A paradigm shift in therapeutic vaccination of cancer patients: the need to apply therapeutic...

download A paradigm shift in therapeutic vaccination of cancer patients: the need to apply therapeutic vaccination strategies in the preventive setting

If you can't read please download the document

Transcript of A paradigm shift in therapeutic vaccination of cancer patients: the need to apply therapeutic...

  • A paradigm shift in therapeuticvaccination of cancer patients:

    the need to apply therapeutic

    vaccination strategies in thepreventive setting

    Summary: An extraordinary variety of potential therapeutic vaccinestrategies directed against a wide variety of tumor antigens has beenexplored in clinical trials. To date, none of these cancer immunotherapieshave been approved by the Food and Drug Administration for use inhumans. A significant problem is that the vast majority of such clinicaltrials are carried out in patients with advanced or metastatic cancer. Theimmune systems of these patients are considerably compromised as aresult of tumor- and treatment-mediated immunosuppression. Even incases where patients are immunized in the adjuvant setting, where there isminimal residual disease, vaccines directed against tumor-associatedantigens have failed to mediate eradication of tumors in the overwhelmingmajority of cases. Recently, we and others have experimented withadministering therapeutic cancer vaccines in the preventive setting. Thisis achieved by vaccinating at the earliest possible stage of carcinogenesis.These studies have demonstrated that early vaccination is extremelyeffective in eliciting an anti-tumor immune response that leads tounprecedented improvements in the survival of mice that spontaneouslydevelop cancer. Certain human cancers, notably prostate adenocarcinomaand cervical cancer, can currently be detected at very early stages ofcarcinogenesis. Therapeutic vaccines are available for these diseases,opening up the possibility of administering vaccinations early to patientsdiagnosed with pre-malignant lesions to halt disease progression. Inaddition, new technologies have become available in the past decade thatwill soon yield very sensitive and specific diagnostic tests for a plethora ofother cancers. Earlier detection of these cancers, combined with existingvaccines directed against them, will soon make them targets for therapeu-tic vaccination in the preventive setting. The ability to immunize patients atthe very earliest stages of carcinogenesis, when they have fully competentimmune systems, has the potential to cause a paradigm shift in howtherapeutic cancer vaccines are tested and used clinically.

    Keywords: therapeutic cancer vaccines, immunotherapies, tumor immunity, vaccination,cancer, cancer prevention

    Introduction

    Immunotherapy has long been proposed as a method of

    treating cancer. Hundreds of animal studies have shown that a

    myriad of cancers can be treated specifically, safely, and

    Andrew Gray

    Adam B. Raff

    Maurizio Chiriva-Internati

    Si-Yi Chen

    W. Martin Kast

    Immunological Reviews 2008

    Vol. 222: 316327

    Printed in Singapore. All rights reserved

    r 2008 The Authors

    Journal compilationr 2008 Blackwell Munksgaard

    Immunological Reviews0105-2896

    Authors addresses

    Andrew Gray1, Adam B. Raff1, Maurizio Chiriva-Internati2,3,

    Si-Yi Chen1,4, W. Martin Kast1,4,5

    1Norris Comprehensive Cancer Center, University of

    Southern California, Los Angeles, CA, USA.2Department of Molecular Microbiology & Immunology,

    Texas Tech University Health Sciences Center and

    Southwest Cancer Treatment and Research Center,

    Lubbock, TX, USA.3Division of Hematology & Oncology, Texas Tech

    University Health Sciences Center and Southwest Cancer

    Treatment and Research Center, Lubbock, TX, USA.4Department of Microbiology & Immunology, University

    of Southern California, Los Angeles, CA, USA.5Department of Obstetrics & Gynecology, University of

    Southern California, Los Angeles, CA, USA.

    Correspondence to:

    W. Martin Kast

    University of Southern California

    1450 Biggy Street, NRT 7507

    Los Angeles, CA 90033, USA

    Tel.:11 323 442 3870

    Fax: 11 323 442 7760

    e-mail: [email protected]

    Acknowledgements

    This review is partially based on studies that were sup-

    ported by the Margaret E. Early Medical Research Trust and

    NIH training grant T32 GM 067587 (Gray). W. Martin Kast

    holds the Walter A. Richter Cancer Research Chair.

    316

  • effectively using a wide variety of immunotherapeutic strate-

    gies. The extraordinary successes in eliciting immune re-

    sponses against tumor-associated antigens (TAAs) that are

    capable of mediating eradication of tumors in animal models

    of cancer have led to an explosion in clinical trials designed to

    test the viability of translating similar strategies to treating

    patients. However, no such immunotherapies have been ap-

    proved by the Food and Drug Administration for human use.

    A significant problem is that virtually all such vaccines have

    been tested in terminally ill cancer patients who have failed

    other therapeutic strategies. These patients generally have large

    and/or metastatic tumors. Patients with advanced cancers are

    not good candidates for immunotherapy, because they are

    immunocompromised as a result of previous treatments and

    the immune evasion mechanisms of their tumors. The field has

    attempted to circumvent this problem by two main

    approaches: (i) patients with minimal residual disease (e.g., in

    patients that have undergone surgical, radiological, and/or

    chemotherapeutic intervention) have been vaccinated in the

    hopes that the immune suppression commanded by the tumor

    will be diminished if tumor volume is reduced, and (ii) the

    mechanisms of tumor immune evasion have been individually

    targeted and eliminated or attenuated in an effort to allow the

    anti-tumor immune response elicited by vaccination to

    proceed more efficiently. These approaches have met with

    limited success. As discussed herein, large tumors can and do

    significantly alter the immune system, and these changes can

    persist even after the tumor has been debulked. Furthermore,

    the complex immunosuppressive networks developed by

    advanced cancers have multiple, often redundant, mechanisms

    of limiting the anti-tumor immune response. As a result,

    targeting one or a few of these immune evasion mechanisms

    within a tumor rarely elicits an improvement in the clinical

    response to vaccination because other immunosuppressive

    mechanisms remain that can blunt the anti-tumor immune

    response induced by vaccination.

    Given these considerations, it is time for a paradigm shift in

    how therapeutic vaccination protocols are tested and applied in

    the clinic. Advances in screening and detection of cancer are

    arguably the greatest victories in the so-called war on cancer

    to date. The improved cancer detection mechanisms that are

    available and that will be coming online over the next decade

    will undoubtedly allow the medical field to detect cancers at

    their very earliest stages. We believe that this provides us a new

    opportunity to apply therapeutic cancer vaccines preventatively

    during the earliest possible stages of carcinogenesis. Here we

    will propose that very early detection of cancer will allow us to

    intervene with tumor immunotherapies before the presence of

    the tumor and before the use of conventional anticancer

    treatments have manifested changes to the immune system of

    the patient. We summarize our own recent studies and those of

    others that show that therapeutic vaccinations in the very early

    stages of carcinogenesis can induce an extremely effective, long

    lasting, and safe immune protection against cancer development.

    Most therapeutic cancer vaccine clinical trials to datehave been carried out in end-stage patients and haveyielded limited clinical benefit

    A plethora of therapeutic vaccination strategies have undergone

    clinical trials in recent years. These have included vaccines

    based on tumor cell lysates, peptides of TAAs, recombinant

    proteins, dendritic cells (DCs) (mature and immature) loaded

    with all of the above, DCs transfected with tumor RNA or with

    DNA encoding TAAs, whole tumor cells, recombinant viral

    vectors expressing TAAs, and direct immunization with plas-

    mid DNA encoding TAAs. In addition, many of these strategies

    have been combined with administration of other immuno-

    modulatory molecules including but not limited to interleukin-

    2 (IL-2), granulocyte macrophage colony-stimulating factor

    (GM-CSF), and costimulatory molecules. Despite these many

    and varied attempts at eliciting an anti-tumor immune re-

    sponse, very few are capable of doing so, and so far none have

    proven consistently capable of eradicating an existing tumor.

    Virtually all therapeutic cancer vaccines that have undergone

    clinical trials have been tested in patients with advanced or

    metastatic cancer who have failed all conventional treatment

    options. This philosophy of clinical trial design is exemplified

    in the case of prostate cancer immunotherapy. Some recent

    trials of therapeutic prostate cancer vaccines have been

    summarized in Table 1 (115). The majority of these trials has

    been conducted in patients with metastatic hormone-

    refractory prostate cancer (HRPC), for whom only palliative

    treatment is available. From an ethical standpoint, these

    individuals make good candidates for experimental therapies.

    However, these trials have produced very little positive data.

    Measurable immunological responses to vaccination, often

    measured by enzyme-linked immunospot assay (ELISPOT),

    tetramer staining, cytotoxicity assay for cellular responses, and

    enzyme-linked immunosorbent assay for the development of

    humoral immunity, are usually limited within these studies.

    Moreover, objective clinical responses are even rarer. Initially,

    the disappointing results obtained led the field to conclude that

    the immunogenicity of vaccines need to be enhanced by

    targeting new TAAs, adopting completely new vaccination

    strategies or by attempting to bolster the effectiveness of

    existing ones, for example by the co-administration of

    Immunological Reviews 222/2008 317

    Gray et al Therapeutic vaccines in cancer prevention

  • Table1.

    Summaryofrecentclinical

    trialsoftherapeu

    ticprostatecancervaccines

    Aut

    hor(

    s)Y

    ear

    Vac

    cine

    Num

    ber

    of

    pat

    ient

    sD

    isea

    sest

    atus

    Pre

    vious

    trea

    tm

    ent(

    s)Pat

    ient

    resp

    ons

    eto

    vacc

    ine

    Clin

    ical

    resp

    ons

    esR

    efer

    ence

    Tjo

    aet

    al.

    1998

    Aut

    olo

    gous

    DC

    spul

    sed

    with

    PSM

    Apep

    tides

    .33

    Met

    asta

    ticpro

    stat

    eca

    ncer

    RP,H

    ,pal

    liativ

    era

    dio

    ther

    apy

    DT

    Hfo

    rPSM

    A9/3

    3PR

    ,11/3

    3SD

    ,13/3

    3N

    R(1

    )

    Mur

    phy

    etal

    .1999

    As

    Tjo

    aet

    al.(

    1)

    37

    Adva

    nced

    pro

    stat

    eca

    ncer

    (sta

    geC

    D

    )R

    P,X

    RT

    ,B,H

    DT

    Hfo

    rPSM

    A1/3

    7C

    R,1

    0/3

    7PR

    ,8/3

    7SD

    ,18/3

    7N

    R(2

    )

    Eder

    etal

    .2000

    Vac

    cini

    a-bas

    edva

    ccin

    ein

    corp

    ora

    ting

    PSA

    33

    Adva

    nced

    (up

    toan

    din

    clud

    ing

    LNin

    volv

    e-m

    ent)

    but

    nobony

    met

    sal

    low

    ed

    XR

    T,R

    P,b

    oth

    14/2

    5as

    sess

    able

    pat

    ient

    sha

    dD

    TH

    resp

    ons

    esto

    vax

    afte

    ral

    l3

    dose

    s.In

    the

    vax1

    GM

    -CSF

    group

    ,7/1

    0pat

    ient

    sha

    dD

    TH

    resp

    ons

    esan

    d5/7

    had

    cellu

    lar

    resp

    ons

    es(E

    LISP

    OT

    )

    SDin

    6/3

    3pat

    ient

    s(3

    )

    Smal

    letal

    .2000

    As

    Smal

    letal

    .(12)

    31

    Met

    asta

    ticH

    PR

    CR

    P,X

    RT

    ,B,C

    RY

    O10/2

    6pat

    ient

    ste

    sted

    dev

    eloped

    T-c

    ellr

    espons

    esto

    PA

    P,1

    6/3

    1te

    sted

    dev

    eloped

    anti-

    PA

    Pan

    tibodie

    s

    Thr

    eepat

    ient

    ssh

    ow

    ed4

    50%

    dec

    reas

    ein

    PSA

    leve

    l,th

    ree

    more

    show

    eda

    dec

    reas

    eof

    2549

    %.M

    edia

    nT

    TP

    for

    pa-

    tient

    sw

    hodev

    eloped

    anim

    -m

    une

    resp

    ons

    ew

    as34

    wee

    ksco

    mpar

    edw

    ith13

    wee

    ksfo

    rth

    ose

    that

    did

    not(Po

    0.0

    27)

    (4)

    Fong

    etal

    .2001

    Aut

    olo

    gous

    DC

    slo

    aded

    with

    xeno

    geni

    cm

    urin

    ePA

    P

    21

    Met

    asta

    ticpro

    stat

    eca

    ncer

    ,eith

    erho

    rmone

    sens

    itive

    or

    HPR

    C

    C,X

    RT

    ,RP

    T-c

    ellp

    rolif

    erat

    ion

    assa

    yaf

    ter

    stim

    ulat

    ion

    with

    hPA

    P/m

    PA

    P,

    ELIS

    Afo

    rIL

    -4,I

    L-10,I

    FN-g

    and

    TN

    F-a,

    ELIS

    PO

    Tfo

    rIF

    N-g

    and

    IL-4

    .All

    pat

    ient

    sdev

    eloped

    re-

    spons

    eto

    mPA

    P.1

    1/2

    1pat

    ient

    sdev

    eloped

    ace

    llula

    rre

    spons

    eto

    hPA

    P

    8/2

    1ha

    ddisea

    sest

    abiliza

    tion.

    Pro

    gres

    sion-

    free

    surv

    ival

    very

    sign

    ifica

    ntly

    bet

    ter

    inth

    egr

    oup

    that

    dev

    eloped

    hPA

    Pre

    spons

    esco

    mpar

    edw

    ithth

    ose

    that

    did

    not(P

    =0.0

    087

    )

    (5)

    Hei

    seret

    al.

    2002

    Aut

    olo

    gous

    DC

    str

    ans-

    fect

    edw

    ithPSA

    RN

    A13

    Met

    asta

    tic(D

    1to

    D3)

    m

    ost

    D3

    XR

    T,R

    P,n

    one

    ,OA

    llpat

    ient

    ssh

    ow

    edPSA

    -spec

    ific

    ELIS

    PO

    Tre

    spons

    eD

    ecre

    ase

    inse

    rum

    PSA

    leve

    lsfo

    r1/7

    eval

    uable

    pat

    ient

    s.D

    ecre

    ase

    inlo

    gPSA

    leve

    lfor

    afu

    rthe

    r5/7

    .6pat

    ient

    sex

    clud

    ed

    [6]

    Bur

    chet

    al.

    2004

    As

    Smal

    letal

    .(1

    2)

    21

    Met

    asta

    ticH

    PR

    CR

    P,A

    A,X

    RT

    ,HPro

    lifer

    atio

    nas

    say

    and

    ELIS

    Afo

    rre

    spons

    esto

    PA

    P,G

    M-C

    SF,a

    ndPA

    2024.

    Sign

    ifica

    ntce

    llula

    rre

    -sp

    ons

    eat

    wee

    k4

    (Po

    0.0

    001),

    13/1

    5pat

    ient

    sdev

    eloped

    anti-

    body

    resp

    ons

    esto

    PA

    202

    4but

    notPA

    P

    2pat

    ient

    ssh

    ow

    ed2550%

    de-

    crea

    sein

    PSA

    (tra

    nsie

    nt).

    One

    pat

    ient

    show

    eddec

    line

    ofP

    SAto

    undet

    ecta

    ble

    leve

    lsby

    wee

    k24

    co

    ntin

    ued

    to52

    mont

    hs

    (7)

    Kau

    fman

    etal

    .2004

    Vac

    cini

    a-an

    dfo

    wlp

    ox-

    bas

    edva

    ccin

    esin

    corp

    ora

    ting

    PSA

    64

    Org

    an-c

    ont

    aine

    dpro

    stat

    eca

    ncer

    C,H

    ,RP,R

    T,o

    ther

    46%

    had

    incr

    ease

    inPSA

    -spec

    ific

    T-c

    ellr

    espons

    esN

    oobje

    ctiv

    ere

    spons

    es,b

    utaf

    ter

    2ye

    ars

    off

    ollo

    w-u

    p,7

    8.1

    %ofm

    enar

    efr

    eefr

    om

    clin

    ical

    disea

    sepro

    gres

    sion

    and

    45.3

    %ar

    efr

    eefr

    om

    PSA

    pro

    gres

    sion

    (8)

    Suan

    dD

    annu

    llet

    al.

    2005

    DC

    str

    ansf

    ecte

    dw

    ithhT

    ERT

    or

    hTER

    T-L

    AM

    Pm

    RN

    A

    20

    Met

    asta

    ticH

    PR

    CR

    P,X

    RT

    ,C,H

    Incr

    ease

    sin

    IFN

    -gby

    ELIS

    PO

    Ton

    CD

    41

    and

    CD

    81

    cells

    No

    obje

    ctiv

    ere

    spons

    es,b

    utPSA

    doub

    ling

    time

    impro

    ved

    to100

    mont

    hsin

    6-c

    ycle

    group

    (P=

    0.0

    4).

    No

    impro

    vem

    ent

    seen

    in3-c

    ycle

    group

    (P=

    0.6

    3).

    (9)

    318 Immunological Reviews 222/2008

    Gray et al Therapeutic vaccines in cancer prevention

  • Tra

    nsie

    ntcl

    eara

    nce

    ofP

    SA-

    expre

    ssin

    gtu

    mor

    cells

    obse

    rved

    inso

    me

    subje

    cts

    (by

    real

    -tim

    ePC

    Rfo

    rPSA

    mR

    NA

    )D

    iPao

    laet

    al.

    2006

    Vac

    cini

    a-an

    dfo

    wlp

    ox-

    bas

    edva

    ccin

    esin

    corp

    or-

    atin

    gPSA

    seque

    nce

    and

    TR

    ICO

    M(B

    7-1,I

    CA

    M-1

    ,an

    dLF

    A-3

    ).Pat

    ient

    str

    eate

    dw

    ithva

    ccin

    iavi

    rus

    then

    boost

    edw

    ithfo

    wlp

    ox

    10

    HPR

    C,s

    om

    em

    etas

    tatic

    No

    prior

    chem

    oth

    erap

    yal

    low

    ed

    Positiv

    ere

    spons

    esfo

    ran

    ti-va

    ccin

    ia,-

    fow

    lpox

    and

    -PSA

    antib

    odie

    sby

    ELIS

    A

    4/1

    0SD

    ,6/1

    0N

    R(1

    0)

    Fues

    sele

    tal

    .2006

    DC

    slo

    aded

    with

    aco

    ck-

    tail

    ofp

    eptid

    esder

    ived

    from

    PSA

    ,PSM

    A,s

    urvi

    -vi

    n,pro

    stei

    n,an

    dtr

    p-p

    8

    8H

    PR

    Cpat

    ient

    s,al

    lbut

    one

    met

    asta

    ticX

    RT

    ,RP,C

    ,none

    4/8

    positiv

    efo

    rre

    spons

    eby

    ELIS

    PO

    Tonl

    y3/4

    ELIS

    PO

    Tre

    spond

    ers

    also

    show

    edcl

    inic

    alre

    spons

    es(1

    1)

    Smal

    letal

    .2006

    Aut

    olo

    gous

    DC

    slo

    aded

    with

    reco

    mbin

    antfu

    sion

    pro

    tein

    ofP

    AP

    com

    -bin

    edw

    ithG

    M-C

    SF(P

    rove

    nge)

    127

    Met

    asta

    ticH

    PR

    CN

    otdiscu

    ssed

    indet

    ail

    Ver

    ysign

    ifica

    nt(Po

    0.0

    01)

    incr

    ease

    inT

    -cel

    lres

    pons

    eto

    antig

    enic

    targ

    et

    Sign

    ifica

    ntim

    pro

    vem

    entin

    ove

    rall

    surv

    ival

    (P=

    0.0

    1)

    (12)

    Tho

    mas

    -Kas

    kele

    tal

    .2006

    DC

    slo

    aded

    with

    PSC

    Aan

    dPSA

    pep

    tides

    .12

    Met

    asta

    ticpro

    stat

    eca

    ncer

    RP,X

    RT

    ,H,C

    ,AA

    5/1

    2ha

    dpositiv

    eD

    TH

    resp

    ons

    e.1/1

    2ha

    dpositiv

    ete

    tram

    erre

    spons

    e

    6/1

    2SD

    ,6/1

    2PD

    incl

    udin

    gone

    that

    had

    aLN

    regr

    ession

    (13)

    Wae

    cker

    le-

    Men

    etal

    .2006

    Aut

    olo

    gous

    DC

    spul

    sed

    with

    pep

    tides

    der

    ived

    from

    PSC

    A,P

    AP,P

    SMA

    ,an

    dPSA

    6H

    PR

    Cpat

    ient

    s,al

    lbut

    one

    met

    asta

    ticX

    RT

    3/6

    positiv

    efo

    rre

    spons

    eby

    ELIS

    PO

    T,t

    etra

    mer

    anal

    ysis

    and

    invivo

    CT

    Las

    say

    All

    thre

    epat

    ient

    sw

    ithim

    mun

    ere

    spons

    esha

    ddec

    reas

    edPSA

    doub

    ling

    times

    (14)

    Nogu

    chie

    tal

    .2007

    Per

    sona

    lized

    pre

    oper

    a-tiv

    epep

    tide

    vacc

    ine,

    follo

    wed

    by

    RP

    18

    Clin

    ical

    lylo

    caliz

    edN

    one

    In8/1

    0pat

    ient

    ste

    sted

    ,cel

    lula

    rre

    spons

    esto

    atle

    astone

    oft

    hefo

    urpep

    tides

    wer

    eobse

    rved

    .Si

    gnifi

    cant

    lym

    ore

    TIL

    sin

    the

    vacc

    inat

    edgr

    oup

    (P=

    0.0

    043)

    but

    nosign

    ifica

    ntdiff

    eren

    cebet

    wee

    nB-c

    ella

    ndC

    D81

    infil

    trat

    ion.

    IgG

    spec

    ific

    toT

    -cel

    lep

    itopes

    det

    ecte

    din

    8/1

    0pat

    ient

    s

    No

    sign

    ifica

    ntdiff

    eren

    cebe-

    twee

    nva

    ccin

    ated

    and

    cont

    rols

    inte

    rms

    ofP

    SAle

    vels,b

    utfo

    uroft

    hec

    ont

    aine

    ddisea

    sev

    acci

    -na

    ted

    pat

    ient

    sac

    tual

    lyha

    dm

    ore

    adva

    nced

    disea

    se.B

    ette

    rst

    udy

    des

    ign

    need

    ed

    (15)

    Tre

    atm

    ents

    :A

    A,an

    dro

    gen

    abla

    tion;

    B,bra

    chyt

    hera

    py;

    C,ch

    emoth

    erap

    y;C

    RY

    O,cr

    yoth

    erap

    y;H

    ,ho

    rmone

    ther

    apy;

    O,orc

    hiec

    tom

    y;R

    P,ra

    dic

    alpro

    stat

    ecto

    my;

    XR

    T,ra

    dio

    ther

    apy.

    Res

    pons

    es:C

    R,

    com

    ple

    tere

    spons

    e;N

    R,n

    ore

    spons

    e;PR

    ,par

    tialr

    espons

    e;SD

    ,sta

    ble

    disea

    se;T

    TP,t

    ime

    topro

    gres

    sion.

    Immunological Reviews 222/2008 319

    Gray et al Therapeutic vaccines in cancer prevention

  • immunostimulatory cytokines or costimulatory molecules.

    Despite these efforts, the results of prostate cancer

    immunotherapy trials have been broadly similar to each other

    regardless of the vaccination strategy used or TAA targeted.

    Commonly, overall survival (OS) and/or time to progression is

    significantly improved in patients that develop antigen-specific

    cellular or humoral immune responses as a result of vaccination,

    implying that the vaccines that have been developed are perfectly

    capable of working in a subset of immunocompetent subjects.

    Similar results have been observed in trials of therapeutic

    vaccines for renal cell carcinoma (16, 17), melanoma (18, 19),

    pancreatic adenocarcinoma (20), and breast cancer (21). It is

    thus most likely that the failure to date of immunotherapeutic

    agents to mediate tumor clearance is mostly due to the immuno-

    compromised status of advanced cancer patients in which they

    are tested. Several studies have attempted to assess the general

    immune competence of test subjects, for example by measuring

    their ability to mount recall responses to common antigens.

    However, the ability of the immune system to respond to an

    antigen to which it has already been exposed is not necessarily a

    measure of its ability to mount a completely new immune

    response upon vaccination. In recent years, the cellular

    and molecular mechanisms underlying immune suppression

    and their implications for therapeutic cancer vaccination have

    been the subject of intense interest. These studies have revealed

    multiple mechanisms of immune suppression in advanced

    cancer patients that render them poor candidates for

    immunotherapeutic intervention.

    Large tumors fundamentally alter the immune systems ofpatients and limit their ability to mount an anti-tumorimmune response

    Large tumors have been shown to have multiple, often redundant

    pathways of immune escape. Tumor immune escape mechan-

    isms have been the subject of excellent reviews (22, 23) and are

    not covered in depth here. In summary, a tolerizing milieu

    consisting of regulatory T cells (Tregs), suppressive/tolerogenic

    DCs, and suppressive cytokines develops within the tumor

    microenvironment. Antigen presentation and T-cell activation

    are often compromised within tumors. In addition, corrupted

    CD81 T-cell memory function results from chronic stimulation

    and contributes to immune failure in cancer patients (24).

    As mechanisms of tumor-mediated immune suppression

    have been elucidated over recent years, a determined research

    drive has been concentrated on eliminating or inhibiting them

    to enhance the efficacy of therapeutic cancer vaccines.

    Naturally occurring CD41CD251 Tregs have been the focus of

    particularly intense study in this regard, although they do not

    always seem to be involved in mediating the suppression

    of immune responses elicited by cancer vaccines (25).

    Nevertheless, natural Tregs have been accepted as central players

    in tumor-mediated immune suppression in many cancers, and

    several methods to deplete them or limit their suppressive

    activity have been developed (26). For example, inhibition of

    CD41CD251 Tregs by cyclophosphamide pretreatment before

    immunization with a vaccine directed against HER-2/neu in

    mice allowed the activation of latent antigen-specific CD81 T

    cells that were capable of mounting an anti-tumor response

    (27). Efforts to abrogate natural Treg activity to boost vaccine

    efficacy have also included depletion via treatment with a

    monoclonal anti-CD25 antibody (28) and administration of

    denileukin diftitox (Ontak, Ligand Pharmaceuticals, San Diego,

    CA, USA) (29, 30). Other approaches to increasing tumor

    vaccine efficacy by limiting tumor-mediated immuno-

    suppression have included attempts to block suppressive

    pathways in tolerogenic DCs and efforts to limit the effects

    of suppressive cytokines, for example by administration of

    neutralizing antibodies. Our recent studies have shown the

    emerging importance of hardwired negative regulators of

    proinflammatory signaling in antigen-presenting cells in the

    maintenance of self-tolerance. Suppressor of cytokine signaling

    1 (SOCS1) is a key regulator of cytokine receptor-mediated Janus

    kinase (JAK)/signal transducer and activator of transcription

    (STAT) signaling. We found that SOCS1 has a central role in

    regulating the duration and intensity of antigen presentation by

    DCs. Inhibition or silencing of SOCS1 in DCs results in the over-

    activation of antigen-presenting DCs that in turn leads to

    enhanced antigen-specific anti-tumor immunity, providing a

    new strategy to break self-tolerance and enhance the potency of

    tumor vaccines (31, 32). However, despite these advances in

    targeting and inhibiting individual mechanisms of tumor-

    mediated immune suppression, abrogating their collective

    effects may prove to be difficult (33).

    Immunosuppressive tumor microenvironments inhibit the

    local anti-tumor immune response, both natural and in response

    to vaccination, but they can also produce suppressive cells of

    multiple phenotypes that migrate from the tumor to lymphoid

    organs where they can mediate systemic immunosuppression.

    These cells can inhibit immune responses even after the original

    suppressive network has been eliminated, for example by tumor

    resection, as discussed below. Moreover, metastasis of tumor cells

    to a sentinel lymph node (LN), the first LN away from the

    primary tumor in the lymphatic drainage pathway, leads to local

    immunosuppression within that LN (34). This is significant as the

    sentinel node is the first lymphoid organ in which antigenic

    stimulation occurs as the first stage in the development of a

    320 Immunological Reviews 222/2008

    Gray et al Therapeutic vaccines in cancer prevention

  • systemic immune response. Although the local immuno-

    suppression mediated by metastatic tumor cells within LNs does

    not normally lead to complete systemic immunosuppression, this

    process nevertheless represents a mechanism by which the

    presence of tumors can compromise the immune response of

    the patient.

    Systemic immune dysfunction was recently demonstrated in

    the patients enrolled in a failed Phase II clinical trial of a

    melanoma vaccine based on the glycoprotein MPS160 (35).

    Although many patients demonstrated an increase in vaccine-

    specific cytotoxic T-lymphocyte (CTL) numbers, as measured by

    tetramer analysis, these cells were broadly incapable of

    producing interferon-g (IFN-g) when stimulated with therelevant MPS160 peptides in vitro. Repeated immunizations

    simply caused an increase in the numbers of these antigen-

    specific cells that fail to produce IFN-g. Furthermore, expansionsin this non-responsive CTL population were correlated with

    tumor progression events. Analysis of the peripheral blood of

    vaccinated patients and healthy control subjects indicated that

    the profile of plasma cytokines was skewed toward

    immunosuppressive molecules. The authors hypothesized that

    these suppressive cytokines were being produced within the

    tumor microenvironment and were spilling over into the

    plasma, resulting in a failure of systemic immune competence.

    Alterations in the plasma cytokine profiles of cancer patients

    compared with healthy subjects and those with benign tumors

    have been highlighted in several studies. We recently completed

    a study profiling the serum levels of 14 cytokines in 187 ovarian

    cancer patients and compared them with those of 45 patients

    with benign ovarian tumors and 50 healthy control subjects

    (36). New cytokine bead array technology allowed the

    simultaneous analysis of multiple cytokines present in serum

    samples. Univariate analyses demonstrated that serum IL-6, IL-

    7, and macrophage chemotactic protein-1 (MCP-1) were

    increased in ovarian cancer patients compared with healthy

    controls and patients with benign ovarian tumors (Po 0.05 inall cases). Many other studies have highlighted differences in

    serum levels of cytokines in prostate cancer patients compared

    with controls (37). Altered serum levels of IL-4, IL-6, IL-10,

    and transforming growth factor-b (TGF-b) have been noted inprostate cancer patients, with several studies demonstrating that

    increased IL-6 levels are a negative predictor of disease outcome.

    Current conventional anticancer therapies havesignificant effects on the immune system

    Current conventional cancer therapies are well known to have

    potent immunomodulatory effects that can either inhibit or

    enhance the anti-tumor immune response elicited by vaccina-

    tion. Radiation therapy and many chemotherapeutic agents are

    known to be immunosuppressive, particularly at high dosages.

    Lymphocytes responding to antigens, including those stimu-

    lated by vaccination against TAAs, rapidly proliferate. Therefore

    they are susceptible to anticancer therapies that preferentially

    kill proliferating cells (38).

    Despite the traditionally held view that radiotherapy and

    chemotherapy are generally immunosuppressive, the effects

    of conventional anticancer treatments on the immune system

    are extremely complex. As a result, chemotherapy and

    radiotherapy can be combined with vaccination strategies to

    improve their effectiveness so long as due attention is paid

    to dosages and the relative timing of each treatment. For

    example, doxorubicin and melphalan were shown not to

    impede the efficacy of two separate vaccination strategies in

    micewhen they were administered shortly before immunization

    (39). An excellent recent review has covered scenarios in

    which cytotoxic chemotherapies have been combined

    with immunotherapy to yield enhanced responses (40).

    Radiotherapy has also been combined with immunotherapy.

    For example, a course of radiotherapy was recently integrated

    into a vaccination schedule in prostate cancer patients (41).

    Thirty patients were either given radiotherapy alone or

    radiotherapy plus vaccination with a recombinant vaccinia-

    prostate-specific antigen (PSA) vaccine followed by monthly

    boosting with recombinant fowlpox-PSA. In the combination

    therapy arm, patients received radiotherapy between the third

    and fifth boosts. Thirteen out of 17 patients in the combination

    arm developed PSA-specific cellular responses, versus none in

    the radiotherapy-only arm (Po 0.0005). Given that there wasno immunotherapy-only treatment arm in this study, it cannot

    be concluded that radiotherapy enhanced the immune response

    to vaccination. However, the study ably demonstrates that local

    radiotherapy does not inhibit antigen-specific responses elicited

    by vaccination. Finally, we demonstrated that a peptide vaccine

    directed against human papilloma virus (HPV) was capable of

    eliciting complete protection against challenge with HPV-16-

    expressing tumors in mice, despite prior treatment with pelvic

    radiation or cisplatin. This was true even though the radiation-

    and cisplatin-pretreated mice had measurably lower peptide-

    specific immune responses to vaccination than untreated

    controls, as measured by IFN-g ELISPOT (42).Despite the success of combining conventional anticancer

    treatments with immunotherapy in certain instances, this

    approach may not always be feasible. In many cases, the

    optimal chemotherapeutic agents and/or radiotherapeutic

    schedules available to physicians are dictated by the nature of

    Immunological Reviews 222/2008 321

    Gray et al Therapeutic vaccines in cancer prevention

  • the cancer they are treating. Not all of these treatment options

    will be suitable for combination with immunotherapeutic

    strategies, and therefore not all cancers will be viable targets

    for developing combination treatment strategies. Furthermore,

    cancer patients are most commonly enrolled in clinical trials

    for therapeutic vaccines as a treatment of last resort. Most

    clinical immunotherapy trials exclude patients that have

    received prior treatments that may affect vaccine efficacy, but

    this is usually limited to treatment received o 1 month beforethe start of the trial. As a result, patients enrolled in cancer

    immunotherapy clinical trials have already received a variety of

    prior treatments that may have had long-lasting effects on their

    immune competence. The complexity of the interactions

    between these various treatments and the immune system

    makes accurate assessment of the results of clinical trials and

    indeed comparisons between otherwise similar trials

    exceedingly difficult.

    Therapeutic cancer vaccines have limited efficacy inpatients with minimal residual disease

    It is well accepted that tumor growth is frequently too rapid for

    the immune system to contain, despite the induction of a

    robust anti-tumor response (40). Given that immunotherapy

    in many cases can be successfully combined with certain

    conventional therapies, the cancer immunotherapy field has

    actively investigated a paradigm in which vaccines are admi-

    nistered in the adjuvant setting, where minimal residual disease

    is present. Clinical trials in which patients have been vaccinated

    after their tumors have been debulked by surgical, chemother-

    apeutic, or radiological means have been conducted for several

    cancers, most notably in melanoma (4355), and lymphoma

    (56, 57). Other examples of therapeutic cancer vaccines being

    used in the adjuvant setting include those directed against

    ovarian cancer (58, 59), lung cancer (60, 61), pancreatic

    carcinoma (20, 62), and breast cancer (63).

    Most clinical trials of therapeutic cancer vaccines in the

    adjuvant setting have had positive but limited results. While

    disease-free survival (DFS) and/or OS are often improved

    (sometimes very significantly so), therapeutic vaccination in

    the adjuvant setting is rarely capable of eradicating the residual

    tumor cells that are present after surgery and eliciting a complete

    response. This inability is probably because debulking tumors

    removes only one component of the systemic immune

    suppression that they establish, namely the intratumoral

    suppressive milieu. One mouse study has demonstrated that

    immunocompetence of the host can be restored upon surgical

    removal of the primary tumor, despite the continued presence of

    metastases in a mouse model of mammary carcinoma (64).

    However, the limited results of human clinical trials suggest that

    this effect is abrogated in humans, indicating that the

    suppressive T cells/DCs that have already escaped the tumor

    may be active in the LN and the immunosuppressive cytokine

    profile present in the peripheral blood of the patient are capable

    of limiting vaccine efficacy even in the absence of the

    immunosuppressive tumor microenvironment. Nevertheless,

    the improvements in DFS and OS observed in many studies of

    therapeutic vaccination in the adjuvant setting indicate that

    tumor-mediated immunosuppression is a major factor in the

    limited efficacy of therapeutic cancer vaccines and that by

    partially disrupting that immunosuppression their effectiveness

    can be increased. These findings have led us and others (65) to

    hypothesize that immunization with a therapeutic cancer

    vaccine at the earliest stages of carcinogenesis before local and

    systemic immunosuppressive environments are established by

    the tumor will yield the best immune responses to vaccination

    and therefore confer excellent protection against the progression

    of cancer development.

    Vaccination at early stages of carcinogenesis is highlyeffective at inducing anti-tumor immune responses andimproving survival

    Patients with advanced cancer display systemic and local

    (intratumoral) immunosuppression that is only partially alle-

    viated by debulking of the tumor. Furthermore, overcoming

    the combined tumor immunosuppressive mechanisms once

    they become established is proving to be difficult. With these

    considerations in mind, our group and others have directed

    their attention to using therapeutic cancer vaccines in the

    preventive setting.

    We have recently demonstrated that a therapeutic vaccination

    strategy directed against two different prostate TAAs (66, 67,

    and authors unpublished observations) at the earliest stage of

    carcinogenesis can elicit superb long-term protection against

    spontaneous prostate cancer development in transgenic

    adenocarcinoma mouse prostate (TRAMP) mice. In both

    studies, we used a heterologous vaccination scheme involving

    priming mice with a plasmid containing cDNA encoding the

    target antigen, followed by boosting with Venezuelan equine

    encephalitis (VEE) virus replicon particles (VRP) expressing the

    same antigen. VEE VRP have been shown previously to efficiently

    induce anti-tumor immune responses (68), and heterologous

    vaccination schemes have been shown to increase

    immunogenicity (69). The TAAs targeted in these studies, six-

    transmembrane epithelial antigen of the prostate (STEAP) and

    322 Immunological Reviews 222/2008

    Gray et al Therapeutic vaccines in cancer prevention

  • prostate stem cell antigen (PSCA), are strongly overexpressed in

    both human and murine prostate cancers (7072).

    TRAMP mice vaccinated against PSCA at an early stage of

    carcinogenesis, at 8 weeks of age when they have developed only

    premalignant prostate intraepithelial neoplasia (PIN) lesions,

    showed dramatically improved survival compared with

    unvaccinated age-matched controls (67). The OS of the PSCA

    vaccinated group was 90% at 360 days, while all of the

    unvaccinated controls had reached the survival endpoint by

    day 360 post-vaccination. In contrast, PSCA vaccination at

    16 weeks of age, when TRAMP mice have developed prostate

    cancer, conferred no significant protection of mice compared

    with unvaccinated age-matched controls (unpublished

    observations). Interestingly, TRAMP mice vaccinated against

    PSCA early (i.e. at 8 weeks of age) did develop prostate tumors,

    but the majority of the mice presented with small well-

    differentiated focal adenocarcinomas with extensive hyperplasia

    and multiple apoptotic zones. Greater numbers of immune

    cells, including macrophages, DCs, and IFN-g-expressingCD41 and CD81 T cells, infiltrated the prostate tumors of

    PSCA vaccinated TRAMP mice compared with unvaccinated

    controls. Intratumoral IL-5, IL-4, and IFN-g cytokine levelswere significantly increased in response to early PSCA

    vaccination. In addition, transcription of IL-2 was significantly

    upregulated within the tumors of mice vaccinated early against

    PSCA compared with unvaccinated controls. Overall, TRAMP

    mice vaccinated early against PSCA appeared outwardly healthy

    at 18 months of age and had small, low-grade prostate tumors

    that were in stark contrast to the very large, high-grade prostate

    tumors observed in control vaccinated mice that typically

    resulted in death before 9 months of age.

    In a parallel study, we investigated the effectiveness of

    vaccination against STEAP using our immunization protocol in

    protecting TRAMP mice from prostate cancer (66). Initial

    studies were carried out to determine whether STEAP

    vaccination could protect C57BL/6 mice against challenge

    with TRAMP-C2 prostate cancer cells. Interestingly, CD41 T

    cells expressing IFN-g, TNF-a, and IL-2 played a major role intumor rejection. Furthermore, the presence of high IL-12 levels

    in the tumor environment was associated with tumor rejection.

    The therapeutic efficacy of STEAP vaccination was more modest

    than its prophylactic effectiveness, but nevertheless it induced a

    small but significant delay in the progression of established

    TRAMP-C2 tumors. To assess the efficacy of STEAP vaccination

    in a more physiologically relevant setting, TRAMP mice were

    vaccinated against STEAP. In a striking parallel with our PSCA

    study, TRAMP mice vaccinated at 8 weeks of age displayed

    vastly superior survival compared with age-matched controls,

    while mice vaccinated at 16 weeks of age showed little

    improvement in survival (authors unpublished observations).

    The extraordinary survival differences between mice

    vaccinated against PSCA or STEAP at 8 versus 16 weeks of age

    are most likely attributable solely to the stage of carcinogenesis

    at which immunization occurs. Identical vaccination protocols

    and reagents were used in each group of mice, and there is still

    significant thymic output at 16 weeks of age in mice (73). We

    hypothesize that by 16 weeks of age a suppressive prostate

    tumor microenvironment has become established that inhibits

    the anti-tumor immune response induced by vaccination. At 8

    weeks of age, there is most likely a microenvironment within

    PIN lesions that is still favorable for the establishment of an

    anti-tumor immune response. This idea is demonstrated by the

    vaccine-induced infiltration by active immune cells into the

    prostate tumor that subsequently develops from these PIN

    lesions and by the immuno-activating cytokine profiles that

    develop within those tumors. We are currently involved in

    studies designed to determine precisely at which stage of

    carcinogenesis the prostate tumor microenvironment switches

    from immune favorable to immunosuppressive. These studies

    will allow us to intervene with cancer immunotherapy at the

    latest possible stage of tumorigenesis at which vaccination can be

    applied and still elicit an effective anti-tumor immune response.

    Prevention of the spontaneous development of tumors by

    vaccination at an early stage of carcinogenesis has also been

    demonstrated in BALB-neuT mice that spontaneously develop

    mammary tumors. Nava-Parada et al. (74) demonstrated that a

    single vaccination of BALB-neuT mice with a peptide vaccine

    derived from the RNEU TAA with concomitant administration

    of the Toll-like receptor agonist CpG can significantly delay

    spontaneous tumor development (74). BALB-neuT mice

    remained completely tumor free until approximately 23

    weeks of age, when they were vaccinated at 8 weeks of age, at

    which time they display diffuse atypical hyperplasia but not

    overt carcinoma. Interestingly, the authors report that peptide

    vaccination at later stages of carcinogenesis was less effective

    in eliciting anti-tumor immune responses and controlling

    tumor growth.

    Several studies have demonstrated that vaccination of

    women with premalignant cervical intraepithelial neoplastic

    lesions (CIN) can cause their complete eradication or partial

    regression to a lower-grade lesion. Muderspach et al. (75)

    demonstrated three complete responses and six partial

    responses in 12 patients with grade II/III CIN that were

    vaccinated with a vaccine directed against HPV E7 peptides.

    Immunization with a recombinant protein (SGN-00101)

    consisting of bacterial heat shock protein (M. bovis Hsp65)

    Immunological Reviews 222/2008 323

    Gray et al Therapeutic vaccines in cancer prevention

  • fused to the complete HPV-16 E7 sequence was recently shown

    by Roman et al. (76) to confer clinical benefit to patients with

    high-grade CIN lesions. Of the 20 women enrolled in the study,

    seven showed complete regression of their CIN lesions, one

    had a partial regression to a low-grade lesion, 11 had stable

    disease, and one patient progressed (76). A phase III

    randomized study of the same vaccine conducted by Einstein

    et al. (77) yielded very similar results. Of the 58 patients that

    had completed the full vaccination protocol, 13 had a complete

    response, 32 had partial responses, 11 had stable disease, and

    two progressed (77). A vaccine based on a vaccinia virus vector

    expressing recombinant MVA E2 has also been demonstrated to

    be effective in treating high-grade CIN lesions (78). Of the 34

    women immunized in a recent phase II clinical trial of the

    vaccine, 19 showed complete regression, and 15 showed partial

    regression (79). Early trials of other therapeutic cervical cancer

    vaccines have demonstrated the generation of excellent immune

    responses in patients with CIN lesions, although no clinical

    responses were assessed (80, 81). Generally, vaccination of

    CIN patients elicits the development of very robust immuno-

    logical responses. This indicates that these patients are generally

    immunocompetent, as is expected at the early stages of carcino-

    genesis (82). Given the availability of efficacious therapeutic

    vaccines and the accessibility and reliability of current screening

    methods, cervical cancer represents an excellent candidate for a

    treatment modality in which premalignant lesions are treated by

    therapeutic vaccination.

    Although the circumvention of immunosuppression

    mediated by tumors and their treatment is the primary

    justification for vaccination against premalignant lesions, in

    some cases administration of therapeutic cancer vaccines has

    had unexpected beneficial effects on the outcomes of

    subsequent conventional treatments. As has been recently

    discussed (83), superior responses to standard therapies have

    been observed in patients who have initially received a

    therapeutic vaccine and have subsequently been given

    conventional treatments upon disease progression. This

    fascinating phenomenon has been observed in trials of

    vaccines directed against small-cell lung cancer (84) and

    prostate cancer (85, 86). These observations are particularly

    significant in the case of prostate cancer. The trials in question

    were conducted in patients with HRPC who were probably

    immunocompromised, given that prostate cancer patients with

    metastatic disease are less able to mount immune responses

    than patients that have less advanced disease (87). Early

    detection of prostate cancer is commonplace as a result of PSA

    screening. The potential side effects of conventional prostate

    cancer therapies are more severe than the consequences of

    living with contained, low-grade prostate cancer. As a result,

    men with rising PSA levels frequently undergo very long

    periods of active surveillance and only elect to undergo

    curative surgery when it becomes apparent that the disease

    has begun to progress. It is conceivable that therapeutic

    vaccination during this watchful waiting phase may clear

    premalignant prostate intraepithelial lesions or delay their

    progression to prostate adenocarcinoma. If and when this

    occurs, prior vaccination may then help enhance the efficacy

    of standard prostate cancer therapies that the patient receives.

    In summary, early vaccination of premalignant prostate lesions

    is an attractive proposition, because it may yield clinical benefit

    to patients at multiple stages of disease progression.

    Several cancers will be excellent candidates forvaccination at the early stages of carcinogenesis becausenovel early detection methods are being developed andtherapeutic vaccines are available

    Advances in genomics and proteomics have led to the develop-

    ment of novel methods of cancer detection. Analysis of genetic

    and epigenetic markers associated with carcinogenesis can be

    performed on DNA obtained from whole tumor cells in the

    peripheral blood, sputum, tumor ascites, and urine, or on

    naked DNA shed into these fluids by dying cancer cells.

    Analysis of DNA markers is exquisitely sensitive thanks to

    polymerase chain reaction-based amplification of the minute

    amounts of tumor DNA that are normally obtained. The

    development of DNA microarray technology and the ability to

    analyze thousands of DNA molecules simultaneously that it

    affords will also revolutionize cancer diagnostics. In addition,

    new proteomics methodologies, including very high-through-

    put mass spectrometry and protein chip analysis, allow scien-

    tists to search for cancer-associated protein markers at an

    unprecedented resolution (88). Very intense efforts to use

    these technologies and others are currently underway to dis-

    cover novel biomarkers that will allow the very early detection

    of many cancers, including ovarian, lung, and breast cancer.

    There are several examples of promising therapeutic cancer

    vaccines that are apparently capable of eliciting substantial

    immune responses but are not able to eradicate tumors in

    clinical trials. The development of new diagnostic tools may

    allow researchers and physicians to reassess the efficacy of these

    vaccines in the preventive setting by administering them at the

    earliest possible stages of carcinogenesis. For example, a

    vaccine based on an anti-idiotypic antibody that mimics the

    ganglioside GD3, Bec2/bacille Calmette Guerin (BCG), was

    recently demonstrated to be ineffective in improving either OS

    or progression-free survival in a phase III study involving 515

    324 Immunological Reviews 222/2008

    Gray et al Therapeutic vaccines in cancer prevention

  • small-cell lung cancer patients (61). Despite the lack of success

    in improving patient survival, this vaccine elicited humoral

    responses in approximately one-third of patients. Furthermore,

    there was a trend toward improved survival in patients with

    humoral responses versus those without (P=0.085), although

    this finding was weakened when the data were stratified for a

    confounding factor, namely the administration of prophylactic

    cranial irradiation being more prevalent in the group that

    developed humoral responses. A requirement for enrollment

    in this study was that patients had only limited disease as

    defined by the Veterans Administration Lung Study Group

    criteria. However, patients with limited disease are defined

    by these standards as having primary tumor and nodal

    involvement limited to one hemithorax (89). Given that even

    this is a relatively advanced stage of disease, it remains possible

    that the Bec2/BCG vaccine may yield improvements in patient

    survival if it is administered at a much earlier stage of

    carcinogenesis. Another example of a currently available

    vaccine that may benefit from early administration resulting

    from improved screening methodologies includes a breast

    cancer vaccine consisting of a HER-2/neu peptide (E75)

    mixed with GM-CSF that has been successful in preventing

    disease recurrence in node-positive patients with minimal

    disease burden (90). Similarly, oregovomab (mAb B43.13)

    has been demonstrated to increase time to progression in

    ovarian cancer, but only in the subgroup of patients that had

    the smallest residual tumors (59). It is possible that one or both

    of these vaccines would be even more successful if

    administered earlier in the development of disease. Improved

    screening and identification of biomarkers for lung, breast, and

    ovarian cancers may soon allow the vaccines that are available

    for them to be tested in the preventive setting, which may lead

    to improved patient outcomes.

    Conclusion

    The results of therapeutic cancer immunotherapy trials con-

    tinue to be disappointing, despite major advances in vaccine

    technology over the past decade. It has become apparent that

    while the vaccines themselves have the potential to elicit potent

    anti-tumor immune responses, the cancer patients in which

    they are tested are simply not capable of mounting robust

    responses to the vaccines. The compromised immunity in

    patients with advanced cancer is largely unavoidable and,

    currently, untreatable. This is broadly true even in instances

    where conventional anticancer treatments have been used to

    generate a scenario of minimal residual disease in which to

    administer immunotherapy. Tumor- and treatment-mediated

    immune suppression in cancer patients can be avoided by

    vaccinating them early in disease, before other treatments are

    dispensed and before the establishment of systemic immune

    failure by the tumor. Rapid advances in cancer screening have

    been made, and improvements in early cancer detection will

    continue to advance as genomic and proteomic technologies

    become more integrated into cancer diagnostics. With todays

    technology, early detection methods and efficacious therapeutic

    vaccines exist for two extremely common human cancers,

    prostate and cervical, that allow the vaccination of patients at the

    very earliest stages of carcinogenesis. With these considerations

    in mind, we propose a paradigm shift away from the use of

    therapeutic cancer vaccines in patients with advanced disease and

    toward their application in the earliest stages of tumorigenesis.

    References

    1. Tjoa BA, et al. Evaluation of phase I/II clinical

    trials in prostate cancer with dendritic cells and

    psma peptides. Prostate 1998;36:3944.

    2. Murphy GP, et al. Phase II prostate cancer

    vaccine trial: report of a study involving 37

    patients with disease recurrence following

    primary treatment. Prostate 1999;39:5459.

    3. Eder JP, et al. A phase I trial of a recombinant

    vaccinia virus expressing prostate-specific

    antigen in advanced prostate cancer. Clin

    Cancer Res 2000;6:16321638.

    4. Small EJ, et al. Immunotherapy of hormone-

    refractory prostate cancer with antigen-

    loaded dendritic cells. J Clin Oncol

    2000;18:38943903.

    5. Fong L, et al. Dendritic cell-based xeno-

    antigen vaccination for prostate cancer

    immunotherapy. J Immunol 2001;167:

    71507156.

    6. Heiser A, et al. Autologous dendritic cells

    transfected with prostate-specific antigen

    RNA stimulate CTL responses against meta-

    static prostate tumors. J Clin Invest

    2002;109:409417.

    7. Burch PA, et al. Immunotherapy (APC8015,

    provenge) targeting prostatic acid

    phosphatase can induce durable remission of

    metastatic androgen-independent prostate

    cancer: a phase 2 trial. Prostate

    2004;60:197204.

    8. Kaufman HL, et al. Phase II randomized study

    of vaccine treatment of advanced prostate

    cancer (E7897): a trial of the eastern coop-

    erative oncology group. J Clin Oncol

    2004;22:21222132.

    9. Su Z, et al. Telomerase mRNA-transfected

    dendritic cells stimulate antigen-specific

    CD81 and CD41 T cell responses in patients

    with metastatic prostate cancer. J Immunol

    2005;174:37983807.

    10. Fuessel S, et al. Vaccination of hormone-

    refractory prostate cancer patients with

    peptide cocktail-loaded dendritic cells: results

    of a phase I clinical trial. Prostate 2006;

    66:811821.

    11. DiPaola RS, et al. A phase I trial of pox PSA

    vaccines (PROSTVAC-VF) with B7-1,

    ICAM-1, and LFA-3 co-stimulatory molecules

    (TRICOM) in patients with prostate cancer. J

    Transl Med 2006;4:1.

    12. Small EJ, et al. Placebo-controlled phase III

    trial of immunologic therapy with sipuleucel-t

    (APC8015) in patients with metastatic,

    asymptomatic hormone refractory prostate

    cancer. J Clin Oncol 2006;24:30893094.

    13. Thomas-Kaskel AK, et al. Vaccination of ad-

    vanced prostate cancer patients with PSCA

    Immunological Reviews 222/2008 325

    Gray et al Therapeutic vaccines in cancer prevention

  • and PSA peptide-loaded dendritic cells in-

    duces DTH responses that correlate with

    superior overall survival. Int J Cancer

    2006;119:24282434.

    14. Waeckerle-Men Y, et al. Dendritic cell-based

    multi-epitope immunotherapy of hormone-

    refractory prostate carcinoma. Cancer Immu-

    nol Immunother 2006;55:15241533.

    15. Noguchi M, et al. Immunological evaluation

    of neoadjuvant peptide vaccination before

    radical prostatectomy for patients with

    localized prostate cancer. Prostate 2007;

    67:933942.

    16. Uemura H, et al. A phase I trial of vaccination

    of CA9-derived peptides for hla-a24-positive

    patients with cytokine-refractory metastatic

    renal cell carcinoma. Clin Cancer Res

    2006;12:17681775.

    17. Berntsen A, Geertsen PF, Svane IM. Thera-

    peutic dendritic cell vaccination of patients

    with renal cell carcinoma. Eur Urol

    2006;50:3443.

    18. Berd D, Sato T, Cohn H, Maguire HC, Jr,

    Mastrangelo MJ. Treatment of metastatic

    melanomawith autologous, hapten-modified

    melanoma vaccine: regression of pulmonary

    metastases. Int J Cancer 2001;94:531539.

    19. Markovic SN, et al. Peptide vaccination of

    patients with metastatic melanoma: improved

    clinical outcome in patients demonstrating

    effective immunization. Am J Clin Oncol

    2006;29:352360.

    20. Gjertsen MK, et al. Intradermal ras peptide

    vaccination with granulocyte-macrophage

    colony-stimulating factor as adjuvant: clinical

    and immunological responses in patients

    with pancreatic adenocarcinoma. Int J Cancer

    2001;92:441450.

    21. Vonderheide RH, et al. Vaccination of cancer

    patients against telomerase induces functional

    antitumor CD81 T lymphocytes. Clin Cancer

    Res 2004;10:828839.

    22. Gajewski TF, et al. Immune resistance orche-

    strated by the tumor microenvironment.

    Immunol Rev 2006;213:131145.

    23. Zou W. Immunosuppressive networks in the

    tumour environment and their therapeutic

    relevance. Nat Rev Cancer 2005;5:263274.

    24. Klebanoff CA, Gattinoni L, Restifo NP.

    CD81T-cell memory in tumor immunology

    and immunotherapy. Immunol Rev

    2006;211:214224.

    25. Zimmermann VS, et al. Tumors hamper the

    immunogenic competence of CD41 T cell-

    directed dendritic cell vaccination. J Immunol

    2007;179:28992909.

    26. Curiel TJ. Tregs and rethinking cancer

    immunotherapy. J Clin Invest 2007;

    117:11671174.

    27. Ercolini AM, et al. Recruitment of latent pools

    of high-avidity CD8(1) T cells to the anti-

    tumor immune response. J Exp Med

    2005;201:15911602.

    28. Viehl CT, et al. Depletion of CD41CD251

    regulatory T cells promotes a tumor-specific

    immune response in pancreas cancer-

    bearing mice. Ann Surg Oncol

    2006;13:12521258.

    29. Litzinger MT, Fernando R, Curiel TJ, Grosen-

    bach DW, Schlom J, Palena C. The IL-2

    immunotoxin denileukin diftitox reduces

    regulatory T cells and enhances vaccine-

    mediated T-cell immunity. Blood

    2007;110:31923201.

    30. Dannull J, et al. Enhancement of vaccine-

    mediated antitumor immunity in cancer

    patients after depletion of regulatory T cells.

    J Clin Invest 2005;115:36233633.

    31. Shen L, Evel-Kabler K, Strube R, Chen SY.

    Silencing of SOCS1 enhances antigen presen-

    tation by dendritic cells and antigen-specific

    anti-tumor immunity. Nat Biotechnol

    2004;22:15461553.

    32. Evel-Kabler K, Song XT, Aldrich M, Huang XF,

    Chen SY. SOCS1 restricts dendritic cells abil-

    ity to break self tolerance and induce anti-

    tumor immunity by regulating IL-12

    production and signaling. J Clin Invest

    2006;116:90100.

    33. Finn OJ. Premalignant lesions as targets for

    cancer vaccines. J Exp Med

    2003;198:16231626.

    34. Shu S, Cochran AJ, Huang RR, Morton DL,

    Maecker HT. Immune responses in the drain-

    ing lymph nodes against cancer: implications

    for immunotherapy. Cancer Metastasis Rev

    2006;25:233242.

    35. Celis E. Overlapping human leukocyte anti-

    gen class I/II binding peptide vaccine for the

    treatment of patients with stage IV melano-

    ma: evidence of systemic immune dysfunc-

    tion. Cancer 2007;110:203214.

    36. Lambeck AJ, et al. Serum cytokine profiling as

    a diagnostic and prognostic tool in ovarian

    cancer: a potential role for interleukin 7. Clin

    Cancer Res 2007;13:23852391.

    37. Miller AM, Pisa P. Tumor escape mechanisms

    in prostate cancer. Cancer Immunol Immun-

    other 2007;56:8187.

    38. Mitchell MS. Combinations of anticancer

    drugs and immunotherapy. Cancer Immunol

    Immunother 2003;52:686692.

    39. Casati A, Zimmermann VS, Benigni F, Berti-

    laccio MT, Bellone M, Mondino A. The im-

    munogenicity of dendritic cell-based vaccines

    is not hampered by doxorubicin and mel-

    phalan administration. J Immunol

    2005;174:33173325.

    40. Emens LA, Jaffee EM. Leveraging the activity

    of tumor vaccines with cytotoxic chemother-

    apy. Cancer Res 2005;65:80598064.

    41. Gulley JL, et al. Combining a recombinant

    cancer vaccine with standard definitive

    radiotherapy in patients with localized pros-

    tate cancer. Clin Cancer Res

    2005;11:33533362.

    42. Small LA, et al. A murine model for the

    effects of pelvic radiation and cisplatin

    chemotherapy on human papillomavirus

    vaccine efficacy. Clin Cancer Res 2001;7:

    876s881s.

    43. Hsueh EC, et al. Prolonged survival after

    complete resection of disseminated melano-

    ma and active immunotherapy with a ther-

    apeutic cancer vaccine. J Clin Oncol

    2002;20:45494554.

    44. Berd D, Sato T, Maguire HC, Jr, Kairys J,

    Mastrangelo MJ. Immunopharmacologic

    analysis of an autologous, hapten-modified

    human melanoma vaccine. J Clin Oncol

    2004;22:403415.

    45. Hsueh EC, Famatiga E, Shu S, Ye X, Morton

    DL. Peripheral blood CD41T-cell response

    before postoperative active immunotherapy

    correlates with clinical outcome in metastatic

    melanoma. Ann Surg Oncol

    2004;11:892899.

    46. Takeuchi H, Morton DL, Elashoff D, Hoon DS.

    Survivin expression by metastatic melanoma

    predicts poor disease outcome in patients

    receiving adjuvant polyvalent vaccine. Int

    J Cancer 2005;117:10321038.

    47. Chung MH, et al. Humoral immune response

    to a therapeutic polyvalent cancer vaccine

    after complete resection of thick primary

    melanoma and sentinel lymphadenectomy.

    J Clin Oncol 2003;21:313319.

    48. Wallack MK, et al. Increased survival of

    patients treated with a vaccinia melanoma

    oncolysate vaccine: second interim analysis of

    data from a phase III, multi-institutional trial.

    Ann Surg 1997;226:198206.

    49. Weber J, et al. Granulocyte-macrophage-

    colony-stimulating factor added to a

    multipeptide vaccine for resected stage II

    melanoma. Cancer 2003;97:186200.

    50. Sosman JA, et al. Adjuvant immunotherapy

    of resected, intermediate-thickness, node-

    negative melanoma with an allogeneic tumor

    vaccine: impact of HLA class I antigen ex-

    pression on outcome. J Clin Oncol

    2002;20:20672075.

    51. Livingston PO, et al. Improved survival in

    stage III melanoma patients with GM2 anti-

    bodies: a randomized trial of adjuvant

    vaccination with GM2 ganglioside. J Clin

    Oncol 1994;12:10361044.

    52. Wang F, et al. Phase I trial of a MART-1

    peptide vaccine with incomplete freunds

    adjuvant for resected high-risk melanoma.

    Clin Cancer Res 1999;5:27562765.

    53. Tagawa ST, Cheung E, Banta W, Gee C, Weber

    JS. Survival analysis after resection of meta-

    static disease followed by peptide vaccines in

    patients with stage IV melanoma. Cancer

    2006;106:13531357.

    54. Sanderson K, et al. Autoimmunity in a phase I

    trial of a fully human anti-cytotoxic T-lym-

    phocyte antigen-4 monoclonal antibody with

    326 Immunological Reviews 222/2008

    Gray et al Therapeutic vaccines in cancer prevention

  • multiple melanoma peptides and Montanide

    ISA 51 for patients with resected stages III and

    IV melanoma. J Clin Oncol

    2005;23:741750.

    55. Kirkwood JM, et al. High-dose interferon

    alfa-2b significantly prolongs relapse-free and

    overall survival compared with the GM2-

    KLH/QS-21 vaccine in patients with resected

    stage IIB-III melanoma: results of intergroup

    trial E1694/S9512/C509801. J Clin Oncol

    2001;19:23702380.

    56. Hsu FJ, et al. Tumor-specific idiotype vaccines

    in the treatment of patients with B-cell lym-

    phoma long-term results of a clinical trial.

    Blood 1997;89:31293135.

    57. Weng WK, Czerwinski D, Timmerman J, Hsu

    FJ, Levy R. Clinical outcome of lymphoma

    patients after idiotype vaccination is corre-

    lated with humoral immune response and

    immunoglobulin G FC receptor genotype. J

    Clin Oncol 2004;22:47174724.

    58. Reinartz S, et al. Vaccination of patients with

    advanced ovarian carcinoma with the anti-

    idiotype ACA125: immunological response

    and survival (phase Ib/II). Clin Cancer Res

    2004;10:15801587.

    59. Berek JS, et al. Randomized, placebo-con-

    trolled study of oregovomab for consolida-

    tion of clinical remission in patients with

    advanced ovarian cancer. J Clin Oncol

    2004;22:35073516.

    60. Kimura H, Yamaguchi Y. A phase III rando-

    mized study of interleukin-2 lymphokine-

    activated killer cell immunotherapy com-

    bined with chemotherapy or radiotherapy

    after curative or noncurative resection of

    primary lung carcinoma. Cancer

    1997;80:4249.

    61. Giaccone G, et al. Phase III study of adjuvant

    vaccination with Bec2/bacille calmette-guer-

    in in responding patients with limited-dis-

    ease small-cell lung cancer (European

    organisation for research and treatment of

    cancer 08971-08971B; silva study). J Clin

    Oncol 2005;23:68546864.

    62. Jaffee EM, et al. Novel allogeneic granulocyte-

    macrophage colony-stimulating factor-

    secreting tumor vaccine for pancreatic

    cancer: a phase I trial of safety and

    immune activation. J Clin Oncol 2001;19:

    145156.

    63. Sportes C, et al. Establishing a platform for

    immunotherapy: clinical outcome and study

    of immune reconstitution after high-dose

    chemotherapy with progenitor cell support

    in breast cancer patients. Biol Blood Marrow

    Transplant 2005;11:472483.

    64. Danna EA, Sinha P, Gilbert M, Clements VK,

    Pulaski BA, Ostrand-Rosenberg S. Surgical

    removal of primary tumor reverses tumor-

    induced immunosuppression despite the

    presence of metastatic disease. Cancer Res

    2004;64:22052211.

    65. Finn OJ, Forni G. Prophylactic cancer vac-

    cines. Curr Opin Immunol 2002;14:

    172177.

    66. Garcia-Hernandez Mde L, Gray A, Hubby B,

    Kast WM. In vivo effects of vaccination with

    six-transmembrane epithelial antigen of the

    prostate: a candidate antigen for treating

    prostate cancer. Cancer Res

    2007;67:13441351.

    67. Garcia-Hernandez Mde L, Gray A, Hubby B,

    Klinger OJ, Kast WM. Prostate stem cell

    antigen vaccination induces a long-term pro-

    tective immune response against prostate

    cancer in the absence of autoimmunity. Can-

    cer Res 2008;68:861869.

    68. Wang X, Wang JP, Maughan MF, Lachman LB.

    Alphavirus replicon particles containing the

    gene for HER2/neu inhibit breast cancer

    growth and tumorigenesis. Breast Cancer Res

    2005;7:R145R155.

    69. Mwau M, et al. A human immunodeficiency

    virus 1 (HIV-1) clade a vaccine in clinical

    trials: stimulation of hiv-specific T-cell re-

    sponses by DNA and recombinant modified

    vaccinia virus ankara (MVA) vaccines in hu-

    mans. J Gen Virol 2004;85:911919.

    70. Hubert RS, et al. STEAP: a prostate-specific

    cell-surface antigen highly expressed in hu-

    man prostate tumors. Proc Natl Acad Sci USA

    1999;96:1452314528.

    71. Gu Z, et al. Prostate stem cell antigen (PSCA)

    expression increases with high gleason score,

    advanced stage and bone metastasis in pros-

    tate cancer. Oncogene 2000;19:12881296.

    72. Yang D, Holt GE, Velders MP, Kwon ED, Kast

    WM. Murine six-transmembrane epithelial

    antigen of the prostate, prostate stem cell

    antigen, and prostate-specific membrane

    antigen: prostate-specific cell-surface anti-

    gens highly expressed in prostate cancer of

    transgenic adenocarcinoma mouse prostate

    mice. Cancer Res 2001;61:58575860.

    73. Hale JS, Boursalian TE, Turk GL, Fink PJ.

    Thymic output in aged mice. Proc Natl Acad

    Sci USA 2006;103:84478452.

    74. Nava-Parada P, Forni G, Knutson KL, Pease LR,

    Celis E. Peptide vaccine given with a toll-like

    receptor agonist is effective for the

    treatment and prevention of spontaneous

    breast tumors. Cancer Res 2007;67:

    13261334.

    75. Muderspach L, et al. A phase I trial of a

    human papillomavirus (HPV) peptide vaccine

    for women with high-grade cervical and

    vulvar intraepithelial neoplasia who are HPV

    16 positive. Clin Cancer Res

    2000;6:34063416.

    76. Roman LD, et al. A phase II study of Hsp-7

    (SGN-00101) in women with high-grade

    cervical intraepithelial neoplasia. Gynecol

    Oncol 2007;106:558566.

    77. Einstein MH, et al. Heat shock fusion protein-

    based immunotherapy for treatment of cer-

    vical intraepithelial neoplasia III. Gynecol

    Oncol 2007;106:453460.

    78. Corona Gutierrez CM, et al. Therapeutic

    vaccination with MVA E2 can eliminate pre-

    cancerous lesions (CIN 1, CIN 2, and CIN 3)

    associated with infection by oncogenic hu-

    man papillomavirus. Hum Gene Ther

    2004;15:421431.

    79. Garcia-Hernandez E, et al. Regression of

    papilloma high-grade lesions (CIN 2 and CIN

    3) is stimulated by therapeutic vaccination

    with MVA E2 recombinant vaccine. Cancer

    Gene Ther 2006;13:592597.

    80. Brinkman JA, et al. Therapeutic vaccination

    for HPV induced cervical cancers. Dis Markers

    2007;23:337352.

    81. Kanodia S, Fahey LM, Kast WM. Mechanisms

    used by human papillomaviruses to escape

    the host immune response. Curr Cancer Drug

    Targets 2007;7:7989.

    82. Ressing ME, et al. Occasional memory cyto-

    toxic T-cell responses of patients with human

    papillomavirus type 16-positive cervical le-

    sions against a human leukocyte antigen-A0201-restricted E7-encoded epitope. CancerRes 1996;56:582588.

    83. Schlom J, Arlen PM, Gulley JL. Cancer

    vaccines: moving beyond current

    paradigms. Clin Cancer Res 2007;13:

    37763782.

    84. Antonia SJ, et al. Combination of p53 cancer

    vaccine with chemotherapy in patients with

    extensive stage small cell lung cancer. Clin

    Cancer Res 2006;12:878887.

    85. Arlen PM, et al. Antiandrogen, vaccine and

    combination therapy in patients with non-

    metastatic hormone refractory prostate can-

    cer. J Urol 2005;174:539546.

    86. Arlen PM, et al. A randomized phase II study

    of concurrent docetaxel plus vaccine versus

    vaccine alone in metastatic androgen-inde-

    pendent prostate cancer. Clin Cancer Res

    2006;12:12601269.

    87. Salgaller ML, et al. Report of immune mon-

    itoring of prostate cancer patients under-

    going T-cell therapy using dendritic cells

    pulsed with HLA-A2-specific peptides from

    prostate-specific membrane antigen (psma).

    Prostate 1998;35:144151.

    88. Cho WC. Contribution of oncoproteomics to

    cancer biomarker discovery. Mol Cancer

    2007;6:25.

    89. Micke P, et al. Staging small cell lung

    cancer: Veterans Administration Lung Study

    Group versus International Association for

    the Study of Lung Cancer what limits

    limited disease? Lung Cancer 2002;

    37:271276.

    90. Peoples GE, et al. Clinical trial results

    of a HER2/neu (E75) vaccine to prevent

    recurrence in high-risk breast cancer

    patients. J Clin Oncol 2005;23:

    75367545.

    Immunological Reviews 222/2008 327

    Gray et al Therapeutic vaccines in cancer prevention