1 (1) 1 (0) .99 b Patient characteristics are de-
Genitourinary 4 (4) 4 (4) 8 (3) scribed in the TABLE. Both groups were
Other 2 (2) 1 (1) 3 (1) similar at baseline, except for the mean
Missing 0 (0) 1 (1) 1 (0)
(SD) pain score of 6.1 (1.7) in group
(continued)
A, duloxetine first, and 5.6 (1.6) in
group B, placebo first (P=.02).
tial treatment were defined as the equations were used to determine
difference between the week 1 and week whether there was a treatment effect Pain (Primary Outcome)
5 scores (measured on day 1 of weeks when combining data from both the ini- At the end of the initial treatment pe-
1 and 6). Changes for crossover treat- tial and crossover periods. Multiple riod, patients in the duloxetine-first
ment used week 8 and week 12 scores imputation and pattern-mixture model group reported a larger decrease in av-
(measured on day 1 of weeks 8 and 13). were used to evaluate the pattern and erage pain (mean change score, 1.06;
To test for a group effect during the potential influence of missing values for 95% CI, 0.72-1.40) than those in the
initial treatment period on the pri- the primary end point (eAppendix placebo-first group (mean change score,
mary and secondary end points, we used available at www.http://www.jama 0.34; 95% CI, 0.01-0.66; P = .003;
3 separate models of analysis of cova- .com). To univariately compare treat- FIGURE 2). The effect size attributed to
riance, each stratified by neurotoxic ment groups, the Wilcoxon rank test duloxetine was moderately large at
agent and risk of painful chemotherapy- was used for continuous variables and 0.513.42 The observed mean differ-
induced peripheral neuropathy, and the ␹2 test for proportions. The 95% ence in the average pain score between
including the baseline measure of the confidence intervals for proportions the duloxetine-first and placebo-first
corresponding end point. Least square used exact binomial methods. Analy- groups was 0.73 (95% CI, 0.26-1.20).
means and their 95% confidence inter- ses included only patients who began Results of the sensitivity analysis tak-
vals were taken from analysis of cova- protocol therapy. Statistical analyses ing missing data into consideration are
riance models. Additionally, for the pri- with 2-sided significance threshold of consistent with the primary findings of
mary end point, generalized estimating P⬍.05 were performed using SAS ver- the trial (eTables 1 and 2 available at
1362 JAMA, April 3, 2013—Vol 309, No. 13 ©2013 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

http://www.jama.com): multiple im-


Table. Patient Demographics and Baseline Features (continued)
putation (P = .002) and pattern-
No. (%)
mixture model control–based imputa- of Participants
tion (P=.004). The primary analysis P Group A Group B Total P
value lies between the slightly more lib- Characteristics (n = 109) (n = 111) (n = 220) Value
eral multiple imputation approach and Disease-related features
Disease stage
the more conservative pattern- Early, I-II 38 (44) 38 (41) 76 (35)
mixture model. A post hoc power cal- III 34 (39) 42 (45) 76 (35) .69
culation is provided in the eAppendix. Metastatic 14 (16) 12 (13) 26 (12)
Of the patients treated with dulox- Missing 1 (1) 2 (2) 3 (1)
etine first, 59% reported any decrease Concurrent medications use
in pain vs 38% of patients treated with No 68 (62) 58 (52) 126 (57)
placebo first. Thirty percent of dulox- Yes 31 (28) 43 (39) 74 (34) .10
etine-treated patients reported no Missing 10 (9) 10 (9) 20 (9)
change in pain and 10% reported in- Performance status
0 60 (55) 60 (54) 120 (55)
creased pain. Based on the explor-
1 44 (40) 47 (42) 91 (41)
atory responder analysis, the propor- .78
2 4 (4) 2 (2) 6 (3)
tion of patients achieving various levels
3 1 (1) 1 (1) 2 (1)
of pain reduction is illustrated in
Missing 0 (0) 1 (1) 1 (0)
FIGURE 3. Compared with placebo, the
Pretreatment sensory neuropathy grade
relative risk of experiencing a 30% pain 1, Asymptomatic 1 (1) 2 (2) 3 (1)
reduction with duloxetine was 1.96 2, Moderate 77 (71) 84 (76) 161 (73)
(95% CI, 1.15-3.35) and experiencing .47
3, Severe 31 (28) 24 (22) 55 (25)
a 50% pain reduction was 2.43 (95% CI, 4, Life-threatening 0 (0) 0 (0) 0 (0)
1.11-5.30; eTable 3). Missing 0 (0) 1 (1) 1 (0)
Although the study was powered to Pretreatment pain score
⬍4 2 (2) 2 (2) 4 (2)
detect differences between treatments
4-5 44 (40) 58 (52) 102 (46)
as main effects only, in exploratory
analyses, we examined the potential in- 6-7 39 (36) 35 (32) 74 (34) .02 a
8-10 24 (22) 15 (14) 39 (18)
teraction between treatment group and
Missing 0 (0) 1 (1) 1 (0)
chemotherapy class. Results sug-
Mean (SD) 6.1 (1.7) 5.6 (1.6) 5.8 (1.7)
gested that patients who received plati-
Abbreviations: CIPN, chemotherapy-induced peripheral neuropathy; GI, gastrointestinal; N/A, not applicable (comparative
nums (oxaliplatin) experienced more testing is not applicable because these are stratification variables).
a Tested as a continuous variable.
benefit from duloxetine than those who b When testing for differences between groups based on primary disease, the groups were collapsed into 3 categories
received taxanes (P = .13). The ob- (breast, gastrointestinal, or other).

Figure 2. Duloxetine and Placebo Effects on Average Pain Severity During the Initial and Crossover Treatment Periods

Initial treatment period Crossover treatment period


6.5 6.5
Duloxetine first (group A) Placebo second (group A)
6.0 Placebo first (group B) 6.0 Duloxetine second (group B)
Mean Pain Score

Mean Pain Score

5.5 5.5

5.0 5.0

4.5 4.5

4.0 4.0

3.5 3.5

3.0 3.0
1 2 3 4 5 6 1 2 3 4 5 6
Week (Washout) Week
No. patients No. patients
Duloxetine first (group A) 87 86 86 87 85 87 Placebo second (group A) 67 65 65 66 63 67
Placebo first (group B) 94 92 91 91 91 94 Duloxetine second (group B) 74 68 71 69 66 74

The mean average pain score was measured on the first day of each week in the initial and crossover treatments periods. Day 1 of the first week begins the initial
treatment period. Day 1 of week 6 begins the washout period when patients received 1 capsule of duloxetine or placebo. Patients took no drug during week 7. Error
bars represent 95% CIs.

©2013 American Medical Association. All rights reserved. JAMA, April 3, 2013—Vol 309, No. 13 1363

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

2.82) for patients treated with placebo


Figure 3. Percent Decrease in Pain Score Due to Duloxetine vs Placebo
first (P=.03). The mean difference be-
100 tween the 2 groups in mean change
90 score was 1.58 (95% CI, 0.15-3.00;
80 P=.03).
Percentage of Patients

70 Adverse Effects. No hematologic or


60 grade 4 (moderately severe) or 5 (se-
50 vere) adverse events were reported. In
40
the initial treatment period, 16% treated
30
with duloxetine and 27% treated with
20
10
placebo reported grade 2 (mild) and 7%
0
Duloxetine (n = 87)
Placebo (n = 94)
treated with duloxetine and 3% treated
with placebo reported grade 3 (mod-
≥0 ≥10 ≥20 ≥30 ≥40 ≥50 ≥60 ≥70 ≥80 ≥90 100
Pain Reduction From Baseline to End Point erate) nonhematologic adverse events.
of Initial Treatment Period, % Fatigue (7%), insomnia (5%), and nau-
sea (5%) were the most common ad-
The plot shows the proportion of patients achieving various levels of pain reduction at the completion of the
initial treatment period. verse effects reported by patients treated
by duloxetine, whereas somnolence
(8%), insomnia (7%), and fatigue (5%)
served mean difference in platinum- The treatment remained significant were the most common adverse ef-
related average pain score between the (P = .002), whereas the period (P =.55) fects reported by patients treated with
duloxetine and placebo treatment was and carryover (P=.95) effects were not. placebo (eTable 7 available at http:
1.06 (95% CI, 0.48 to 1.63) vs 0.19 The change in mean pain score during //www.jama.com).
(95% CI, ⫺0.61 to 0.98) for patients the crossover treatment period for
treated with taxane. Results of the ex- group A (placebo second) was 0.41 Other Findings
ploratory responder analysis revealed (95% CI, 0.06-0.89) and for group B Nonpainful Symptoms. After the ini-
that compared with placebo, the rela- (duloxetine second) was 1.42 (95% CI, tial treatment period, 41% of patients
tive risk of experiencing a reduction in 0.97-1.87). The mean difference be- treated with duloxetine reported a de-
pain by 30% with duloxetine among pa- tween the 2 groups in mean change crease in numbness and tingling in the
tients treated with platinum was 3.05 score during the crossover period was feet (95% CI, 31%- 52%) vs 23% of pa-
(95% CI, 1.49-6.27) and a reduction by 1.01 (95% CI, 0.36-1.65). tients treated with placebo (95% CI, 15%-
50% was 3.78 (95% CI, 1.32-10.84; 33%). The trend held through the cross-
eTables 4-6 available at http://www Secondary Outcome over period with 41% of patients treated
.jama.com). However, compared with Pain Interference With Daily Function. with duloxetine (95% CI, 31%-53%) vs
placebo, the relative risk of experienc- At the end of the initial treatment pe- 21% of patients treated with placebo
ing a 30% pain reduction with dulox- riod, when compared with placebo, pa- (95% CI, 13%-32%). The proportion of
etine among patients treated with tax- tients treated with duloxetine first re- patients with improved hand numb-
ane was 0.97 (95% CI, 0.41-2.32) and ported a greater decrease in the amount ness and tingling at the end of the ini-
a 50% pain reduction was 1.22 (95% CI, that pain had interfered with daily func- tial treatment period was similar among
0.35-4.18), neither of which was sta- tioning (P=.01; eFigure 1). The change those treated with duloxetine (36%) and
tistically significant. There was no dif- in mean interference score for patients those treated with placebo (34%).
ference in duloxetine efficacy based on treated with duloxetine first was 7.9 Ancillary Analgesics. Compared
risk of developing painful chemo- (95% CI, 5.4-10.5) vs 3.5 (95% CI, 1.1- with group A (duloxetine first), a higher
therapy-induced peripheral neuropathy. 5.9) for patients treated with placebo proportion of those in group B (pla-
We also concurrently evaluated first. The mean difference between the cebo first) were taking concomitant
changes in pain severity during the 2 groups in mean change score was 4.40 medications at the start (43% of pa-
crossover treatment period. After ad- (95% CI, 0.93-7.88). tients in group B vs 31% in group A)
justing for the study stratifiers (che- Quality of Life. Pain-related QOL im- and at the end of the initial treatment
motherapy class and pain risk), there proved to a greater degree for those period (36% in group B vs 29% in group
was a statistically significant treat- treated with duloxetine during the ini- A).Twenty-seven percent of patients
ment effect on change in pain score tial treatment than for those treated with who started out taking concomitant
(P⬍ .001), but an order effect was not placebo. The mean change in the FACT/ medications in the group A discontin-
significant (P = .43). An additional GOG-Ntx total score was 2.44 (95% CI, ued all medications by the end of the
generalized estimating equation model 0.43-4.45) for patients treated with du- initial treatment period compared with
with carryover effect was performed. loxetine first vs 0.87 (95% CI, 1.09- 19% of patients in group B.
1364 JAMA, April 3, 2013—Vol 309, No. 13 ©2013 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

COMMENT
Figure 4. Comparison of the Mean Differences in Average Pain Scores Across Duloxetine
Treatment of painful chemotherapy- Chronic Pain Studies
induced peripheral neuropathy contin-
ues to be a challenge because most drugs No. of Mean Difference in Mean Favors Favors
Source Patients Pain Scores (95% CI) Placebo Duloxetine
tested to date have fallen short of pro- Goldstein et al,13 2005 176 0.98 (0.37 to 1.60)
viding adequate pain relief.43-47 To our Russell et al,30 2008 294 0.60 (0.05 to 1.16)
knowledge, the current study is the first Chappell et al,26 2011 225 0.79 (0.26 to 1.32)
Current study 181 0.73 (0.26 to 1.20)
large phase 3 trial to elucidate an effec-
Neurotoxic agents, current study
tive intervention for painful chemo- Platinums 106 1.06 (0.48 to 1.63)
therapy-induce peripheral neuropathy Taxanes 75 0.19 (–0.61 to 0.98)
caused by platinum and taxane agents
–2 –1 0 1 2
(mainly paclitaxel or oxaliplatin). Dur-
Mean Difference (95% CI)
ing initial treatment, the mean differ-
ence between the 2 groups of the change Duloxetine was compared against placebo in studies involving patients with diabetic neuropathy, Goldstein et
in average pain score was 0.73 (P=.003), al; fibromyalgia, Russell et al; osteoarthritis, Chappell et al; and chemotherapy-induced peripheral neuropa-
thy, the current study, including a breakdown by neurotoxic agent that patients in the current study took.
which compares favorably to mean dif-
ferences in average pain scores (range,
0.60-0.98) observed in patients receiv- ment of approximately 10% that is con- nerve injury mechanisms,16,52-55 the
ing duloxetine for US Food and Drug sistent with the IMMPACT definition of mechanisms of taxane- vs platinum-
Administration–approved indications a minimal clinically important difference. induced peripheral nerve injury are quite
for painful diabetic neuropathy, fibro- In addition, results of the exploratory re- different,16 possibly explaining why pa-
myalgia, and osteoarthritis (eTable 8 sponder analysis suggest that the relative tients treated with platinum reported less
available at www.http://www.jama risk of experiencing a 30% and 50% im- pain in the current study.
.com).13,26,30 The observed mean differ- provement in pain severity statistically fa- The current trial has several strengths
ence in the average pain score between vored duloxetine. and limitations. The strengths include the
the duloxetine and placebo groups in pa- Other factors to consider when judg- prospective, randomized, placebo-
tients treated with platinum was larger ing clinical significance include how controlled trial design and the geographi-
than results reported by Goldstein and quickly the drug takes effect, tolerabil- cally diverse sample. Regarding limita-
colleagues13 (FIGURE 4). However, du- ity, and the drug’s influence on other ef- tions, first, there was an imbalance in the
loxetine-related clinically meaningful ficacy end points such as function and dropout rate due to adverse effects in pa-
improvement in other painful condi- QOL.39 In the current study, pain scores tients treated with duloxetine vs pla-
tions may not be directly comparable decreased in patients treated with du- cebo (11% vs 1%, respectively), despite
with painful chemotherapy-induced pe- loxetine relatively quickly, within the first similar adverse effect rates in both
ripheral neuropathy. week of therapy (Figure 2). Consistent groups. One reason for this differential
In addition to the magnitude of the im- with our results (eTable 7), several stud- in the dropout rate may be the higher
provement, several other factors should ies show that duloxetine is safe and well- proportion of grade 3 adverse events re-
be considered when judging clinical sig- tolerated.13,14,26,30,49-51 Furthermore, du- ported by patients treated with dulox-
nificance, such as the treatment effect loxetine is associated with improved etine. These patients may have been able
size.39 Our results revealed a moderately function and QOL. Therefore, after con- to guess which drug they were taking,
large treatment effect size (0.513). Based sidering the many factors in addition to and those experiencing no or minimal
on the IMMPACT recommendations, the magnitude of improvement in pain pain relief may have dropped out.
clinical meaningfulness is also based on scores, study results strongly suggest that Another potential study limitation has
the results of a responder analysis, spe- duloxetine treatment is associated with to do with how baseline pain was deter-
cifically the proportion of patients expe- a clinically meaningful improvement in mined. Oncology providers commonly
riencing a 30% or a 50% improvement in chemotherapy-induced peripheral neu- use the NCI CTCAE, or other similar
painseverity.21,39,48 A10%to20%decrease ropathic pain. grading scales, to guide a history and
in pain severity is considered to represent Resultsfromourexploratorysubgroup physical examination focused on
a minimal clinically important change, a analysis lend support to the premise that chemotherapy-induced peripheral neu-
30% change represents a moderately im- differences in pathophysiologic mecha- ropathy and to grade its severity.9,16
portant improvement and a 50% change nisms may help to explain duloxetine re- Therefore, we relied on standard
represents a substantially important im- sponse rate variations across neuropathic CTCAE-guided practices for determin-
provement.21,39,48 Duringinitialtreatment, painconditions.Justasresponseratesmay ing severity at baseline. We did not spe-
the mean change in average pain score re- vary when duloxetine is used to treat dia- cifically train the examiners regarding
portedbypatientstreatedwithduloxetine betic vs chemotherapy-induced periph- CTCAE use because its grading is deeply
inthecurrentstudywas1.06,animprove- eral neuropathy due to differences in embedded into oncology practice. Of
©2013 American Medical Association. All rights reserved. JAMA, April 3, 2013—Vol 309, No. 13 1365

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

note, despite its everyday use in oncol- Ohio State University Comprehensive Cancer Center, and Asa B. Smith, Student (University of Michigan). Dr
College of Medicine, Department of Internal Medi- Wang, Mr Taylor, Ms Pitcher, and Ms Jasinski received
ogy clinical settings, the CTCAE is cine, Columbus (Dr Paskett); Department of Psychia- salary support from the Alliance Cooperative Group Grant
known for its suboptimal interrater reli- try and Behavioral Sciences, Memorial Sloan- for their contributions to this work. All others provided
Kettering Cancer Center, New York, New York (Dr assistance without receiving financial compensation.
ability and poor sensitivity to detect Ahles); Cancer and Leukemia Group B/Alliance Central
subtle changes.32,56-60 As such, use of the Office, Chicago, Illinois (Dr Bressler); Department of
Medicine (Hematology/Oncology) and Neurology, REFERENCES
CTCAE could have resulted in misdi- Norris Cotton Cancer Center, Dartmouth-Hitchcock
agnosis. Despite its limitations, CTCAE Medical Center, Lebanon, New Hampshire (Dr Fadul); 1. Smith EM, Cohen JA, Pett MA, Beck SL. The reli-
Illinois Oncology Research Association (Ms Knox and ability and validity of a modified total neuropathy score-
use is consistent with community and reduced and neuropathic pain severity items when used
Dr Le-Lindqwister); Main Line Hematology and On-
academic clinical practice,9 and thus the cology Associates, Wynnewood, Pennsylvania (Dr Gil- to measure chemotherapy-induced peripheral neu-
generalizability of our findings is man); and Division of Oncology, Department of In- ropathy in patients receiving taxanes and platinums.
ternal Medicine, The Ohio State University Wexner Cancer Nurs. 2010;33(3):173-183.
enhanced. Other limitations are that Medical Center and Comprehensive Cancer Center, 2. Kautio AL, Haanpää M, Kautiainen H, Kalso E, Saarto
changes in concurrent ancillary analge- and James Cancer Hospital, College of Medicine, De- T. Burden of chemotherapy-induced neuropathy—a
partment of Internal Medicine Columbus (Dr cross-sectional study. Support Care Cancer. 2011;
sic dosage were not assessed, study find- Shapiro). 19(12):1991-1996.
ings may not be applicable to patients Author Contributions: Drs Smith and Pang had full 3. Loprinzi CL, Reeves BN, Dakhil SR, et al. Natural
access to all of the data in the study and take respon- history of paclitaxel-associated acute pain syndrome:
with painful chemotherapy-induced prospective cohort study NCCTG N08C1. J Clin Oncol.
sibility for the integrity of the data and the accuracy
peripheral neuropathy caused by other of the data analysis. 2011;29(11):1472-1478.
neurotoxic agents, and the study did not Study concept and design: Smith, Pang, Fleishman, 4. Smith EML, Bakitas MA, Homel P, et al. Prelimi-
Paskett, Ahles, Bressler, Fadul, Shapiro. nary assessment of a neuropathic pain treatment and
address long-term duloxetine treat- Acquisition of data: Cirrincione, Fleishman, Knox, referral algorithm for patients with cancer. J Pain Symp-
ment (beyond 5 weeks). Le-Lindqwister, Gilman. tom Manage. 2011;42(6):822-838.
Analysis and interpretation of data: Smith, Pang, 5. Bakitas MA. Background noise: the experience of
Despiteduloxetine’sacceptableadverse chemotherapy-induced peripheral neuropathy. Nurs
Cirrincione, Fleishman, Ahles, Fadul.
effect profile, the risk of duloxetine-drug Drafting of the manuscript: Smith, Pang, Cirrincione, Res. 2007;56(5):323-331.
6. Shimozuma K, Ohashi Y, Takeuchi A, et al. Taxane-
interactions should not be overlooked. Fleishman, Bressler, Knox.
induced peripheral neuropathy and health-related qual-
Critical revision of the manuscript for important in-
Duloxetine should not be used with other tellectual content: Smith, Pang, Cirrincione, Fleish-
ity of life in postoperative breast cancer patients un-
dergoing adjuvant chemotherapy: N-SAS BC 02, a
drugs that inhibit serotonin reuptake due man, Paskett, Ahles, Fadul, Le-Lindqwister, Gilman,
randomized clinical trial. Support Care Cancer. 2012;
Shapiro.
to the associated increased risk of sero- Statistical analysis: Smith, Pang, Cirrincione.
20(12):3355-3364.
tonin syndrome.61 Also, because dulox- 7. Dodd MJ, Cho MH, Cooper BA, Miaskowski C. The
Obtained funding: Smith.
effect of symptom clusters on functional status and
etine is a moderate cytochrome (CYP) Administrative, technical, or material support: Pang, quality of life in women with breast cancer. Eur J On-
Fleishman, Paskett, Ahles, Bressler, Knox,
P450 2D6 enzyme-inhibiter, coadminis- Le-Lindqwister, Shapiro.
col Nurs. 2010;14(2):101-110.
8. Tofthagen C. Surviving chemotherapy for colon can-
tration with CYP P450 2D6 substrates Study supervision: Smith, Pang, Fleishman, Paskett, cer and living with the consequences. J Palliat Med.
can lead to increased substrate drug Fadul, Gilman. 2010;13(11):1389-1391.
Conflict of Interest Disclosures: All authors have com- 9. Stubblefield MD, Burstein HJ, Burton AW, et al.
serum concentrations and associated pleted and submitted the ICMJE Form for Disclosure NCCN task force report: management of neuropa-
toxicities.62-65 Concurrent use of dulox- of Potential Conflicts of Interest. Dr Smith reported thy in cancer. J Natl Compr Canc Netw. 2009;
receiving support from CALGB/Alliance for travel to 7(suppl 5):S1-S26.
etine with warfarin, nonsteroidal anti- meetings. Dr Paskett reported institutional support from 10. Saarto T, Wiffen PJ. Antidepressants for neuro-
inflammatory drugs, or both may also CALGB/Alliance for travel to meetings. Dr Ahles re- pathic pain. Cochrane Database Syst Rev. 2007;
ported receiving support from CALBG/Alliance for
increase bleeding risk.61,66 Lastly, if du- travel to meetings. Dr Fadul reported pending insti-
(4):CD005454.
11. Willis WD, Westlund KN. Neuroanatomy of the
loxetineandtamoxifenaretakentogether, tutional grants from Genentech. Dr Gilman reported pain system and of the pathways that modulate pain.
duloxetine-induced CYP P450 2D6 en- institutional and direct grants pending from the NCI. J Clin Neurophysiol. 1997;14(1):2-31.
No other financial disclosures were made. 12. Bril V, England JD, Franklin GM, et al; American
zyme inhibition could inhibit tamoxifen Funding/Support: This study was supported by grant Academy of Neurology; American Association of Neu-
conversion to its active metabolite, en- CA31946 from the NCI Division of Cancer Preven- romuscular and Electrodiagnostic Medicine; American
tion, the Alliance Statistics and Data Center, and the Academy of Physical Medicine and Rehabilitation. Evi-
doxifen.61,63,67,68 Alliance Chairman. Drug and placebo were supplied dence-based guideline: treatment of painful diabetic neu-
In conclusion, 5 weeks of duloxetine by Eli Lilly. The NCI provided funding for data man- ropathy—report of the American Association of Neu-
agement and statistical analysis. romuscular and Electrodiagnostic Medicine, the American
treatment was associated with a statisti- Role of the Sponsor: The NCI and Eli Lilly each re- Academy of Neurology, and the American Academy of
cally and clinically significant improve- viewed and approved the study concept via the usual Physical Medicine & Rehabilitation. Muscle Nerve. 2011;
ment in pain compared with placebo. Ex- peer-review process. Minor suggestions were made 43(6):910-917.
by each group regarding aspects of the study design. 13. Goldstein DJ, Lu Y, Detke MJ, Lee TC, Iyengar S.
ploratory analyses raise the possibility Neither the NCI Division of Cancer Prevention nor Eli Duloxetine vs placebo in patients with painful dia-
that duloxetine may work better for Lilly had a role in data collection or management, analy- betic neuropathy. Pain. 2005;116(1-2):109-118.
sis, interpretation of the data, or with manuscript prepa- 14. Wernicke JF, Pritchett YL, D’Souza DN, et al. A
oxaliplatin-induced rather than taxane- ration, review, or approval. randomized controlled trial of duloxetine in diabetic
induced painful chemotherapy-induced Online-Only Material: The eAppendix, 8 eTables, eFig- peripheral neuropathic pain. Neurology. 2006;
ure, and Author Video Interview are available at http: 67(8):1411-1420.
peripheral neuropathy. //www.jama.com. 15. Raskin J, Pritchett YL, Wang F, et al. A double-
Author Affiliations: University of Michigan School of Additional Contributions: We thank the study partici- blind, randomized multicenter trial comparing dulox-
Nursing, Ann Arbor, Michigan (Dr Smith); Alliance Sta- pants and the research staff at all participating sites. Spe- etine with placebo in the management of diabetic pe-
tistics and Data Center (Dr Pang and Ms Cirrincione) cial thanks are extended to the CALGB Oncology Nurs- ripheral neuropathic pain. Pain Med. 2005;6(5):
and Department of Biostatistics and Bioinformatics (Dr ing Committee, Richard Schilsky, MD (American Society 346-356.
Pang), Duke University, Durham, North Carolina; Can- of Clinical Oncology), Xiaofei Wang, PhD (Duke Uni- 16. Argyriou AA, Bruna J, Marmiroli P, Cavaletti G.
cer Supportive Services, Continuum Cancer Centers versity), John Taylor, MA (Alliance), Brandelyn Pitcher, Chemotherapy-induced peripheral neurotoxicity
of New York, Beth Israel Hospital, and St Luke’s– MS (Duke University), Sara Jasinski, BA (Duke Univer- (CIPN): an update. Crit Rev Oncol Hematol. 2012;
Roosevelt, New York, New York (Dr Fleishman); The sity), Celia Bridges, BSN, RN (University of Michigan), 82(1):51-77.

1366 JAMA, April 3, 2013—Vol 309, No. 13 ©2013 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
DULOXETINE ON CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY PAIN

17. Tan G, Jensen MP, Thornby JI, Shanti BF. Vali- 34. Cella D, Eton DT, Fairclough DL, et al. What is a low back pain. Spine (Phila Pa 1976). 2010;35
dation of the Brief Pain Inventory for chronic nonma- clinically meaningful change on the Functional As- (13):E578-E585.
lignant pain. J Pain. 2004;5(2):133-137. sessment of Cancer Therapy-Lung (FACT-L) ques- 51. Raskin J, Wang F, Pritchett YL, Goldstein DJ. Du-
18. Cleeland CS. The brief pain inventory user guide. tionnaire? results from Eastern Cooperative Oncol- loxetine for patients with diabetic peripheral neuro-
http://www.mdanderson.org/education-and-research ogy Group (ECOG) study 5592. J Clin Epidemiol. 2002; pathic pain: a 6-month open-label safety study. Pain
/departments-programs-and-labs/departments-and 55(3):285-295. Med. 2006;7(5):373-385.
-divisions/symptom-research/symptom-assessment 35. Norman GR, Sloan JA, Wyrwich KW. Interpreta- 52. Spallone V, Lacerenza M, Rossi A, Sicuteri R,
-tools/BPI_UserGuide.pdf. Updated 2009. Accessed tion of changes in health-related quality of life: the Marchettini P. Painful diabetic polyneuropathy: ap-
July 28, 2009. remarkable universality of half a standard deviation. proach to diagnosis and management. Clin J Pain.
19. Cleeland CS, Ryan KM. Pain assessment: global Med Care. 2003;41(5):582-592. 2012;28(8):726-743.
use of the Brief Pain Inventory. Ann Acad Med 36. Dubois D, Dhawan R, van de Velde H, et al. De- 53. Morales-Vidal S, Morgan C, McCoyd M, Hornik
Singapore. 1994;23(2):129-138. scriptive and prognostic value of patient-reported out- A. Diabetic peripheral neuropathy and the manage-
20. Atkinson TM, Mendoza TR, Sit L, et al. The Brief comes: the bortezomib experience in relapsed and re- ment of diabetic peripheral neuropathic pain. Post-
Pain Inventory and its “pain at its worst in the last 24 fractory multiple myeloma. J Clin Oncol. 2006; grad Med. 2012;124(4):145-153.
hours” item: clinical trial end point considerations. Pain 24(6):976-982. 54. Smith AG, Singleton JR. Diabetic neuropathy. Con-
Med. 2010;11(3):337-346. 37. Cella D, Eton DT, Lai JS, Peterman AH, Merkel tinuum (Minneap Minn). 2012;18(1):60-84.
21. Dworkin RH, Turk DC, Peirce-Sandner S, et al. Re- DE. Combining anchor and distribution-based meth- 55. Callaghan BC, Cheng HT, Stables CL, Smith AL,
search design considerations for confirmatory chronic ods to derive minimal clinically important differences Feldman EL. Diabetic neuropathy: clinical manifesta-
pain clinical trials: IMMPACT recommendations. Pain. on the Functional Assessment of Cancer Therapy tions and current treatments. Lancet Neurol. 2012;
2010;149(2):177-193. (FACT) anemia and fatigue scales. J Pain Symptom 11(6):521-534.
22. Kaur H, Hota D, Bhansali A, Dutta P, Bansal D, Manage. 2002;24(6):547-561. 56. Postma TJ, Heimans JJ, Muller MJ, Ossenkoppele
Chakrabarti A. A comparative evaluation of amitrip- 38. Common terminology criteria for adverse events GJ, Vermorken JB, Aaronson NK. Pitfalls in grading se-
tyline and duloxetine in painful diabetic neuropathy: version 4.02 (CTCAE). http://evs.nci.nih.gov/ftp1 verity of chemotherapy-induced peripheral neuropathy.
a randomized, double-blind, cross-over clinical trial. /CTCAE/About.html. Updated 2009. Accessed Janu- Ann Oncol. 1998;9(7):739-744.
Diabetes Care. 2011;34(4):818-822. ary 7, 2010. 57. Kuroi K, Shimozuma K, Ohashi Y, et al. Prospec-
23. Yang YH, Lin JK, Chen WS, et al. Duloxetine im- 39. Dworkin RH, Turk DC, McDermott MP, et al. In- tive assessment of chemotherapy-induced peripheral
proves oxaliplatin-induced neuropathy in patients with terpreting the clinical importance of group differ- neuropathy due to weekly paclitaxel in patients with
colorectal cancer: an open-label pilot study. Support ences in chronic pain clinical trials: IMMPACT advanced or metastatic breast cancer (CSP-HOR 02
Care Cancer. 2011;20(7):1491-1497. recommendations. Pain. 2009;146(3):238-244. study). Support Care Cancer. 2009;17(8):1071-
24. Pritchett YL, McCarberg BH, Watkin JG, Robinson 40. Dworkin RH, Turk DC, Wyrwich KW, et al. In- 1080.
MJ. Duloxetine for the management of diabetic pe- terpreting the clinical importance of treatment out- 58. Cavaletti G, Frigeni B, Lanzani F, et al. Chemo-
ripheral neuropathic pain: response profile. Pain Med. comes in chronic pain clinical trials: IMMPACT therapy-induced peripheral neurotoxicity assess-
2007;8(5):397-409. recommendations. J Pain. 2008;9(2):105-121. ment: a critical revision of the currently available tools.
25. Mease PJ, Spaeth M, Clauw DJ, et al. Estimation 41. Kajdasz DK, Iyengar S, Desaiah D, et al. Dulox- Eur J Cancer. 2010;46(3):479-494.
of minimum clinically important difference for pain in etine for the management of diabetic peripheral neu- 59. Lavoie Smith EM, Cohen JA, Pett MA, Beck SL.
fibromyalgia. Arthritis Care Res (Hoboken). 2011; ropathic pain: evidence-based findings from post hoc The validity of neuropathy and neuropathic pain mea-
63(6):821-826. analysis of three multicenter, randomized, double- sures in patients with cancer receiving taxanes and
26. Chappell AS, Desaiah D, Liu-Seifert H, et al. A blind, placebo-controlled, parallel-group studies. Clin platinums. Oncol Nurs Forum. 2011;38(2):133-
double-blind, randomized, placebo-controlled study Ther. 2007;29(suppl):2536-2546. 142.
of the efficacy and safety of duloxetine for the treat- 42. Cohen J. Statistical Power Analysis for the Be- 60. Frigeni B, Piatti M, Lanzani F, et al. Chemotherapy-
ment of chronic pain due to osteoarthritis of the knee. havioral Sciences. 2nd ed. Hillsdale, NJ: Lawrence Earl- induced peripheral neurotoxicity can be misdiag-
Pain Pract. 2011;11(1):33-41. baum; 1988. nosed by the National Cancer Institute Common
27. Chappell AS, Ossanna MJ, Liu-Seifert H, et al. Du- 43. Hammack JE, Michalak JC, Loprinzi CL, et al. Phase Toxicity scale. J Peripher Nerv Syst. 2011;16(3):
loxetine, a centrally acting analgesic, in the treat- III evaluation of nortriptyline for alleviation of symp- 228-236.
ment of patients with osteoarthritis knee pain: a 13- toms of cis-platinum-induced peripheral neuropathy. 61. Spina E, Trifirò G, Caraci F. Clinically significant
week, randomized, placebo-controlled trial. Pain. 2009; Pain. 2002;98(1-2):195-203. drug interactions with newer antidepressants. CNS
146(3):253-260. 44. Kautio AL, Haanpää M, Leminen A, Kalso E, Drugs. 2012;26(1):39-67.
28. Marangell LB, Clauw DJ, Choy E, et al. Compara- Kautiainen H, Saarto T. Amitriptyline in the preven- 62. Patroneva A, Connolly SM, Fatato P, et al. An as-
tive pain and mood effects in patients with comorbid tion of chemotherapy-induced neuropathic symptoms. sessment of drug-drug interactions: the effect of des-
fibromyalgia and major depressive disorder: second- Anticancer Res. 2009;29(7):2601-2606. venlafaxine and duloxetine on the pharmacokinetics
ary analyses of four pooled randomized controlled trials 45. Barton DL, Wos EJ, Qin R, et al. A double-blind, of the CYP2D6 probe desipramine in healthy subjects.
of duloxetine. Pain. 2011;152(1):31-37. placebo-controlled trial of a topical treatment for Drug Metab Dispos. 2008;36(12):2484-2491.
29. Frakes EP, Risser RC, Ball TD, Hochberg MC, chemotherapy-induced peripheral neuropathy: NC- 63. Skinner MH, Kuan HY, Pan A, et al. Duloxetine
Wohlreich MM. Duloxetine added to oral nonsteroi- CTG trial N06CA. Support Care Cancer. 2011; is both an inhibitor and a substrate of cytochrome
dal anti-inflammatory drugs for treatment of knee pain 19(6):833-841. P4502D6 in healthy volunteers. Clin Pharmacol Ther.
due to osteoarthritis: results of a randomized, double- 46. Rao RD, Flynn PJ, Sloan JA, et al. Efficacy of la- 2003;73(3):170-177.
blind, placebo-controlled trial. Curr Med Res Opin. motrigine in the management of chemotherapy- 64. Hua TC, Pan A, Chan C, et al. Effect of dulox-
2011;27(12):2361-2372. induced peripheral neuropathy: a phase 3 random- etine on tolterodine pharmacokinetics in healthy
30. Russell IJ, Mease PJ, Smith TR, et al. Efficacy and safety ized, double-blind, placebo-controlled trial, N01C3. volunteers. Br J Clin Pharmacol. 2004;57(5):652-
of duloxetine for treatment of fibromyalgia in patients Cancer. 2008;112(12):2802-2808. 656.
with or without major depressive disorder: results from 47. Rao RD, Michalak JC, Sloan JA, et al; North Cen- 65. Preskorn SH, Greenblatt DJ, Flockhart D, et al.
a6-month,randomized,double-blind,placebo-controlled, tral Cancer Treatment Group. Efficacy of gabapentin Comparison of duloxetine, escitalopram, and sertra-
fixed-dose trial. Pain. 2008;136(3):432-444. in the management of chemotherapy-induced pe- line effects on cytochrome P450 2D6 function in
31. Calhoun EA, Welshman EE, Chang CH, et al. Psy- ripheral neuropathy: a phase 3 randomized, double- healthy volunteers. J Clin Psychopharmacol. 2007;
chometric evaluation of the Functional Assessment of blind, placebo-controlled, crossover trial (N00C3). 27(1):28-34.
Cancer Therapy/Gynecologic Oncology Group- Cancer. 2007;110(9):2110-2118. 66. Glueck CJ, Khalil Q, Winiarska M, Wang P. In-
Neurotoxicity (FACT/GOG-Ntx) questionnaire for pa- 48. Farrar JT, Young JP Jr, LaMoreaux L, Werth JL, teraction of duloxetine and warfarin causing severe
tients receiving systemic chemotherapy. Int J Gyne- Poole RM. Clinical importance of changes in chronic elevation of international normalized ratio. JAMA.
col Cancer. 2003;13(6):741-748. pain intensity measured on an 11-point numerical pain 2006;295(13):1517-1518.
32. Griffith KA, Merkies ISJ, Hill EE, Cornblath DR. Mea- rating scale. Pain. 2001;94(2):149-158. 67. Jin Y, Desta Z, Stearns V, et al. CYP2D6 geno-
sures of chemotherapy-induced peripheral neuropa- 49. Hall JA, Wang F, Oakes TM, Utterback BG, Crucitti type, antidepressant use, and tamoxifen metabolism
thy: a systematic review of psychometric properties. A, Acharya N. Safety and tolerability of duloxetine in during adjuvant breast cancer treatment. J Natl Can-
J Peripher Nerv Syst. 2010;15(4):314-325. the acute management of diabetic peripheral neuro- cer Inst. 2005;97(1):30-39.
33. Huang HQ, Brady MF, Cella D, Fleming G. Vali- pathic pain: analysis of pooled data from three placebo- 68. Caraci F, Crupi R, Drago F, Spina E. Metabolic drug
dation and reduction of FACT/GOG-Ntx subscale for controlled clinical trials. Expert Opin Drug Saf. 2010; interactions between antidepressants and anticancer
platinum/paclitaxel-induced neurologic symptoms: a 9(4):525-537. drugs: focus on selective serotonin reuptake inhibi-
gynecologic oncology group study. Int J Gynecol 50. Skljarevski V, Desaiah D, Liu-Seifert H, et al. Ef- tors and hypericum extract. Curr Drug Metab. 2011;
Cancer. 2007;17(2):387-393. ficacy and safety of duloxetine in patients with chronic 12(6):570-577.

©2013 American Medical Association. All rights reserved. JAMA, April 3, 2013—Vol 309, No. 13 1367

Downloaded From: https://jamanetwork.com/ Mexico | Access Provided by JAMA by Salvador Macias Diaz on 08/19/2022
EUROPEAN JOURNAL OF CANCER 4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9

available at www.sciencedirect.com

journal homepage: www.ejconline.com

Review

Efficacy of homeopathic therapy in cancer treatment

Stefania Milazzoa, Nancy Russellb, Edzard Ernst a,*


a
Department of Complementary Medicine, Peninsula Medical School, Universities of Exeter and Plymouth,
Institute of Health and Social Care, 25 Victoria Park Road, Exeter EX2 4NT, United Kingdom
b
Integrative Medicine Program Education, Unit 147, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston,
TX 77030-0049, United States

A R T I C L E I N F O A B S T R A C T

Article history: Many cancer patients use homeopathic approaches to increase their bodyÕs ability to fight
Received 23 September 2005 cancer, improve their physical and emotional well-being, and alleviate their pain resulting
Accepted 28 September 2005 from the disease or conventional treatments. Homeopathy is highly controversial as there
Available online 11 January 2006 is no plausible mode of action for these highly diluted remedies. The aim of this systematic
review is to summarize and critically evaluate the efficacy of homeopathic remedies used
Keywords: as a sole or additional therapy in cancer care. We have searched the literature using the
Homeopathy databases: Amed (from 1985); CINHAL (from 1982); EMBASE (from 1974); Medline (from
Homeopathic remedy 1951); and CAMbase (from 1998). Randomised and non-randomised controlled clinical trials
Cancer including patients with cancer or past experience of cancer receiving single or combined
homeopathic interventions were included. The methodological quality of the trials was
assessed by Jadad score. Six studies met our inclusion criteria (five were randomised clin-
ical trials and one was a non-randomised study); but the methodological quality was var-
iable including some high standard studies. Our analysis of published literature on
homeopathy found insufficient evidence to support clinical efficacy of homeopathic ther-
apy in cancer care.
Ó 2005 Elsevier Ltd. All rights reserved.

1. Introduction As a palliative or supportive treatment, homeopathy is


used mainly to strengthen the body in its fight against can-
Cancer is the second most frequent cause of death in devel- cer, to improve general well-being, and to alleviate pain
oped countries according to a World Health Organization resulting from disease or conventional treatments [2,3].
(WHO) report from 2003 [1]. Even though non-surgical Homeopathy is controversial as no plausible mode of action
orthodox treatments can control or even cure cancer, many has been identified for substances that are so highly diluted
adverse effects limit their use [2]. Cancer patients therefore that they can not be measured [4]. Homeopathic remedies
often turn towards complementary therapies, including are believed to be most effective when they are selected to
homeopathy [2]. A recent European survey has shown that address a total set of symptoms and characteristics [5] and
homeopathy is amongst the most commonly used comple- in classical or individualized homeopathy, choice of remedies
mentary therapies for cancer in 7 out of 14 European are based on the match of a patientÕs particular symptoms
countries [3]. with a remedy picture rather than conventional diagnosis

* Corresponding author: Tel.: +44 1392 424839; fax: +44 1392 427562.
E-mail addresses: stesincro@yahoo.com (S. Milazzo), nrussell@mdanderson.org (N. Russell), edzard.ernst@pms.ac.uk (E. Ernst).
0959-8049/$ - see front matter Ó 2005 Elsevier Ltd. All rights reserved.
doi:10.1016/j.ejca.2005.09.025
EUROPEAN JOURNAL OF CANCER 4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9 283

[6]. Prescribing homeopathic substances is based on its pro- toxicological remedies are prepared according to the rules of
posed law of similars that suggests that ‘‘like cures like’’ [7]. homeopathy and are used in combinations as complex reme-
Although Hahnemann initially diluted these substances in dies. Some experts fail to differentiate between homeopathic
order to reduce toxicity, he came to believe that the actual and homotoxicological medicines. However, there are impor-
process of diluting and shaking imparted additional potency tant differences. Homeopathy follows the ‘‘like cures like’’
to each solution [5]. His process of testing natural substances principle, while homotoxicology does not [12]. Homotoxicol-
in healthy individuals became known as ‘‘drug proving’’ and ogy often makes use of biological material that would be
results continue to be collected into an encyclopaedia of atypical in homeopathy, such as material from pigs. Some re-
homeopathic drug effects known as the Materia Medica ports suggest the efficacy of homotoxicology for defined con-
[8,9]. In ‘‘classical homeopathy’’ single remedies are given to ditions, but many caveats exist [12].
patients, whereas in ‘‘complex homeopathy’’ several homeo- Isopathy is another subset of homeopathy that was devel-
pathic medicines are combined into one formula, where con- oped by Johann Lux in the 1830s. It differs from homeopathy
centration tends to be below 24X and usually below 12X [10] in that remedies are prepared from those substances that
(the numbers indicate the dilution of the homeopathic rem- cause the illness (e.g., allergens or bacteria) [13] and several
edy; that is, remedies are obtained by ‘‘decimal dilution’’, one trials have suggested its clinical efficacy [14].
part substance to nine parts alcohol, and then labelled by the The aim of this systematic review is to summarize and crit-
letter X or D). ically evaluate the clinical trial evidence for the effectiveness
In the 1950s, Hans H. Reckeweg developed a new form of of any type of homeopathic remedy in cancer care.
homeopathy known as homotoxicology [11], which generally
uses formulations that contains measurable amounts of 2. Methods
homeopathically prepared active ingredients, designed to
work with the bodyÕs defence mechanisms and facilitate the Electronic literature searches were conducted using the
bodyÕs elimination of toxic substances (homotoxins). Homo- following databases: Amed (from 1985); CINHAL (from 1982);

Table 1 – Search terms to identify the studies for this systematic review
Database Intervention(s) Cancer patients Types of study design Location of studies

PubMed 1. Homeopath*.ab, de. 9. Cancer*.ti, ab, rw, sh. 21. Clinical trial.pt. 33. 8 AND 20 AND 32
2. Homeopath* remed*.ab, de. 10. Neoplasm*.ti, ab, rw, sh. 22. Controlled clinical trial.pt.
3. Homeopath* drug*.ab, de. 11. Terminal adj.illness.ti,ab 23. Randomised controlled
trial.pt.
4. Simil*.ab, de. 12. Carcin*.ti, ab, rw, sh. 24. Random*.tw.
5. Homotoxicology.ab, de. 13. Oncol*.ti, ab, rw, sh. 25. Double blind.tw.
6. Single remedy.ab, de. 14. Sarcoma.ti, ab, rw, sh. 26. Placebo*.tw.
7. Combination remedies 15. Tumour.ti, ab, rw, sh. 27. Control*.tw.
8. OR/1–7 16. Leuk*.ti, ab, rw, sh. 28. Symptom*.tw.
17. Aden*.ti, ab, rw, sh. 29. Quality of life.tw.
18. Malignant.ti, ab, rw, sh. 30. Palliative treatment.tw.
19. Lymphoma.ti, ab, rw, sh. 31. Palliative care.tw.
20. OR/9–19 32. OR/21–31

NHS dialog 1. Homeopath$2.AB.DE. 9. Cancer$6.AB. 21. Clinical trial.AB. 33. 8 AND 20 AND 32
(AMED, CINHAL, 2. Homeopath$2 10. Neoplasm$4.AB. 22. Control$3.AB.
EMBASE, MEDLINE) adj.remed$3.AB.DE
3. Homeopath$2 11. Terminal 23. Study.AB.
adj.drug$1.AB.DE adj.illness.AB.DE
4. Simil$4.DE. 12. Carcin$6.AB. 24. Random$7.AB.
5. Homotoxicology.DE. 13. Oncol$4.AB. 25. Double adj.Blind.AB.
6. Single adj.remedy.DE. 14. Sarcoma.AB. 26. Placebo.AB.
7. Combination 15. Tumour$3.AB. 27. Control$4.AB.DE
adj.remedies.DE.
8. OR/1–7 16. Leuk$5.AB. 28. Sympto$2.AB.
17. Aden$6.AB. 29. Quality adj.life.AB.
18. Malignant.AB. 30. Palliative adj.treatment.DE.
19. Lymphoma.AB. 31. Palliative adj.care.DE.
20. OR/9–19 32. OR/21–31

CAMbase N/A N/A N/A Homeopathy for cancer;


homeopathic complex
remedies for cancer;
trial of homeopathy
for cancer

sh, subheading; ab, abstract; tw, textword; *, truncation; ti, title word; $, truncation; de, drug effect; pt, publication type.
284 EUROPEAN JOURNAL OF CANCER 4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9

EMBASE (from 1974); Medline (from 1951); and CAMbase (from severe degree of reaction. Reaction index was lower in both
1998). AMED, CINHAL, and EMBASE were searched through intervention groups compared to placebo (5 for homeopathic
the NHS Dialog portal, whereas MEDLINE was searched groups vs. 8.5 for placebo group). No significant differences in
through NHS Dialog as well as through PubMed. CAMbase tumour reduction were observed in the study.
was searched by simply typing a naturally spoken sentence, A study by Oberbaum [16] tested he effects of TraumeelSÒ
as the system requires. on chemotherapy-induced stomatitis (mouth sores) in a non-
Randomised and non-randomised controlled clinical trials randomized CCT. This treatment for 20 children and teenag-
(RCTs and CCTs, respectively) were included if they investi- ers was compared with seven randomly chosen controls from
gated patients with cancer or past experience of cancer who the same age group with similar stages of cancer, who re-
received single or combined homeopathic interventions as ceived no treatments for stomatitis.
sole treatment or as adjuvant to conventional treatments, TraumeelSÒ is an homeopathic preparation containing: ar-
and their outcomes were compared to any other intervention nica 2X, calendula 2X, millefolium 3X, chamomilla 3X, sym-
or no intervention. Details of the literature searches are sum- phytum 6X, belladonna 2X ana 0.1 ml, aconitum 2X 0.06 ml,
marised in Table 1. bellis perennis 2X 0.05 ml, hypericum 2X 0.03 ml, echinacea
The main outcome measures we considered were efficacy angustifolia 2X, echinacea purpurea 2X ana 0.025 ml, hama-
of homeopathic remedies for treating symptoms in cancer melis 1X 0.01, mercurius sol. 6X 0.05 g, and hepar sulfuris
patients and cancer survivors. Secondary outcome measures 6X 0.1 g. It was administered orally using liquid doses pro-
included tumour response and quality of life. All articles were vided in vials. The primary outcome measure was the level
read by two independent reviewers and data from the articles of pain measured according to opiate requirements. Ulcer
were validated and extracted according to pre-defined crite- severity was assessed according to the WHO staging of dis-
ria. The methodological quality of all studies was indepen- ease: 0 for no ulcer; 1 for oral pain with no ulcers; 2 for oral
dently assessed by the two reviewers using Jadad score [26]. pain with ulcers but the ability to eat is retained; 3 for liquid
Discrepancies between reviewers were resolved by a third diet only; and 4 for inability to eat or to drink. In all treated
independent reviewer. Statistically significant results of each patients, the treatment was followed by an immediate de-
trial were documented. crease in pain. There was a non-significant trend suggesting
less patients in the intervention group required opiates com-
3. Results pared to the control group (P = 0.09) and some patients in this
group also reported a mood improvement. Symptom duration
The searches identified 55 potentially relevant studies, of was numerically different between the two groups favouring
which six met our inclusion criteria (Fig. 1). Three were dou- the treated group (6 vs. 13 days). Oberbaum [17] subsequently
ble-blinded RCTs, one was a triple-blinded study, and another carried out a larger RCT to assess the effectiveness of Traume-
one was a CCT. Key data are summarised in Tables 2 and 3. elSÒ for chemotherapy-induced stomatitis in cancer patients
Kulkarni [15] conducted an RCT in cancer patients to as- after allogeneic or autologous stem-cell transplantation. Pa-
sess the effectiveness of homeopathy on the severity of radio- tients (ages 3–25 years) were randomised to two groups
therapy-related side effects. Patients with different types of receiving, in addition to standard mouth-washes, TraumeelSÒ
cancer were randomized into three parallel arms: placebo; oral rinse or a placebo rinse. TraumeelSÒ preparation con-
cobaltum 30; and causticum 30 (types of dilution were not tained high dilutions (10 1–10 9) of different extracts (see
specified). These homeopathic remedies were selected be- above). The main outcomes were occurrence of stomatitis
cause they mimic various symptoms of radiation reaction. and time to worsening of symptoms. Subjective symptom
All the patients were evaluated once a week according to scores were recorded during the first 7 days of the trial. Signif-
an 18-point radiation reaction profile, and the average grading icant differences occurred in the reduction of severity and/or
was calculated at the end of the study: 0–5 for minimal reac- duration of stomatitis in the intervention group (P < 0.01)
tion; 6–10 for moderate but tolerable reaction; and >11 for compared to the placebo group. Mean ‘‘area under the curve’’

Total articles retrieved for homeopathic and


homotoxicology treatments in cancer care: 55

Animal and in vitro


studies: 12
Observational studies:
37

Included studies: 6*

*Included studies:
Randomized Clinical trials: 5
Controlled Clinical trials: 1

Fig. 1 – Flowchart of inclusion process.


Table 2 – List of included studies
First Jadad Study designa Sample Patient Intervention Control Condition Outcome Main results P value Adverse
author score and follow up size condition (n) intervention investigated measures events
(year) [26] (n) (nP:nI)

Kulkarni 1 RCT with three 82 Cancer Cobaltum 30 Placebo (28) Radiation Primary outcome: Lower radiation NS None
1988 [15] parallel arms patients (28) reaction Degree of reaction in reported
Follow up: NS undergoing Causticum 30 reaction (0–5; intervention groups
radiation (26) 6–10; >11)
therapy

Oberbaum 0 CCT 27 Leukemia TraumeelÒ No treatment Chemotherapy Primary outcome: Lower opiate 0.09 None
1998 [16] Induced- Opiate requirements in reported

EUROPEAN JOURNAL OF CANCER


Stomatitis requirements treated group
for pain
Follow up: NS Secondary: Decrease in duration NS
Duration of of symptoms in
symptoms; intervention group
Quality of life
Mood improvement NS
in intervention
group

Balzarini 4 Randomised 61 Breast Belladonna Placebo (32) Radiodermatitis Primary outcome: Skin temperature 0.008, 0.016, Hot flushes,
2000 [18] double-blind cancer and 7cH and X-ray Skin heat; decrease at week 3, 0.023, 0.011, perspiration
placebo- undergoing 15cH (29) Hyperpig- 4, 6, 8 and 10 0.250 and
controlled radio- mentation; migraine

4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9
clinical trial therapy Erythema; (0:1)
Oedema
Follow up: 10 Secondary: Total Less frequent 0.050,
weeks severity of hyperpigmentation 0.060
symptoms at week 5 and 10
Less intense colour 0.280
of the skin at week
10
High frequency of 0.025
oedema at week 5, 6
and 10
Decrease of total 0.025
severity of
symptoms during
recovery
0.890
0.05

Report of main results in the intervention groups.


nP:nI number of patients in the placebo group:number of patients in the intervention group; NS, not specified.
a If not stated otherwise trials have two parallel arms.

285
286
Table 3 – List of included studies. Report of main results in the intervention group
First author Jadad Study design* Sample Patient Intervention Control Condition Outcome Main results P value Adverse
(year) score and follow up size condition (n) intervention investigated measure events
[26] (n) (nP:nI)

Oberbaum 4 Random. 30 Patients TraumeelSÒ Placebo (15) Chemotherapy- Primary outcome: Reduction of <0.01 Graft versus
2001[17] double-blind affected by (15) induced Occurrence of severity and host disease
placebo blood malignant stomatitis stomatitis; Time duration of (6:3)
controlled cancer, who to worsening of stomatitis in
clinical trial underwent BMT symptoms intervention group
Follow up: Secondary: Oral Decrease of time to <0.001 Sepsis (8:3)
44 weeks pain worsening of

EUROPEAN JOURNAL OF CANCER


symptoms in
intervention group
Reduction in oral NS
pain and discomfort;
dryness of mouth
and tongue;
difficulty to swallow;
and dysphagia in the
intervention group
Gastrointe-
stinal
complication
(5:0)

4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9
Venous
occlusive
disease (0:4)
Pneumonitis
(0:4)

Jacobs 2005 5 Triple-blind RCT 83 Breast cancer HylandÕs Identical Menopausal Primary outcome: Increase of hot 0.01; Headaches in
[19] with three survivors menopause placebo (27) symptoms Hot flushes flushes severity <0.001 combination
parallel arms (30) severity; Hot score in remedy group
flushes combination (NS)
frequency remedy group
compared to placebo
and single remedy
Follow up: 35 different Secondary: Increase of hot 0.006;
1 year single Quality of life flushes frequency in 0.002
remedies (26) comb. group
compared to placebo
and single remedy
Lower KMI score in Not
single remedy group signific.
Improvement of <0.05
quality of life in both
intervention groups
EUROPEAN JOURNAL OF CANCER 4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9 287

score was 10.4 compared to 24.3. There was also a statistically


significant difference between the two groups in the mean
time to worsening of symptoms with an average of 6.9 days
Adverse
events
(nP:nI)

in the intervention group compared to 4.3 days in the placebo


group (P < 0.001). However the median times were similar (4.7
NS

compared to 4.0, respectively). In a subgroup of patients, aged


P value

less than 15 years, the lower severity score in the intervention


group (11 compared to 25.9 of placebo) was also statistically
/

significant (P < 0.01). Patients in the TraumeelSÒ group


Neither positive nor

showed a reduction in oral pain and discomfort; dryness of


significant results

mouth and tongue; difficulty to swallow; and dysphagia (no


were observed
Main results

P values of statistical significance for these symptoms were


provided). Nausea was reported in two patients in each group
at the beginning of the trial and they were not included in the
analysis. Serious complications occurred in both treatment
groups without significant differences between the two
severity; Quality
MYMOP Activity
Primary outcome:

groups. Graft vs. host disease, sepsis, and gastrointestinal


score; MYMOP

frequency and
Secondary: Hot
Profile score

complications occurred mainly in the placebo group. On the


Outcome
measure

other hand, more patients in the intervention group experi-


flushes

of life

enced venous occlusive disease and pneumonitis.


Balzarini [18] investigated the effectiveness of homeopathic
treatment for skin reactions during radiotherapy for breast
investigated

cancer treatment. Patients were randomised into two groups


withdrawal
Condition

symptoms

receiving homeopathic remedies or placebo. Both groups also


Estrogen

received a topical medication containing fluocortolone.


Homeopathic treatment consisted of three granules of Bella-
nP:nI number of patients in the placebo group:number of patients in the intervention group; NS, not specified.

donna 7cH (twice a day) and X-ray 15cH (once a day). The main
able placebo (25)

outcomes measured in this study were: erythema; skin heat;


intervention

Indistinguish-

cutaneous and subcutaneous oedema; and hyperpigmenta-


tion. Patients treated with homeopathy appeared to experience
Control

transient benefits. Less hyperpigmentation (P value at 5th


(n)

week = 0.050) and decrease of skin heat (P value at 8th


week = 0.011) were observed although these differences were
Intervention

Tablets liquid

no longer significant by the end of the 10-week follow up. Total


71 different

or granules
remedies.

severity score was positive in favour of homeopathic treatment


during radiotherapy and recovery, but statistical significance
(28)

for the difference was noticed only during recovery (P = 0.05).


(n)

High frequency of oedema was observed in the interven-


tion group, and statistical significance was reached at the
Breast cancer

5th and 6th week (P = 0.025). Otherwise, adverse events were


condition

equally distributed between the two groups.


survivor
Sample Patient

Jacobs and colleagues [19] evaluated the homeopathic ef-


fects on menopausal symptoms in breast cancer survivors. Pa-
tients who suffered from menopausal symptoms; had a
* If not stated otherwise trials have 2 parallel arms.

history of carcinoma in situ or stage I–III breast cancer; had


53
size

completed all surgical, chemo- or radiation therapy; and had


an average of three hot flushes per day for a month before
score and follow up
Jadad Study design*

the trial were included in the trial. Following a ‘‘double-dum-


double-blind

clinical trial

my’’ design, each patient was prescribed an individualized


Follow up:
controlled

16 weeks
Random.

placebo-

homeopathic medication and randomized into three treat-


ment groups: a placebo combination and a verum single rem-
edy; a verum combination medicine and a verum single
remedy; and two placebo combinations. Single remedies con-
[26]

sisted of 35 different homeopathic medications, mainly: sepia,


calcarea carbonica, sulfur, lachesis, and kali carbonicum. The
First author

combination remedy was ‘‘HylandÕs menopause’’, which con-


Thompson
2005 [20]

tained: amyl nitrate, sanguinaria canadensis, and lachesis.


Main outcome measures were hot flush severity and num-
(year)

ber of hot flushes, measured according to the Kupperman


Menopausal Index (KMI) score. Quality of life score was
288 EUROPEAN JOURNAL OF CANCER 4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9

measured using the Short Form 36 (SF-36), which evaluated Among the six studies we included, five were randomised
mental and physical health status. Although the overall single [15,17–20] and only one was not randomised [16]. Statistical
remedy homeopathic group tended to experience lower sever- analysis for significance was performed in all the studies,
ity score and fewer hot flushes, no significant differences but only four provided statistical features in their result sec-
were found among the three groups in the univariate model tions. Oberbaum [16] discussed highly significant differences
of all patients adjusted for baseline, time, and tamoxifen between the groups regarding duration of symptoms, but no
use over 1 year period. A statistically significant improvement statistical features for those differences were given. This
in general health score was observed in both homeopathy was a pilot study conducted in order to test the rationale for
groups compared to placebo (combination: P < 0.03; single: performing a more rigorously designed trial [17]. The study
P = 0.02). A subgroup analysis was carried out for the patients by Kulkarni [15], investigating the effectiveness of some
taking (60%) or not taking (40%) tamoxifen. In the subgroup homeopathic remedies for radiation protection, concluded
not receiving tamoxifen, there was a statistical significant in- that the remedies reduced the degree of radiation reaction
crease in the hot flush severity score in the combination significantly, but results of their statistical analysis were not
homeopathy group compared to placebo (P = 0.01, 95% CI, shown. This study also lacked complete information regard-
6.2–47.1) and a highly significant difference when compared ing patients and remedies, as well as essential methodologi-
to single remedy (P < 0.001, 95% CI, 51.9–15.0). In the group cal details, such as randomisation method.
not receiving tamoxifen, the total number of hot flushes in- Even trials with a Jadad score of 5 were not devoid of flaws.
creased statistically significantly in the combination homeo- The small sample size in Jacobs [19] precluded definitive con-
pathic remedy group compared to the placebo and to the clusions, and a major flaw in the study was the use of combi-
single remedy (combination P = 0.006; single remedy nation remedies in an ongoing daily regimen, without
P = 0.002; 95% CI not provided). Statistically significant in- following over-the-counter instructions that suggested to dis-
crease of headaches was also observed in the combination continue use if adverse effects occurred. In the trial con-
homeopathic group (P = 0.03). ducted by Thompson [20], the high placebo response can
Thompson and co-workers [20] conducted a double-blind probably be related to a type II error in the study, since their
RCT to evaluate the effect of homeopathy in 53 breast cancer sample size was only adequate for detecting large differences
survivors affected by oestrogen withdrawal symptoms. Selec- in response.
tion criteria of patients included: more than three hot flushes Of the 6 trials included in this systematic review, only two
per day; not having metastatic disease; no concurrent treat- reported statistically significant positive results of their pri-
ment for hot flushes; no severe concurrent illness; and not mary outcomes. One of these was a RCT conducted by Ober-
undergoing chemotherapy or about to receive any adjuvant baum [17] that showed encouraging results confirming the
chemotherapy. Patients were randomized to receive homeo- need to perform the trial on a larger scale. The other study
pathic remedies or placebo. Patients randomized to homeop- was the RCT carried out by Balzarini [18], although this trial
athy were individually prescribed 71 different remedies, most showed statistically significant differences in favour of the
commonly: sulfur, sepia, carcinosin, natrum muriaticum, bel- intervention group, these differences were not consistently
ladonna, and arnica. Primary outcome measures were activity observed at all time points. The rest of the studies indicated
score and overall profile score, measured according to the a positive trend towards homeopathic interventions for
Measure Yourself Medical Outcome Profile (MYMOP). Second- improvements in quality of life and symptom management,
ary outcome measures included: hot flush frequency and hot which seems to justify further investigations.
flush severity (measured with Menopausal symptom ques- Homeopathic remedies are thought to trigger the bodyÕs
tionnaire); and quality of life (measured according to EORTC own defence and self-regulatory response. However, their
QLQ-C30, plus Breast module). No significant differences be- mode of action is unclear. As homeopathic remedies are often
tween intervention and placebo group for both MYMOP activ- diluted beyond AvogadroÕs number, no pharmacological ac-
ity and overall profile scores were noted (P = 0.17, 95% CI tion can be expected. Sceptics therefore insist that homeopa-
1.0–0.2; P=0.13, 95% CI 0.9–0.1, respectively). There were thyÕs clinical success is solely due to a placebo response [21].
no differences between the groups for any other secondary Homeopaths counter this criticism by postulating that
outcomes measured at final follow up. Adverse events were homeopathic remedies work through mechanisms other than
experienced by approximately one quarter of women in both pharmacological ones [22,23]. The evidence for homeopathic
groups and no significant difference was observed between remedies in cancer care may not be fully conclusive but it
active remedy and placebo group. does seem to warrant further study. Clinical trials of homeop-
athy should be rigorously designed to minimize bias. The
4. Discussion existing trials have a number of limitations, e.g. sample size,
which should be addressed in future research. Such research
Five out of six trials included in this systematic review yielded can be expensive and it is therefore a precondition that ade-
positive results, which suggest the effectiveness of homeo- quate research funds for homeopathy are made available.
pathic remedies for cancer care. Cancer patients appear to Considering that positive results have been obtained in some
have benefited from homeopathic interventions specifically cases [24] using the controversial remedy ‘‘Carcinosin’’ (a car-
for chemotherapy-induced stomatitis, radiodermatitis and cinogenic substance) and in some animals studies [25] inves-
general adverse events from radiotherapy. Breast cancer sur- tigating the possible anticancer effect of homeopathic
vivors, suffering from menopausal symptoms, experienced interventions, further studies testing homeopathy for tumour
a general improvement on their quality of life. response should also be undertaken.
EUROPEAN JOURNAL OF CANCER 4 2 ( 2 0 0 6 ) 2 8 2 –2 8 9 289

The main limitation of our systematic review is the lack 12. Ernst E, Schmidt K. Homotoxicology – a review of randomised
and sometimes poor quality of the primary data. The studies clinical trials. Eur J Clin Pharmacol 2004;60(5):299–306. Epub
we evaluated were highly heterogeneous in virtually every re- 2004 Jun 9.
13. http://www.bioresourceinc.com/articles/vol1no2.html
spect. In some studies, individualized remedies were applied.
[accessed 12.06.05].
Although individualization of therapy allows homeopathy to 14. Reilly DT, Taylor MA, McSharry C, et al. Is homoeopathy a
be practiced in its traditional fashion, this increases the com- placebo response? Controlled trial of homoeopathic potency,
plexity of comparing outcomes. In conclusion, the evidence with pollen in hayfever as model. Lancet 1986;2:881–6.
emerging from this systematic review is encouraging but 15. Kulkarni A, Nagarkar BM, Burde GS. Radiation protection by
not convincing. Further research should attempt to answer use of homoeopathic medicines. Hahnemann Homoeopath Sand
the many open questions related to homeopathy. 1988;12:20–3.
16. Oberbaum M. Experimental treatment of
chemotherapy-induced stomatitis using a homeopathic
Conflict of interest statement complex preparation: a preliminary study. Biol Med
1998;3(6):104–8.
None declared. 17. Oberbaum M, Yaniv I, Ben-Gal Y, et al. A randomized,
controlled clinical trial of the homeopathic medication
TRAUMEEL S in the treatment of chemotherapy-induced
stomatitis in children undergoing stem cell transplantation.
R E F E R E N C E S
Cancer 2001;92(3):684–90.
18. Balzarini A, Felisi E, Martini A, et al. Efficacy of homeopathic
treatment of skin reactions during radiotherapy for breast
1. http://www.who.int/cancer/media/en/788.pdf [accessed cancer: a randomised, double-blind clinical trial. Brit
01.06.05]. Homeopath J 2000;89(1):8–12.
2. Montfort H. A new homeopathic approach to neoplastic 19. Jacobs J, Herman P, Heron K, et al. Homeopathy for
diseases: from cell destruction to carcinogen-induced menopausal symptoms in breast cancer survivors: a
apoptosis. Brit Homeopath J 2000;89(2):78–83. preliminary randomized controlled trial. J Altern Complem Med
3. Molassiotis et al. Use of complementary and alternative 2005;11(1):21–7.
medicine in cancer patients: a European survey. Ann Oncol 20. Thompson EA, Montgomery A, Douglas D, et al. A pilot,
2005;16(4):655–63. randomized, double-blinded, placebo-controlled trial of
4. Shang A, Huwiler-Müntener K, Nartey L, et al. Are the clinical individualized homeopathy for symptoms of estrogen
effects of homoeopathy placebo effects? Comparative study withdrawal in breast-cancer survivors. J Altern Complem Med
of placebo-controlled trials of homoeopathy and allopathy. 2005;11(1):13–20.
The Lancet 2005;366(9487):726–32. 21. Vandenbroucke JP. Homoeopathy and ‘‘the growth of truth’’.
5. Jonas WB, Kaptchuk TJ, Linde K. A critical overview of The Lancet 2005;366(9487):691–2.
homeopathy. Ann Intern Med 2003;138:393–9. 22. Fisher P. The information medicine hypothesis. In: Shulte J,
6. Linde K, Melchart D. Randomized controlled trials of Endler PC, editors. Fundamental research in high
individualized homeopathy: a state-of -the art review. J Altern dilutions. Dordrecht: Kluwer; 1998. p. xi–xiv.
Complem Med 1998;4:371–88. 23. Witt C, Ludtke R, Weisshuhn TE, et al. High homeopathic
7. Robinson K. Homeopathy: questions and answers. New potencies are different from potentized solvent when
Mexico: Elegance publishing; 1994. investigated with the REDEM technology. Forsch Komp Klass
8. Bellavite P, Signorini A. Homeopathy: a frontier in medical Nat 2005;12(1):6–13.
science. North Atlantic Books. 24. Masiello DJ. Melanoma. Am J Holistic Med 2003–2004;96(4):
9. Ernst E, Kaptchuk TJ. Homeopathy revisited. Arch Int Med 318–20.
1996;156(19):2162–4. 25. Sato DY, Wal R, de Oliveira CC, et al. Histopathological and
10. https://www.cedrugstorenews.com/userapp/lessons/ immunophenotyping studies on normal and sarcoma
page_view_ui.cfm?lessonuid=&pageid= 180-bearing mice treated with a complex homeopathic
E44F4F4C5554FDD448D601472A02FFA0 medication. Homeopathy 2005;94(1):26–32.
[accessed 01.06.05]. 26. Jadad A, Moore A, Carroll D. Assessing the quality of
11. http://www.heel.com.au/homotoxicology/index.shtml randomized trials: is blinding necessary?. Control Clin Trial
[accessed 12.06.05]. 1996;17:1–12.
Tumores sólidos 2022 (actualizado).

49 . Cáncer en el embarazo:
consideraciones generales y
agentes antineoplásicos
Rachel Cossetti, Ana Carolina Salles de Mendonça y Antonio Carlos Buzaid

DEFINICIÓN
El cáncer asociado al embarazo se define como el cáncer diagnosticado
desde el primer día de la concepción hasta 1 año después del parto [ Best
Pract Res Clin Obstet Gynaecol 33:2, 2016 ].

EPIDEMIOLOGIA
El diagnóstico de cáncer asociado con el embarazo ocurre en
aproximadamente uno de cada 1000 a 1500 embarazos [ Oncologist 7:279,
2002 ; Am J Obstet Gynecol 189:1128, 2003 ; Int J Gynecol Cancer 27:613,
2017 ]. Esta incidencia ha ido en aumento durante las últimas décadas y
parece reflejar una mayor incidencia de cáncer en general y el retraso en
la edad del primer embarazo hasta la tercera y cuarta décadas de vida para
un número cada vez mayor de mujeres [ Surg Oncol 20:e175 , 2011 ]. Los
tipos de cáncer más comunes durante el embarazo son el cáncer de
mama, el melanoma, el cáncer de cuello uterino, el cáncer de ovario, las
neoplasias malignas hematológicas, el cáncer de tiroides y el cáncer
colorrectal [ J Clin Oncol 27:45, 2009 ].

CONSIDERACIONES GENERALES
La aparición de cáncer durante el embarazo agrega más complejidad al
manejo oncológico de la paciente, ya que la madre y el feto pueden verse
afectados por el tratamiento. El melanoma, las neoplasias malignas
hematológicas y el cáncer de pulmón son las únicas neoplasias malignas
con diseminación a la placenta y al feto descritas en la bibliografía [ J Clin
Oncol 21:2179, 2003 ]. Aunque no se comprende por completo, los cambios
hormonales, la supresión inmunitaria y el aumento de la vascularización y
la permeabilidad capilar están asociados con la fisiopatología del cáncer
en el embarazo [ World J Oncol 10:28, 2019 ]. El manejo ideal de estos casos
debe buscar las recomendaciones terapéuticas estándar para la
enfermedad oncológica de base, de acuerdo al pronóstico, guías
terapéuticas y supervivencia estimada, pero se debe considerar la
seguridad fetal al tomar decisiones, además de factores éticos,
psicológicos, religiosos y legales [ Best Pract Res Clin Obstet Gynaecol 33:2,
2016 ]. El embarazo debe manejarse como un embarazo de alto riesgo, con
monitoreo fetal regular mediante ecografía morfológica (US) y Doppler de
la arteria umbilical [ ESMO Open 1:e000016, 2016 ]. El objetivo debe ser
lograr un parto a término (≥ 37 semanas), ya que la prematuridad es uno
de los principales factores pronósticos que afectan el desarrollo pediátrico
hasta los 3 años de edad [ Lancet Oncol 19:337, 2018]. La modalidad de
parto debe elegirse con base en la recomendación obstétrica, excepto en
casos de neoplasias del tracto ginecológico. Sin embargo, en muchos
casos se prefiere la cesárea por la posibilidad de determinar la fecha exacta
del parto, lo que permite una mejor planificación de la fecha del último
tratamiento sistémico, entre otras intervenciones. Siempre se debe
evaluar cuidadosamente la placenta en busca de signos de
micrometástasis, particularmente en casos de melanoma [ ESMO Open
1:e000016, 2016 ]. En la mayoría de los casos es posible ofrecer un
tratamiento oncológico adecuado a la paciente con mantenimiento del
bienestar fetal, pero la falta de experiencia y conocimiento de los
profesionales sanitarios en el manejo de estos casos puede dar lugar a
retrasos en el diagnóstico, tratamiento incorrecto y mayor riesgo materno
-fetal [Mundo J Oncol 10:28, 2019 ]. Cabe señalar que, en muchos casos, los
retrasos en el diagnóstico y tratamiento pueden ser más perjudiciales que
los posibles riesgos fetales. Se debe considerar la edad gestacional al
determinar los posibles efectos nocivos de las intervenciones diagnósticas
y terapéuticas sobre el bienestar fetal [ Best Pract Res Clin Obstet
Gynaecol 33:2, 2016 ].

— Hasta 10 días después de la concepción: fase de “todo o nada”. Si el


embrión sobrevive a la exposición a agresiones externas, es probable que
no ocurra ningún efecto adverso.

— 10 días a 8 semanas: período de organogénesis. La exposición a


agentes teratogénicos puede causar malformaciones fetales importantes.

— 2° y 3° trimestres: período de crecimiento fetal. Algunos sistemas


fetales en crecimiento siguen siendo vulnerables, como el sistema
nervioso central (SNC) y el sistema hematológico. Pueden ocurrir
complicaciones como el parto prematuro y la restricción del crecimiento
intrauterino con la exposición a agresiones externas.
El embarazo y el impacto en la evolución del
cáncer
En general, las características patológicas y el pronóstico del cáncer
diagnosticado durante el embarazo son similares a las de las pacientes no
embarazadas, con HR = 1,03 para el grupo de embarazadas (IC 95 %: 0,86-
1,22) y HR = 1,02 (IC 95 %: 0,86). -1,22) para el grupo lactante. Estudios previos
sugieren que las mujeres diagnosticadas de mama (HR=1,95; IC 95%: 1,36-
2,78) y de ovario (HR=2,23; IC 95%: 1,05-4,73) durante la lactancia tenían un
mayor riesgo de muerte por causa específica. El riesgo de mortalidad por
causas específicas se redujo en mujeres que experimentaron un
embarazo después del diagnóstico de cáncer (HR=0,49; IC del 95 %: 0,41-
0,9) [ J Clin Oncol 27:45, 2009]. Sin embargo, un estudio de cohortes que
incluyó mujeres diagnosticadas con cáncer de mama en el embarazo
después de la década de 2000 evaluó el pronóstico materno de pacientes
que recibieron quimioterapia (QT) durante el embarazo y mostró una
supervivencia libre de progresión (PFS) y una supervivencia general (SG)
similares para mujeres embarazadas y embarazadas. mujeres no
embarazadas (SLP: HR=1,0; IC 95 %: 0,82-1,27; p=0,83; OS: HR=1,08; IC 95 %:
0,81 -1,45; p=0,59). En un análisis de subgrupos de pacientes que recibieron
más del 60 % de QT durante el embarazo, la supervivencia fue comparable
a la de mujeres no embarazadas (SLP: HR=0,81; IC 95 %: 0,62-1,06; p =0,13;
SG: HR= 0,85; IC del 95 %: 0,58-1,23; p=0,39) [ J Clin Oncol 39:abstr 515, 2021 ].

Interrupción electiva del embarazo


El mantenimiento del embarazo no parece estar asociado con un peor
pronóstico, siempre que el tratamiento del cáncer se realice
correctamente. La interrupción puede ser discutida en pacientes con
enfermedad metastásica, especialmente en casos de crisis visceral en el
primer trimestre, en los que existe riesgo inminente de muerte para la
madre o en casos de enfermedad avanzada e imposibilidad de
tratamiento adecuado, en vista de los riesgos al paciente feto. Se debe
realizar un abordaje multidisciplinar del paciente, con apoyo y
seguimiento del equipo de psicología [ ESMO Open 1:e000016, 2016 ].

SÍNTOMAS Y PRESENTACIÓN
Los cambios fisiológicos del embarazo suponen un reto para el
diagnóstico oncológico, con frecuentes retrasos, especialmente en los
casos de cáncer de mama. El diagnóstico temprano es esencial para lograr
mejores resultados, pero lamentablemente se retrasa por factores que
confunden su alcance [ Best Pract Res Clin Obstet Gynaecol 33:19,
2016 ]. Por otro lado, el embarazo provoca cambios tisulares que afectan a
condiciones benignas y pueden conducir a cambios sugestivos de
malignidad. El patólogo siempre debe estar informado del estado de
embarazo de la paciente para minimizar los errores de diagnóstico [ Eur J
Cancer 46:3158, 2010 ].

COMO ESTADIAR
Es necesario realizar pruebas de estadificación, según el sitio primario,
respetando las particularidades relacionadas con el embarazo, lo que
minimiza los riesgos maternos y fetales [ Cancer Biol Med 15:6, 2018]. Se
deben preferir las técnicas de imagen sin exposición a la radiación
ionizante, como la ecografía y la resonancia magnética (RM) sin contraste,
durante el embarazo. La ecografía suele estar ampliamente disponible y
representa el método de elección para la evaluación inicial de una masa
abdominal o lesiones en la mama, la cabeza y el cuello y los tejidos blandos,
y se puede realizar en cualquier trimestre del embarazo. La RM sin
contraste debe realizarse preferentemente después del primer trimestre
del embarazo, lo que permite una valoración más detallada de órganos y
tejidos, con valoración funcional de las lesiones mediante la técnica de
difusión, que se considera segura durante el embarazo. Existen reportes
en la literatura de la asociación causal del uso de gadolinio durante el
embarazo con malformaciones fetales, aborto y muerte neonatal, [JAMA
316:952, 2016 ; Best Pract Res Clin Obstet Gynaecol 33:19, 2016 ]. La
resonancia magnética de cuerpo entero sin contraste es una prueba de
estadificación importante, que contribuye a la evaluación del tórax, el
abdomen y los huesos, y debe realizarse preferiblemente después del
período de organogénesis [ Radiographics 32:897, 2012 ; Cáncer Biol Med
15:6, 2018]. Se debe considerar el uso de técnicas de imagen con radiación
ionizante cuando sea esencial para el manejo del caso, pero se debe
monitorear la exposición a la radiación, con una dosis fetal acumulada
máxima recomendada de 100 mGy. Con este umbral de dosis, existe un
riesgo aproximadamente un 1 % mayor de malformación fetal y cáncer
infantil. Varios métodos de imagen con radiación ionizante se consideran
seguros, con exposición fetal por debajo de la dosis máxima recomendada
si se usa con protección abdominal adecuada, y no deben evitarse cuando
sea necesario, como la radiografía simple no abdominal, la mamografía y
la tomografía computarizada (TC) de cabeza y cuello. La tomografía
computarizada por emisión de positrones (PET-CT), la gammagrafía ósea,
las TC de abdomen y pelvis y los exámenes guiados por fluoroscopia
cruzan el umbral de seguridad fetal, deberían ser evitados. También se
debe evitar el uso de contraste yodado en mujeres embarazadas
[Tratamiento del cáncer Rev 27:1, 2001 ; Lancet Oncol 6:328, 2005 ; J Vasc
Interv Radiol 23:19, 2012 ; Mundo J Oncol 10:28, 2019 ].

Marcadores tumorales
Los niveles séricos de marcadores tumorales pueden tener menor
sensibilidad y especificidad como resultado de cambios fisiológicos
durante el embarazo. Los niveles de CA 15-3, SCC (antígeno de carcinoma
de células escamosas), CA 125 y alfa-fetoproteína están aumentados en el
embarazo, y no se recomienda su uso. Otros marcadores, como ACE, CA
19-9, DHL y la hormona antimulleriana, no se ven alterados por el estado
del embarazo y pueden ser útiles. Como excepciones, la inhibina B está
elevada en el último trimestre del embarazo y la DHL es un marcador de
cambios hipertensivos en el embarazo [ BMC Med 10:86, 2012 ; Mundo J
Oncol 10:28, 2019 ].

TRATAMIENTO
Cirugía
La cirugía materna durante el embarazo puede estar asociada con un
mayor riesgo de parto prematuro y perfusión uteroplacentaria alterada,
con un mayor riesgo de hipoxia, daño neurológico y muerte fetal. El
segundo trimestre del embarazo es el período más propicio para realizar
procedimientos quirúrgicos, debido al menor riesgo de aborto
espontáneo y parto prematuro [ Asian Pac J Clin Oncol 15:296,
2019 ]. Cuando esté indicado, el procedimiento quirúrgico no debe
posponerse debido al riesgo de progresión de la enfermedad materna y
compromiso fetal por malignidad. Una revisión sistemática de 12 452
mujeres que se sometieron a procedimientos quirúrgicos no obstétricos
durante el embarazo no identificó un mayor riesgo de aborto espontáneo
o defectos fetales [ Am J Surg 190:467, 2005]. Se recomienda reducir el
tiempo bajo anestesia general, minimizar la dosis y la concentración de los
agentes utilizados y evitar la anestesia por inhalación [ Am J Obstet
Gynecol 218:98, 2018 ].

Radioterapia
La exposición a la radiación se asocia con teratogénesis, aborto
espontáneo, retraso mental y mayor riesgo de malignidad en niños y
adultos después de la exposición intrauterina. Por lo tanto, se recomienda
esperar al final del embarazo para el tratamiento con radioterapia. Hay, sin
embargo, reportes de radioterapia durante el embarazo sin asociación con
efectos adversos fetales en casos de cáncer de mama, linfoma, cáncer de
cabeza y cuello y SNC, probablemente relacionados con una protección
uterina eficiente y baja dispersión de la radiación con el uso de tecnologías
más modernas. Los eventos adversos relacionados con la exposición a la
radiación durante el embarazo dependen del período de desarrollo fetal y
de la dosis de radiación. Las dosis inferiores a 0,1 Gy no parecen afectar el
desarrollo fetal. En el primer trimestre (organogénesis), los riesgos de
malformaciones, la restricción del crecimiento de la cabeza y el retraso
mental son mayores. En el segundo trimestre, los principales efectos
adversos son similares a los del primer trimestre. En el tercer trimestre, el
riesgo de retraso mental y restricción del crecimiento es menor, pero aún
puede ocurrir [Crit Rev Oncol Hematol 136:13, 2019 ].

TRATAMIENTO SISTÉMICO
Consideraciones generales
Las indicaciones de tratamiento sistémico durante el embarazo son
similares a las de las pacientes diagnosticadas de cáncer fuera de este
período, es decir, se consideran los mismos factores de riesgo. Sin
embargo, al proponer el tratamiento a la mujer embarazada, se debe
evaluar cuidadosamente la seguridad para el feto. El gran reto es no
infratratar a la paciente oncológica y elegir el tratamiento que mejor se
adapte al trimestre gestacional para preservar la salud fetal. Es imperativo
que el oncólogo tenga un conocimiento profundo de los medicamentos
contra el cáncer seguros durante el embarazo. No se recomienda el
tratamiento sistémico en el primer trimestre del embarazo, considerando
el riesgo de teratogenicidad fetal, prematuridad y aborto espontáneo. En
los casos de pacientes diagnosticados de enfermedad avanzada o
metastásica en el primer trimestre, se debe considerar la gravedad de la
enfermedad de base, riesgo para la salud materna, necesidad de
tratamiento específico y mantenimiento o no del embarazo. A partir del
segundo trimestre del embarazo, existen datos de estudios retrospectivos
así como series de casos que sugieren la seguridad del uso de ciertos
agentes antineoplásicos. [Ann Intern Med 94:547, 1981 ; J Clin Oncol
23:4192, 2005 ; Tratamiento del cáncer Rev 41:301, 2015]. En su mayoría, los
agentes antineoplásicos, la terapia hormonal (HT) y los agentes biológicos
utilizados en el tratamiento del cáncer se consideran categoría D para el
tratamiento de mujeres embarazadas con cáncer por la FDA y deben
evitarse. Un estudio observacional multicéntrico que evaluó a niños que
estuvieron expuestos a QT en el período prenatal mostró que la exposición
fetal a QT no se asoció con un aumento de la morbilidad cardíaca o
auditiva, ni con daño a la salud general, incluidos los hitos en el
crecimiento y desarrollo infantil, en comparación con la población en
general. Es importante destacar que, en este estudio, la prematuridad fue
común entre los fetos que fueron expuestos a quimioterapia, y este fue un
factor asociado con el deterioro cognitivo y del desarrollo. Por lo tanto, el
parto prematuro iatrogénico debe evitarse tanto como sea posible.
[Lancet Oncol 19:337, 2018 ]. Otro estudio de casos y controles corrobora
estos hallazgos [ N Engl J Med 373:1824, 2015 ]. Los cambios fisiológicos del
embarazo, tales como los cambios en la función renal y hepática, el
volumen plasmático y el efecto del tercer espacio relacionado con el
líquido amniótico, pueden alterar la farmacocinética de los agentes
antineoplásicos [ Semin Oncol 16:337, 1989 ; Best Pract Res Clin Obstet
Gynaecol 33:86, 2016 ]. El cálculo de la dosis de quimioterapia en pacientes
embarazadas también debe basarse en el área de superficie corporal, con
ajustes basados en el aumento de peso continuo. Deben evitarse las
reducciones de dosis debido a la probable mayor eliminación de fármacos
durante el embarazo [ Lancet Oncol 5:283, 2004 ].

Amamantamiento
Las mujeres que se someten a un tratamiento oncológico activo no deben
amamantar. Hay informes de excreción de fármacos citotóxicos en la
leche materna con riesgo de toxicidad neonatal, como leucopenia y
trombocitopenia. Todavía son escasos los datos sobre la seguridad del uso
de la terapia sistémica con nuevas clases de medicamentos, y se debe
evitar la lactancia [ Cancer Treat Rev 39:207, 2013 ; Medicina de la Lactancia
Materna 12:91, 2017 ].

Quimioterapia
La mayoría de los agentes quimioterapéuticos pueden atravesar la barrera
placentaria (peso molecular < 500 kDa) y llegar al feto [ Clin
Pharmacokinet 43:487, 2004 ]. Los riesgos para el feto asociados con el uso
de quimioterapia durante el embarazo dependen de los fármacos
utilizados y de la edad gestacional [ Best Pract Res Clin Obstet Gynaecol
33:86, 2016 ]. En el primer trimestre del embarazo (período de
organogénesis), el uso de agentes quimioterapéuticos está
contraindicado debido al alto riesgo de teratogenicidad, con mayor riesgo
de anomalías congénitas, anomalías cromosómicas, prematuridad y
aborto espontáneo. El riesgo de malformaciones es mayor con los
fármacos combinados que con el uso de un solo agente [ Ann Oncol 15:146,
2004 ; Defectos de nacimiento Res A Clin Mol Teratol 94:626, 2012; Best
Pract Res Clin Obstet Gynaecol 33:86, 2016 ]. Algunos medicamentos se
consideran seguros durante el segundo y tercer trimestre [ J Clin Oncol
23:4192, 2005 ]. El riesgo de malformación fetal es del 3-4% en la población
general, y el tratamiento con fármacos considerados seguros en los
últimos trimestres se asoció con un riesgo bajo (alrededor del 3%), pero
existe el riesgo de restricción del crecimiento intrauterino (RCIU),
prematuridad y bajo peso al nacer hasta en un 50% de los casos. Aunque
el uso de quimioterapia durante el segundo y tercer trimestre del
embarazo se considera seguro, debe evitarse durante las 3 semanas
previas a la fecha prevista del parto para reducir el riesgo de
mielosupresión fetal y complicaciones durante el parto como hemorragia,
sepsis y muerte neonatal. [Defectos de nacimiento Res A Clin Mol Teratol
94:626, 2012 ; Best Pract Res Clin Obstet Gynaecol 33:86, 2016 ].

Antraciclinas: El uso de doxorrubicina y epirrubicina se considera


seguro en el segundo y tercer trimestre del embarazo. Debe evitarse el uso
de idarubicina debido al riesgo de teratogenicidad. Un estudio
prospectivo del MD Anderson Cancer Center (MDACC) que incluyó a 87
pacientes con cáncer de mama en el embarazo tratadas, en el segundo o
tercer trimestre, con el régimen FAC la terapia adyuvante o neoadyuvante
ha demostrado seguridad en el uso de antraciclinas en el embarazo. La
gran mayoría de los recién nacidos no presentó complicaciones
perinatales, ocurriendo complicaciones en solo el 3% de ellos, tasa similar
a la población general. Hubo un caso de síndrome de Down, un caso de
reflujo urotelial y un caso de pie zambo congénito. No hubo aborto
espontáneo ni muerte perinatal [ Breast Cancer Res 16:500, 2014 ; BMC
Cancer 17:777, 2017 ].

Taxanos: El uso de taxanos en el segundo y tercer trimestre del embarazo


parece ser seguro. Una revisión sistemática de la literatura que incluye 16
estudios (50 embarazos) que usan taxanos durante el embarazo informó
un 76,7 % de los resultados con el nacimiento de niños completamente
sanos [ Clin Breast Cancer 13:16, 2013 ]. Además, otros estudios describen su
uso en cáncer de ovario, cervical y de pulmón de células no pequeñas con
resultados prometedores [ Arch Gynecol Obstet 283:97, 2011 ; Arco Gynecol
Obstet 284:779, 2011 ; J Thorac Oncol 4:559, 2009 ].

Platino: los datos sugieren seguridad con el uso de derivados de platino


en el segundo y tercer trimestre del embarazo. En un metaanálisis que
incluyó 39 estudios, con 88 mujeres con cáncer de cuello uterino, la terapia
neoadyuvante basada en platino se consideró una opción favorable para
el tratamiento de pacientes, con recién nacidos completamente normales
en el 80,7 % de los casos [ Drug Des Devel Ther 13:79, 2018 ]. La revisión de
la literatura que usa platino combinado con taxanos para el tratamiento
del cáncer de ovario también informó datos de seguridad de la
combinación [ Int J Clin Pharmacol Ther 55:753, 2017 ]. En una revisión
sistemática de 43 mujeres tratadas con platino durante el embarazo (36
recibieron cisplatino , 6 recibieron carboplatino y 1 recibió ambos
fármacos), 8 pacientes expuestas a cisplatino tuvieron algún impacto en
el embarazo o deterioro de la salud fetal, 3 tenían RCIU, 1 polihidramnios, 1
ventriculomegalia y 3 casos de prematuridad. Ninguno de los pacientes
tratados con carboplatino experimentó complicaciones [ Cancer 113:3069,
2008 ].

Ciclofosfamida: Se considera seguro para el tratamiento de mujeres


embarazadas en el segundo o tercer trimestre del embarazo. La mayoría
de los estudios evaluaron el uso de ciclofosfamida en regímenes
asociados con antraciclinas [ Rev Assoc Med Bras 59:174, 2013 ; Cáncer de
mama Res 16:500, 2014 ].

Fluoruracilo: Hay informes de seguridad del tratamiento del cáncer en


mujeres embarazadas con este agente de quimioterapia después del
primer trimestre del embarazo. Los datos provienen de estudios que
evaluaron la seguridad y eficacia de la combinación de antraciclinas
más ciclofosfamida con o sin fluorouracilo (régimen FAC ) en el
tratamiento del cáncer de mama en el embarazo [ Breast Cancer Res
16:500, 2014 ; Cáncer 107:1219, 2006 ].

Metotrexato: el uso de metotrexato conlleva un alto riesgo de


teratogenicidad y debe evitarse durante el embarazo [ Birth Defects Res A
Clin Mol Teratol 94:626, 2012 ].

Inhibidores de la tirosina quinasa


Los inhibidores de la tirosina cinasa ( tyrosine kinase inhibitors - TKI)
actúan de forma selectiva sobre los receptores de tirosina cinasa (TKR) que
se activan en algunas neoplasias. Aunque se considera selectivo para el
TKR de interés, los sitios de unión de ATP están muy conservados, lo que
permite la unión a TKR no específicos [ Sci Rep 9:2535, 2019 ]. La
transferencia placentaria de imatinib al feto está documentada en
humanos. La tasa de malformación después de la exposición en el primer
trimestre es de alrededor del 11%, sin reportes en el segundo y tercer
trimestre del embarazo. Por lo tanto, se debe evitar el uso de imatinib en
el primer trimestre del embarazo. Datos del uso de imatinib después del
primer trimestre están restringidas a un pequeño número de pacientes, y
su uso debe evaluarse cuidadosamente en cada caso [ Blood 111:5505,
2008 ; Cancer Treat Rev 41:301, 2015 ]. Los TKI de segunda
generación, dasatinib y nilotinib , parecen tener un perfil de toxicidad
similar al de imatinib durante el embarazo [ Anticancer Drugs 23:754,
2012 ; JHematol Oncol 2:42, 2009 ]. Sin embargo, se debe evitar su uso
hasta que haya más datos disponibles [ Cancer Treat Rev 41:301,
2015 ]. La vía del receptor del factor de crecimiento epidérmico– EGFR)
participa en la proliferación y diferenciación celular y juega un papel
importante en varias etapas del desarrollo embrionario. Los informes de
uso involuntario de erlotinib (2 casos), gefitinib (1 caso), erlotinib seguido
de gefitinib (1 caso) y crizotinib (2 casos) durante el embarazo no
mostraron anomalías fetales. Sin embargo, el uso de estos medicamentos,
así como los inhibidores de EGFR de segunda y tercera generación, no se
recomienda durante el embarazo debido a la escasez de datos [ J Adv Res
7:571, 2016 ]. Ácido transretinoico ( ATRA) es esencial para el desarrollo
embrionario, regulando la morfogénesis, la proliferación celular y la
diferenciación [ Mol Med Rep 16:5915, 2017 ]. Su transferencia placentaria
está bien documentada en humanos y está contraindicada en el primer
trimestre del embarazo [ Br J Haematol 94:699, 1996 ; Acta Haematol
114:167, 2005 ]. Su uso parece ser seguro después del primer trimestre del
embarazo [ Cancer Treat Rev 41:301, 2015 ]. El interferón -alfa no atraviesa
la barrera placentaria en cantidades significativas y se considera una
opción terapéutica plausible para su uso durante el embarazo [ Reprod
Toxicol 33:265, 2012 ].

Agentes biológicos
Los agentes biológicos, como los anticuerpos monoclonales, son
moléculas de alto peso molecular que requieren un transporte placentario
activo para llegar al feto, lo que no suele ocurrir hasta la semana 14 de
gestación, lo que sugiere que la exposición a estos agentes en el primer
trimestre presenta un riesgo menor. efectos adversos, mientras que los
riesgos son mayores en el segundo y tercer trimestre del embarazo, pero
hay escasez de datos sobre el uso de estos agentes en el primer trimestre,
por lo que se debe tener precaución y evitar su uso [ Cancer Treat Rev
41:301, 2015 ]. La tasa de eventos adversos con el uso de anticuerpos
monoclonales es de hasta un 40 % [ Birth Defects Res A Clin Mol Teratol
94:626, 2012 ].

Terapia anti-HER-2: El uso de trastuzumab en el tratamiento del cáncer


de mama HER-2 positivo en el embarazo está contraindicado en cualquier
trimestre. Su uso se ha asociado con casos de oligohidramnios, que
pueden estar relacionados con casos de hipoplasia pulmonar, anomalías
esqueléticas e incluso muerte neonatal [ Cancer Treat Rev 41:301, 2015 ]. En
un meta-análisis de 17 estudios, la aparición de oligohidramnios fue del 61,1
%, con una mayor incidencia de prematuridad [ J Clin Oncol 36:abstr 10065,
2018 ]. De manera similar, el uso de lapatinib [ Clin Breast Cancer 7:339,
2006 ], pertuzumab [ Anticancer Drugs 29:810, 2018 ] y ado-trastuzumab
emtansine[ Ther Adv Med Oncol 6:202, 2014 ] también está contraindicado
en el embarazo.

Rituximab: existe un riesgo potencial de citopenia neonatal y posneonatal


con el uso de rituximab durante el embarazo, pero se deben sopesar los
beneficios de usar este fármaco en el control de la enfermedad y la SG en
portadoras de linfomas de células B durante el embarazo [ Cancer Treat
Rev 41:301, 2015 ; Front Pharmacol 12:621247, 2021 ].

Inhibidores de EGFR: Cetuximab y panitumumab son anticuerpos


monoclonales anti-EGFR. A pesar de ser moléculas grandes, ambas
atraviesan la barrera placentaria. La vía EGFR es importante para el
crecimiento fetal y el mantenimiento del embarazo, con teratogenicidad
reportada en modelos animales. No existen datos sobre la seguridad de su
uso en humanos durante el embarazo, y debe evitarse [ Oncologist 21:563,
2016 ; Cancer Treat Rev 41:301, 2015 ].

Agentes antiangiogénicos: debido a la escasez de datos en la literatura,


se debe evitar el uso de antiangiogénicos durante el embarazo [ Cancer
Treat Rev 41:301, 2015 ]. Bevacizumab es un anticuerpo monoclonal que
inhibe la vía del receptor del factor de crecimiento endotelial vascular
(VEGF). En un modelo animal, hay transferencia placentaria dependiente
de la dosis en todas las etapas del embarazo [ Birth Defects Res B Dev
Reprod Toxicol 95:363, 2012 ], con hallazgos de eventos adversos fetales [ J
Matern Fetal Neonatal Med 27:1744, 2014 ] . También hay datos de
teratogenicidad fetal en modelos animales que
usan sunitinib y sorafenib anti-VEGFR multi-TKI [Defectos de nacimiento
Res B Dev Reprod Toxicol 86:204, 2009 ]. No hay datos sobre el uso de estos
medicamentos durante el embarazo en humanos.

Inmunoterapia
Durante el embarazo, las vías de señalización de puntos de control, en
particular el eje PD-1/PDL-1, son importantes para la tolerancia inmunitaria
fetal. La evidencia sugiere que los inhibidores de los puntos
de control inmunitarios pueden atravesar la barrera placentaria y causar
daño al feto y un mayor riesgo de aborto espontáneo [ Cancer Treat Rev
41:301, 2015 ; Best Pract Res Clin Obstet Gynaecol 33:86, 2016 ; Semín Oncol
45:164, 2018 ].

Terapia hormonal
Estudios con el uso de tamoxifeno durante el embarazo en humanos
reportan 16 casos de nacidos vivos con malformaciones congénitas y 122
de nacidos vivos sin malformaciones. De estos últimos, se informaron 85
casos en el estudio de prevención, sin que se informaran hallazgos de
anomalías fetales [ Lancet 342:168, 1993 ]. La base de datos de seguridad
de AstraZeneca informó 11 casos de malformaciones congénitas en 44
nacidos vivos, 12 abortos espontáneos, 17 interrupciones sin hallazgos de
anomalías fetales, 6 interrupciones con anomalías fetales, 1 muerte fetal
sin anomalía fetal y 2 muertes fetales con anomalías fetales, además de 57
embarazos con resultado desconocido [ Oncólogo 16:1547, 2011]. Aunque
representan casos con una descripción retrospectiva, sin establecer una
relación causal entre el uso de tamoxifeno en el embarazo y el resultado
gestacional, se considera prudente suspender el tamoxifeno antes de
planificar un embarazo y evitar su uso durante todas las etapas del
embarazo. En los casos de embarazo involuntario usando tamoxifeno , se
debe considerar y discutir el riesgo materno de recaída debido a la
interrupción temprana del tratamiento. Los niños que han estado
expuestos al tamoxifeno intrauterino deben someterse a un seguimiento
a largo plazo [ Oncologist 16:1547, 2011 ; Cáncer J del sur de Asia 5:209,
2016]. No hay datos de seguridad sobre el uso de inhibidores de la
aromatasa durante el embarazo, y debe evitarse, al igual que el uso de
análogos de LHRH.

Terapia de suporte
Bisfosfonatos: Los datos de la literatura actual sugieren que el uso de
bisfosfonatos durante el embarazo no está asociado con un mayor riesgo
de efectos teratogénicos fetales, pero puede haber una mayor tasa de
complicaciones neonatales. En caso de exposición fetal a uno de estos
agentes, se recomienda monitorear la posible hipopotasemia neonatal y
síntomas cardíacos y neuromusculares [ Am J Health Syst Pharm 71:2029,
2014 ; Osteoporos Int 30:221, 2019 ].

Denosumab: El uso de denosumab , un anticuerpo monoclonal humano


que inhibe el ligando RANK, utilizado en el tratamiento de metástasis
óseas, está contraindicado en el embarazo y la lactancia. Los estudios en
animales muestran un aumento de las tasas de mortinatalidad y
mortalidad neonatal [ Reprod Toxicol 42:27, 2013 ; Biochem Biophys Res
Commun 491:614, 2017 ].

Antieméticos: el uso
de meclozina , difenhidramina y metoclopramida se considera seguro
durante el embarazo, incluido el primer trimestre [ N Engl J Med 360:2528,
2009 ; JAMA 316:1392, 2016 ]. El uso de ondansetrón durante el embarazo
se considera seguro, pero debe evitarse en el primer trimestre del
embarazo, ya que se asocia con un mayor riesgo de paladar hendido
[ JAMA 320:2429, 2018 ]. El uso de dexametasona puede afectar la
osteogénesis fetal. Debe evitarse su uso crónico en el embarazo [ Curr
Pharm Des 20:5430, 2014]. Debe evitarse el uso de antagonistas de la
neuroquinina 1 (NK1) durante el embarazo debido a la falta de datos de
seguridad.

Factor estimulante de colonias: el uso de factores estimulantes de


colonias parece ser seguro durante el embarazo. Usando datos
del Registro de Cáncer y Embarazo de la Universidad de Cooper, un
estudio evaluó retrospectivamente el papel y los posibles efectos adversos
de los factores estimulantes de colonias en pacientes diagnosticadas con
cáncer en el embarazo tratadas con quimioterapia y que usaron
o filgrastim o pegfilgrastim durante el tratamiento de quimioterapia. De
las 176 gestantes que recibieron quimioterapia, 142 no usaron factor
estimulante de colonias y 34 lo usaron. No se observó un aumento en la
incidencia de teratogenicidad, abortos o partos prematuros con el uso del
factor estimulante de colonias. Además, las pacientes que usaron estos
medicamentos tenían un menor riesgo de infecciones oportunistas
posparto. Los datos son limitados y el número de pacientes evaluados es
pequeño [ J Cancer Ther 3:157, 2012 ; Obstet Gynecol 125:197,
2015 ]. Recientemente, la Red Internacional de Cáncer, Infertilidad y
Embarazo (INCIP) publicó una serie de casos que evaluó a 42 pacientes
que recibieron factor estimulante de colonias ( G-CSF). como parte del
tratamiento del cáncer durante el embarazo. En su mayor parte, las
mujeres se vieron afectadas por el cáncer de mama. Las principales
indicaciones para el uso de G-CSF fueron: protocolo QT de dosis densa (67
%), protocolo de terapia multimedicamentosa (17 %), neutropenia febril (12
%) y prevención de la neutropenia perinatal (5 %). Las complicaciones
observadas en la serie de casos son similares a los datos de otras
publicaciones sobre mujeres embarazadas que reciben quimioterapia, lo
que en un principio no sugiere un mayor riesgo de malformaciones y
prematuridad relacionado con el uso del factor estimulante de colonias
[ Cancers (Basel) 13:1214 , 2021 ].

You might also like