immunological risks of adult t-cell leukemia at primary htlv-i infection

7
Immunological risks of adult T-cell leukemia at primary HTLV-I infection Mari Kannagi, Takashi Ohashi, Nanae Harashima, Shino Hanabuchi and Atsuhiko Hasegawa Department of Immunotherapeutics, Medical Research Division, Tokyo Medical and Dental University, Tokyo 113-8519, Japan A small percentage of human T-cell leukemia virus type-I (HTLV-I)-infected individuals develop adult T-cell leukemia (ATL). In animal experiments, inoculation of HTLV-I via the oral route, which is the main route of mother-to-child viral transmission in humans as a result of breastfeeding, induced host HTLV-I-specific T-cell unresponsiveness and resulted in increased viral load. This strongly suggested that the known epidemiologi- cal risk factors for ATL (i.e. vertical HTLV-I infection and elevated viral load) are linked by an insufficient HTLV-I- specific T-cell response. Recent findings on the anti- tumor effects of Tax-targeted vaccination in rats and the reactivation of Tax-specific T cells in ATL patients as a result of hematopoietic stem cell transplantation imply promising immunological approaches for the prophylaxis and therapy of ATL. Human T-cell leukemia virus type-I (HTLV-I) is etiologically linked to adult T-cell leukemia (ATL) [1,2] and is found in Japan, the Caribbean, South America, Africa, Melanesia and the Middle East [3]. In Japan, it is estimated that approximately one million people are infected with HTLV-I, and that 1–5% of infected individuals develop ATL [4]. Most other HTLV-I carriers are asymptomatic throughout their lives, however, a small fraction develop a chronic inflam- matory neurological disorder termed HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) [5,6] or other inflammatory disorders. HTLV-I transmission occurs via both vertical and horizontal routes. The main vertical transmission route is via breastfeeding [7,8]. Horizontal transmission routes include sexual transmission (mainly from male to female) and blood transfusion or intravenous injection of contami- nated blood [9]. HTLV-I infection occurs mainly through cell-to-cell contact of infected cells with a wide variety of target cells in vitro [10]. Sera from HTLV-I-infected individuals are capable of neutralizing HTLV-I-mediated membrane fusions [11]. Recently, it was shown that the ubiquitous glucose transporter, known as GLUT-1, is a specific component of the HTLV receptor and plays an essential role in HTLV envelope-mediated infection [12]. ATL is characterized by CD4 þ and CD25 þ mature T lymphocyte phenotypes, disease onset during middle age or later, immune suppression and poor prognosis as a result of resistance to combination chemotherapy [13,14]. Recently, hematopoietic stem cell transplantation (HSCT) was applied to a limited number of ATL patients. Initial studies of autologous HSCT revealed a frequent recur- rence of ATL [15], however, more recent reports have revealed that allogeneic HSCT can produce better results, although there is a risk of graft-versus-host disease (GVHD), which is sometimes lethal [16]. Further improve- ments or new approaches are therefore required for treatment of ATL. HTLV-I Tax, one of the viral proteins, transactivates and interacts with many cellular proteins that regulate or dysregulate cell growth [17], partly accounting for the mechanisms of HTLV-I-induced leukemogenesis. How- ever, it has been suggested that in half of the cases of ATL the leukemia cells might not be able to express Tax because of mutations and deletions, mainly within the 5 0 long terminal repeat (LTR) region of the gene [18,19]. In the remainder of ATL cases, leukemia cells retain the ability to express HTLV-I antigens, but the viral antigens are only detectable after several hours of in vitro cultivation [20–22]. A similar transient silencing of HTLV-I is observed not only in leukemia cells but also in peripheral blood mononuclear cells (PBMCs) of HAM/TSP patients and HTLV-I carriers [23,24]. These observations suggest that although Tax contributes to the early steps of HTLV-I leukemogenesis, multiple additional steps that substitute for Tax functions might be required for ATL development. Constitutive activation of nuclear factor (NF)-kB [25] and JAK–STAT (Janus kinase–signal transducers and activators of transciption) [26] pathways has been shown in ATL cells. It remains to be clarified whether anything in addition to Tax is activating these path- ways in ATL cells. Although consistent differences have not been observed among HTLV-I strains isolated from ATL and HAM/TSP patients [27,28], immunological studies have found a clear difference in HTLV-I-specific T-cell immune responses among HTLV-I-related diseases. HTLV-I-specific cytotoxic T lymphocytes (CTLs) are highly activated in HAM/TSP patients and can be induced from PBMCs of asymptomatic carriers but only rarely from those of ATL patients in vitro [23,29–33]. The nature of insufficient HTLV-I-specific T-cell immunity in ATL patients is not fully understood. A recent report indicated that some CD8 þ HTLV-I-specific CTLs persist in ATL patients although they mostly fail to Corresponding author: Mari Kannagi ([email protected]). Available online 4 June 2004 Review TRENDS in Microbiology Vol.12 No.7 July 2004 www.sciencedirect.com 0966-842X/$ - see front matter q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.tim.2004.05.005

Upload: mari-kannagi

Post on 29-Oct-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

Immunological risks of adult T-cellleukemia at primary HTLV-I infectionMari Kannagi, Takashi Ohashi, Nanae Harashima, Shino Hanabuchi and

Atsuhiko Hasegawa

Department of Immunotherapeutics, Medical Research Division, Tokyo Medical and Dental University, Tokyo 113-8519, Japan

A small percentage of human T-cell leukemia virus

type-I (HTLV-I)-infected individuals develop adult T-cell

leukemia (ATL). In animal experiments, inoculation of

HTLV-I via the oral route, which is the main route of

mother-to-child viral transmission in humans as a result

of breastfeeding, induced host HTLV-I-specific T-cell

unresponsiveness and resulted in increased viral load.

This strongly suggested that the known epidemiologi-

cal risk factors for ATL (i.e. vertical HTLV-I infection and

elevated viral load) are linked by an insufficient HTLV-I-

specific T-cell response. Recent findings on the anti-

tumor effects of Tax-targeted vaccination in rats and

the reactivation of Tax-specific T cells in ATL patients as

a result of hematopoietic stem cell transplantation

imply promising immunological approaches for the

prophylaxis and therapy of ATL.

Human T-cell leukemia virus type-I (HTLV-I) is etiologicallylinked to adult T-cell leukemia (ATL) [1,2] and is found inJapan, the Caribbean, South America, Africa, Melanesiaand the Middle East [3]. In Japan, it is estimated thatapproximately one million people are infected with HTLV-I,and that 1–5% of infected individuals develop ATL [4]. Mostother HTLV-I carriers are asymptomatic throughout theirlives, however, a small fraction develop a chronic inflam-matory neurological disorder termed HTLV-1-associatedmyelopathy/tropical spastic paraparesis (HAM/TSP) [5,6] orother inflammatory disorders.

HTLV-I transmission occurs via both vertical andhorizontal routes. The main vertical transmission routeis via breastfeeding [7,8]. Horizontal transmission routesinclude sexual transmission (mainly from male to female)and blood transfusion or intravenous injection of contami-nated blood [9]. HTLV-I infection occurs mainly throughcell-to-cell contact of infected cells with a wide variety oftarget cells in vitro [10]. Sera from HTLV-I-infectedindividuals are capable of neutralizing HTLV-I-mediatedmembrane fusions [11]. Recently, it was shown that theubiquitous glucose transporter, known as GLUT-1, is aspecific component of the HTLV receptor and plays anessential role in HTLV envelope-mediated infection [12].

ATL is characterized by CD4þ and CD25þ matureT lymphocyte phenotypes, disease onset during middle ageor later, immune suppression and poor prognosis as a

result of resistance to combination chemotherapy [13,14].Recently, hematopoietic stem cell transplantation (HSCT)was applied to a limited number of ATL patients. Initialstudies of autologous HSCT revealed a frequent recur-rence of ATL [15], however, more recent reports haverevealed that allogeneic HSCT can produce better results,although there is a risk of graft-versus-host disease(GVHD), which is sometimes lethal [16]. Further improve-ments or new approaches are therefore required fortreatment of ATL.

HTLV-I Tax, one of the viral proteins, transactivatesand interacts with many cellular proteins that regulate ordysregulate cell growth [17], partly accounting for themechanisms of HTLV-I-induced leukemogenesis. How-ever, it has been suggested that in half of the cases of ATLthe leukemia cells might not be able to express Tax becauseof mutations and deletions, mainly within the 50 longterminal repeat (LTR) region of the gene [18,19]. In theremainder of ATL cases, leukemia cells retain the ability toexpress HTLV-I antigens, but the viral antigens are onlydetectable after several hours of in vitro cultivation[20–22]. A similar transient silencing of HTLV-I isobserved not only in leukemia cells but also in peripheralblood mononuclear cells (PBMCs) of HAM/TSP patientsand HTLV-I carriers [23,24]. These observations suggestthat although Tax contributes to the early steps of HTLV-Ileukemogenesis, multiple additional steps that substitutefor Tax functions might be required for ATL development.Constitutive activation of nuclear factor (NF)-kB [25] andJAK–STAT (Janus kinase–signal transducers andactivators of transciption) [26] pathways has beenshown in ATL cells. It remains to be clarified whetheranything in addition to Tax is activating these path-ways in ATL cells.

Although consistent differences have not been observedamong HTLV-I strains isolated from ATL and HAM/TSPpatients [27,28], immunological studies have found a cleardifference in HTLV-I-specific T-cell immune responsesamong HTLV-I-related diseases. HTLV-I-specific cytotoxicT lymphocytes (CTLs) are highly activated in HAM/TSPpatients and can be induced from PBMCs of asymptomaticcarriers but only rarely from those of ATL patients in vitro[23,29–33]. The nature of insufficient HTLV-I-specificT-cell immunity in ATL patients is not fully understood. Arecent report indicated that some CD8þ HTLV-I-specificCTLs persist in ATL patients although they mostly fail to

Corresponding author: Mari Kannagi ([email protected]).

Available online 4 June 2004

Review TRENDS in Microbiology Vol.12 No.7 July 2004

www.sciencedirect.com 0966-842X/$ - see front matter q 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.tim.2004.05.005

Page 2: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

respond ex vivo [34], suggesting the involvement ofimmune suppressive or tolerating mechanisms.

In HAM/TSP, the co-presence of Tax mRNA and anactivated HTLV-I-specific T-cell response in the centralnervous system strongly indicates the involvement ofimmunopathological mechanisms [35,36]. Neurologicalsymptoms improve in response to oral prednisone treat-ment in less advanced cases of HAM/TSP [37]. The use ofanti-retroviral treatment for HAM/TSP patients partlyreduces HTLV-I proviral load, suggesting that HTLV-Imight spread from cell-to-cell in these patients [38].Spontaneous HTLV-I infection in peripheral HTLV-I-specific CTLs from some HAM/TSP patients [24] alsoindicates active interaction between host immunity andthe virus.

HTLV-I core, envelope, polymerase, Tax, Tof and Rofproteins are known to be recognized by HTLV-I-specificCTLs [31,32,39–41]. Among these antigens, HTLV-I Tax(a crucial viral protein for T cell immortalization) is themost popular target for HTLV-I-specific CTLs in infectedindividuals [31,39]. Infrequent sequence variations arefound in the Tax gene, even in HAM/TSP patients [42],indicating the importance of this protein for maintenanceof the virus. HTLV-I Tax-specific CTLs are capable oflysing short-term cultured ATL cells in vitro [23,33]. Thesefindings together with the insufficient HTLV-I-specificCTL response in ATL patients suggests that HTLV-I-specific CTLs contribute to anti-tumor surveillance inHTLV-I-infected individuals, and that an insufficiency ofT-cell immune responses to HTLV-I might be a risk factorfor ATL development.

To attest this hypothesis and understand the mechan-isms of HTLV-I leukemogenesis in vivo, a series of ratmodels for HTLV-I infection and T-cell lymphomas wereinvestigated. From these experiments, as described later,it was discovered that the route of HTLV-I transmissionseriously affects host immune responses [43,44], and thatthe proviral load increases under insufficient HTLV-I-specific T-cell responses. Furthermore, HTLV-I Tax waswidely recognized by rat and human CTLs, and vacci-nation directed at Tax eradicated ATL-like lymphomas inrats [45,46].

More recently, HTLV-I Tax-specific CTL responses werefound to be strongly reactivated in ATL patients followingHSCT [47], also supporting the notion that HTLV-I-specificT-cell immune responses contribute towards the surveil-lance of HTLV-I-infected lymphoproliferative disease.

Risk factors for ATL development

In cohort studies of HTLV-I carriers, it appears that therisk factors for ATL might include vertical HTLV-Iinfection, gender (male . female) and increasing numbersof abnormal lymphocytes [48,49]. Because the proportionof vertically infected female HTLV-I carriers is relativelylow compared with male carriers (owing to horizontal viraltransmission from husbands to wives), the higher inci-dence of ATL in male HTLV-I carriers is attributed tovertical infection [9]. Genetic analysis has indicated thepresence of typical human leukocyte antigen (HLA)haplotypes for ATL in an endemic area [50,51], whichalso implies that vertical infection might transmit some

determinants of HTLV-I leukemogenesis. Althoughimmunological studies have pointed out clear differencesin the magnitude of HTLV-I-specific cytotoxic T-cellresponses between ATL and HAM/TSP patients, longi-tudinal studies on T-cell immunity in HTLV-I carrierstoward ATL development have never been conducted.Therefore, it is not known whether the insufficiency inHTLV-I-specific T-cell responses in ATL patients isspontaneously associated or acquired after the onset ofATL. The reasons for the wide variety of immuneresponses to HTLV-I among carriers of the virus are alsounknown. One possibility is the influence of verticalHTLV-I infection, as ATL occurs mainly in verticallyinfected individuals but not in those who become infectedlater in life [9]. To assess this possibility, animalexperiments have been conducted to examine the variousroutes of HTLV-I infection.

Immune unresponsiveness to HTLV-I in animal models

Rats can be persistently infected with HTLV-I [52]. Toexamine whether the route of primary HTLV-I infectionaffects host T-cell immunity to HTLV-I, 5 £ 107 MitomycinC (MMC)-treated MT-2 cells were inoculated into immu-nocompetent adult rats either orally or intraperitoneally.HTLV-I-specific antibody responses were significantlylower in orally infected rats [43,44]. Significant levels ofHTLV-I-specific T-cell immune responses were detected byT-cell proliferation or interferon (IFN)-g production inintraperitoneally infected rats but scarcely in orallyinfected rats. The responses of T cells from rats inoculatedorally were indistinguishable from those of naive rats. Thisclearly indicates that the conditions of primary infection,especially when occurring via the oral route, severelyaffect host immune responses to HTLV-I, even if thegenetic backgrounds of the host are identical.

By contrast, real-time PCR analysis revealed that theHTLV-I proviral load in the spleen cells of orally infectedrats was significantly higher than that detected inintraperitoneally infected rats, although some individualdifferences did exist. Such variation in the levels of T-cellimmunity and proviral loads detected implies that the lowimmune response in orally infected rats is not due to ascarcity of antigens.

When rats were orally or intraperitoneally inoculatedwith low doses of HTLV-I (50 MMC-treated MT-2 cells),viral-specific antibody responses were undetectable in allthose infected, and slight IFN-g production was detectableonly in Tcells from intraperitoneally infected rats. Some ofthe rats orally inoculated with 50 cells showed high levelsof proviruses, whereas intraperitoneally infected rats hadlow provirus levels. Therefore, a scarcity of viral antigensin vivo might explain the poor immune responses of theintraperitoneally infected rats but not those orallyinfected.

Some of the rats that had been orally inoculated with 50cells showed elevated levels of viral load. This suggeststhat even when the initial number of HTLV-I-infected cellsis extremely small, once such cells begin to grow in vivo,the orally infected host might not have any control overthem. The initial amount of virus used at primary infectiondid not markedly affect the persistent proviral load within

Review TRENDS in Microbiology Vol.12 No.7 July 2004 347

www.sciencedirect.com

Page 3: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

either the orally or the intraperitoneally infected group.This observation suggests that the route of infection morestrongly influences the persistent HTLV-I load in the hostthan does the amount of virus delivered.

Relationship between HTLV-I load and virus-specific

T-cell immunity

In the above rat experiments, the relationship betweenHTLV-I proviral load and T-cell proliferative responses toHTLV-I was analyzed. As shown in Figure 1, the rats canbe roughly divided into three groups: (A) a high viral loadand low T-cell response group, (B) a low viral load and lowT-cell response group, and (C) a low viral load and highT-cell response group. Group A and C consisted exclu-sively of orally and intraperitoneally infected rats,respectively, whereas group B contained both. Overall,there is an inverse correlation between HTLV-I pro-viral load and HTLV-I-specific T-cell proliferation, sug-gesting that HTLV-I-specific T-cell immune responsesmight contribute to the limited expansion of HTLV-I-infected cells in vivo, and that the magnitude of hostimmune response at primary infection might be acrucial determinant of persistent HTLV-I levels in vivo.

In humans, vertical HTLV-I infection, high viral loadand low T-cell responses to HTLV-I are incidentallyassociated with ATL. This exhibits a striking similarityto the oral HTLV-I infection profile demonstrated in the ratexperiments. Because oral infection is a major route ofvertical HTLV-I transmission, a similar immune unre-sponsiveness to HTLV-I and subsequent enlargement ofthe HTLV-I-infected cell population might also occur in

humans, providing conditions that favor the evolution ofinfected cells towards more malignant phenotypes.

Recovery of HTLV-I-specific T-cell immunity in orally

infected rats

It remains unclear how HTLV-I infection occurs throughintestinal mucosa and why T-cell responses are insufficientin orally infected rats. Oral administration of proteinantigens is known to induce peripheral tolerance, in whichhost immunity does not respond to challenge adminis-tration of the original inoculum. [53,54]. However, HTLV-I-specific T-cell responses in orally infected rats wererestored when they were re-immunized with syngeneicHTLV-I-infected cells. Therefore, such immune unrespon-siveness might differ from the typical oral toleranceobserved in the case of non-infectious protein antigens.Alternatively, the syngeneic HTLV-I-infected cells used asa challenging immunogen could be strong antigen-pre-senting cells that are able to break T cell anergy.

HTLV-I-specific cellular immune unresponsivenessassociated with oral infection was reversible, suggestingthat potential immune tolerance in human babies infectedthrough maternal milk might also be reversible. Restor-ation of HTLV-I-specific cellular immunity by vaccinationin the early stage of their lives might reduce theunderlying risk of ATL. However, it appears thatsuch T-cell immunity restoration might also occurspontaneously in human HTLV-I carriers.

In addition to oral infection, there are some other minorroutes of vertical infection such as perinatal or transpla-cental infection, as a small percentage of children born byHTLV-I-infected mothers, but which are bottle-fed, areinfected with HTLV-I [55]. The immunological status ofthese children remains unknown.

Anti-tumor effects of Tax-specific CTL in animal models

Because HTLV-I exists mainly in association with cells andnot as a free virus in vivo, an increase in the levels ofproviruses implies HTLV-1-infected cell proliferation orcell-to-cell infection. However, even though there might besome expansion of infected cells in persistently HTLV-I-infected rats, malignant tumors failed to develop withintwo years of observation. This is partly explained by thetime taken for clonal evolution of the infected cells.

Several animal models have been developed to studythe pathogenesis of ATL by inoculating syngeneic HTLV-I-infected T-cell lines into newborn rats [56] or rabbits [57],or by inoculating human leukemia cells from ATL patientsinto immunodeficient mice [58]. However, there were somelimitations in applying these models for immunologicalstudies on HTLV-I-associated tumors. To analyze anti-tumor immunity during HTLV-I infection, another experi-mental model of HTLV-I-infected T-cell lymphomaswas established [59]. In this model, athymic F344/NJcl-rnu/rnu (nu/nu) rats were subcutaneously inoculatedwith the HTLV-I-transformed CD4þ T-cell line FPM1,derived from the thymocytes of a syngeneic nu/þ rat.Although FPM1 failed to cause tumors in the nu/nu rats,the cell line underwent phenotypic progression throughseveral passages in newborn nu/nu rats, resulting in theestablishment of a sub-line FPM1-V1AX; this sub-line

Figure 1. The relationship between human T-cell leukemia virus type-I (HTLV-I)

proviral loads and Tax-specific T-cell responses. A mild inverse correlation

(r ¼ 20.315, p ¼ 0.094) was observed between the relative copy number of HTLV-I

proviruses (vertical axis) and HTLV-I Tax-specific T-cell proliferative index (hori-

zontal axis) of rats that were inoculated intraperitoneally (circle) or orally (triangle)

with either 50 (open symbol) or 5 £ 107 (closed symbol) MMC-treated MT-2 cells

[44].

TRENDS in Microbiology

1

2

3

4

-10 5 10 25 100

5

6

50

HT

LV-I

pro

vira

l loa

d

T-cell proliferation index

A

B C

Review TRENDS in Microbiology Vol.12 No.7 July 2004348

www.sciencedirect.com

Page 4: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

reproducibly caused fatal systemic lymphomas in adultnu/nu rats with infiltration in the lymph nodes, lungs,liver and occasionally the ovaries and skin, as well as locallesions at the site of inoculation, within two months ofreceiving the inoculum. However, FPM1-V1AX failed togrow in syngeneic immunocompetent nu/þ rats.

The outline of the rat model of HTLV-I-infected T-celllymphomas is shown in Figure 2. The development ofFPM1-V1AX lymphomas in nu/nu rats was inhibited byadoptively transferred T-cell lymphocytes from syngeneicnu/þ rats that had been immunized with FPM1-V1AX.The effector Tcells that caused anti-tumor effects consistedof CTLs that predominantly recognized HTLV-I Tax [45].Fine mapping analysis revealed that Tax180–188(GAFLTNVPY) is the central target epitope of CTLs thatare restricted by the rat major histocompatibility complexclass I (MHC-I) molecule, known as RT1.Al. Furthermore,freshly prepared T cells from nu/þ rats vaccinated withTax180–188 oligopeptides prevented the development ofFPM1-V1AX-induced lymphomas in nu/nu rats [45].Similarly, DNA vaccinations with plasmids encoding TaxcDNA evoked effective anti-tumor T-cell immunity in vivo[46]. These results indicated that Tax is a major target ofHTLV-I-specific CTLs in rats as well as humans, and thatTax-specific CTLs play a crucial role in surveillanceagainst HTLV-I-infected tumor development in vivo.

Activation of Tax-specific CTLs in ATL patents following

HSCT

Although Tax-targeted vaccines in rat models of HTLV-I-induced T-cell lymphomas shows promising anti-tumoreffects, the rat model differs from the human disease inmany aspects. The most apparent difference is in viralexpression. FPM1-V1AX cells express much higher levelsof Tax than leukemia cells from ATL patients, and theirgrowth in vivo depends on Tax [60]. In another mousemodel with artificially expressed Tax–enhanced greenfluorescent protein (EGFP) fusion proteins that wereunder the control of the HTLV-I LTR, tumor growth was

suppressed in vivo by Tax-specific CTLs, whereas thetumor cells with low Tax–EGFP expression remained [61].

To investigate anti-tumor immunity in ATL, the T-cellresponses in ATL patients who underwent completeremission following non-myeloablative allogeneic periph-eral blood HSCT from HLA-identical sibling donors wereanalyzed [47] (Figure 3). These patients were presumed tohave raised T-cell immunity mediating graft-versus-hostand graft-versus-leukemia effects. As a target of suchresponses, a phytohemagglutinin (PHA)-stimulated inter-leukin (IL)-2-dependent T-cell line (ILT) derived from anATL patient before HSCT was established. ILT cells werespontaneously infected with HTLV-I. When PBMCs iso-lated from the same patient after HSCT were stimulatedwith autologous ILT cells in vitro, CD8þ CTLs capable ofkilling autologous ILT cells proliferated vigorously.Further analysis revealed that these CTLs predominantlyrecognized a single epitope Tax11–19 restricted withHLA-A2 (Figure 3). They also contained a minor popu-lation probably related to the graft-versus-host response.PBMCs obtained from the same patient before HSCT didnot respond to autologous ILT cells.

The reconstituted T-cell immunity derived from theHLA-identical donor, but not the original ATL patient,strongly reacted with HTLV-I Tax in the ATL patientsfollowing HSCT, indicating that the insufficiency ofHTLV-I-specific CTLs in the original ATL patient wasnot related to HLA or the absence of Tax expression.A similar oligoclonal expansion of Tax11–19-specific CTLshas been reported in HLA-A2 þ HAM/TSP patients [62],reflecting a highly activated host CTL response. Althoughthe direct contribution of Tax-specific CTLs to graft-versus-leukemia effects in the ATL patient remainscontroversial, a similar reactivation of Tax-specific CTLswas observed in other ATL patients who obtained completeremission following HSCT, supporting the notion thatthese CTLs might participate in the maintenance ofremission from ATL.

Hypothetical dynamics of HTLV-I-specific T-cell immune

conversion

For the previously described ATL patient who receivedHLA-identical HSCT, a new balance between host immu-nity and HTLV-I-infected cells was established. Thevigorous HTLV-I-specific CTL response observed in thispatient was very similar to that in HAM/TSP patients. Inthis sense, allogeneic HSCT converted HTLV-I-specificT-cell immunity in the recipient from one extreme tothe other, whereby host immunity might control themalignant expansion of HTLV-I-infected cells in vivo.

The immune unresponsiveness observed in orallyinfected rats might partially mimic insufficient T-cellresponses in some human HTLV-I carriers. Infants thatare born to HTLV-I-carrying mothers are estimated tointake,1 £ 108 HTLV-I-infected cells before weaning [63],and several infantile carriers remain seronegative forHTLV-I for a certain period of time [64], suggesting thatimmunological tolerance for HTLV-I infection might beestablished during this period. The majority of theseHTLV-I-infected individuals exhibit seroconversion withina few years [65]. Although T-cell immune responses to

Figure 2. A syngeneic rat model of human T-cell leukemia virus type-I (HTLV-I)-

infected lymphomas and tumor vaccines. An HTLV-I-transformed rat T-cell line

FPM1-V1AX derived from F344 Jcl rnu/þ (nu/þ) rat causes fatal systemic lympho-

mas in syngeneic athymic Jcl rnu/rnu (nu/nu) but not nu/þ rats when subcu-

taneously inoculated. Transfer of the spleen T cells from nu/þ rats inoculated with

FPM1-V1AX cells can eradicate tumors. By using this system, anti-tumor effects of

vaccines with Tax-coding DNA or synthetic peptides were observed [45,46,59].

TRENDS in Microbiology

nu/nu nu/+(no tumor)

nu/+

Fatallymphoma

Tumorregression

SpleenT-cell

VACCINEInfected cellTax-DNATax-peptide

FPM1-V1AX

(Inoculation)

(Transfer)

Review TRENDS in Microbiology Vol.12 No.7 July 2004 349

www.sciencedirect.com

Page 5: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

HTLV-I in children have not been investigated, it ispossible that they might also recover spontaneously laterin life, as many adult HTLV-I carriers exhibit HTLV-I-specific T-cell responses.

Figure 4 schematically demonstrates our currenthypothesis on immunological risks of ATL in the naturalcourse of HTLV-I infection. Vertically infected HTLV-Icarriers harbor risks of ATL (i.e. insufficient HTLV-I-specific T-cell responses and expansion of infected cells),although the levels can vary among individuals. However,such risks might be reduced in many of HTLV-I carriers byspontaneous recovery of HTLV-I-specific T cells. OnceT-cell immunity to HTLV-I recovers, the magnitude ofthe immune response would positively correlate withpre-existing viral load in vivo [66,67].

Some HTLV-I carriers within the low-risk group for ATLdevelop HAM/TSP. It remains unclear what determinesthe risk of HAM/TSP, but genetic factors might be involved[68]. The presence of a large amount of viral load andelevated levels of HTLV-I-specific immunity in HAM/TSPpatients might be partially explained by T-cell immuneconversion long after the establishment of persistent viralloads following vertical infection.

Nevertheless, a small population of vertically infectedHTLV-I carriers remains in a state consisting of low T-cellresponses to HTLV-I despite an abundant viral load, untillater in life. We assume that this population might be thehigh-risk group for ATL. This hypothesis should beattested by a wide survey of cellular immune responsesin HTLV-I carriers, especially at young ages. This is alsorequired for identification of high-risk groups for ATL.

Concluding remarks

Although the exact mechanisms of ATL developmentremain unclear, these results indicate that immuneunresponsiveness to HTLV-I might be established at avery early stage of HTLV-I infection, and that it is one

Figure 3. Analysis of T cell immunity in adult T-cell leukemia (ATL) patients following hematopoietic stem cell transplantation (HSCT). Peripheral blood mononuclear cells

(PBMCs) from an ATL patient after HSCT were co-cultured with formalin-treated HTLV-I-infected T-cell line (ILT) cells that had been established from the same patient

before HSCT. In this culture, HLA-A2-restricted Tax11–19-specific CD8þ CTLs capable of killing ILT cells predominantly proliferated. Reproduced, with permission, from

Ref. [47].

CD8

HLA-identicaldonor

HTLV-I+

T-cell line (ILT)

ATL patientafter HSCT

(complete remission)

ATL patientbefore HSCT

HSCT

PBMC

Coculture in thepresence of IL-2 H

LA-A

2/Ta

x11-

19 T

etra

mer 63%

101

102

103

104

100

100 101 102 103 104

Figure 4. Hypothesis on immunological risks for adult T-cell leukemia (ATL) in the

natural course of human T-cell leukemia virus type-I (HTLV-I) infection. Vertically

infected HTLV-I carriers harbor risks of ATL (i.e. insufficient HTLV-I-specific T-cell

response and expansion of infected cells). At a later time-point, HTLV-I-specific

T-cell responses spontaneously recover in most of these carriers and the risks for

ATL decrease. However, a small population remains in the high-risk group. HTLV-I

carriers infected through other routes might have less risks of ATL. Under insuffi-

cient T-cell immunity, HTLV-I-infected cells accumulate additional mutations

towards ATL.

TRENDS in Microbiology

Vertical infection

AsymptomaticHTLV-I carriers

(high risk group)

HAM/TSP

ATL

Additionalmutations

(Risk for ATL)

AsymptomaticHTLV-I carriers(low risk group)

Immunetoleranceto HTLV-I

Expansion ofinfected cells

Horizontalinfection

T-cell immuneconversion

Prophylacticvaccine

Therapeuticvaccine?

Escapemutants?

Immunesuppressive

reagents

Review TRENDS in Microbiology Vol.12 No.7 July 2004350

www.sciencedirect.com

Page 6: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

of the risk factors of ATL as it allows the expansion ofHTLV-I-infected cells. In this regard, the reactivationof HTLV-I Tax-specific T-cell immunity by vaccinationsmight contribute to prophylaxis for ATL developmentand also to ATL therapy to some extent.

AcknowledgementsWe thank Atae Utsunomiya (Imamura Bun-in Hospital, Kagoshima),Ryuji Tanosaki (National Cancer Center Hospital, Tokyo) and MasatoMasuda (University of the Ryukyus, Okinawa) for clinical collaborationand their excellent bedside achievements.

References

1 Hinuma, Y. et al. (1981) Adult T-cell leukemia: antigen in an ATL cellline and detection of antibodies to the antigen in human sera. Proc.Natl. Acad. Sci. U. S. A. 78, 6476–6480

2 Poiesz, B.J. et al. (1980) Detection and isolation of type C retrovirusparticles from fresh and cultured lymphocytes of a patient withcutaneous T-cell lymphoma. Proc. Natl. Acad. Sci. U. S. A. 77,7415–7419

3 Maloney, E.M. and Blattner, W.A. (2003) HTLV-I worldwide patternsand disease associations. In Gann Monograph on Cancer Research(Vol. 50) (Sugamura, K. et al., eds), pp. 339–361, Japan ScienceSocieties Press

4 Tajima, K. (1990) The 4th nation-wide study of adult T-cell leukemia/lymphoma (ATL) in Japan: estimates of risk of ATL and itsgeographical and clinical features. The T- and B-cell MalignancyStudy Group. Int. J. Cancer 45, 237–243

5 Gessain, A. et al. (1985) Antibodies to human T-lymphotropic virustype-I in patients with tropical spastic paraparesis. Lancet 2, 407–410

6 Osame, M. et al. (1986) HTLV-I associated myelopathy, a new clinicalentity. Lancet 1, 1031–1032

7 Hino, S. et al. (1985) Mother-to-child transmission of human T-cellleukemia virus type-I. Jpn. J. Cancer Res. 76, 474–480

8 Kinoshita, K. et al. (1987) Milk-borne transmission of HTLV-I fromcarrier mothers to their children. Jpn. J. Cancer Res. 78, 674–680

9 Tajima, K. and Hinuma, Y. (1992) Epidemiology of HTLV-I/II in Japanand the world. In Gann Monograph on Cancer Research (Vol. 39)(Yoshida, M., ed.), pp. 129, Japan Science Societies Press

10 Miyoshi, I. et al. (1981) Type C virus particles in a cord T-cell linederived by co-cultivating normal human cord leukocytes and humanleukaemic T cells. Nature 294, 770–771

11 Nagy, K. et al. (1983) Human T-cell leukemia virus type I: induction ofsyncytia and inhibition by patients’ sera. Int. J. Cancer 32, 321–328

12 Manel, N. et al. (2003) The ubiquitous glucose transporter GLUT-1 is areceptor for HTLV. Cell 115, 449–459

13 Uchiyama, T. et al. (1977) Adult T-cell leukemia: clinical andhematologic features of 16 cases. Blood 50, 481–492

14 Shimoyama, M. et al. (1988) Major prognostic factors of adult patientswith advanced T-cell lymphoma/leukemia. J. Clin. Oncol. 6,1088–1097

15 Tsukasaki, K. et al. (1999) Poor outcome of autologous stem celltransplantation for adult T cell leukemia/lymphoma: a case report andreview of the literature. Bone Marrow Transplant. 23, 87–89

16 Utsunomiya, A. et al. (2001) Improved outcome of adult T cellleukemia/lymphoma with allogeneic hematopoietic stem cell trans-plantation. Bone Marrow Transplant. 27, 15–20

17 Yoshida, M. (2001) Multiple viral strategies of HTLV-1 for dysregula-tion of cell growth control. Annu. Rev. Immunol. 19, 475–496

18 Konishi, H. et al. (1984) Defective human T-cell leukemia virus in adultT-cell leukemia patients. Mol. Biol. Med. 2, 273–283

19 Tamiya, S. et al. (1996) Two types of defective human T-lymphotropicvirus type I provirus in adult T-cell leukemia. Blood 88, 3065–3073

20 Hinuma, Y. et al. (1982) retrovirus associated with human adult T-cellleukemia: in vitro activation. Gann 73, 341–344

21 Kinoshita, T. et al. (1989) Detection of mRNA for the tax1/rex1 gene ofhuman T-cell leukemia virus type I in fresh peripheral bloodmononuclear cells of adult T-cell leukemia patients and viral carriersby using the polymerase chain reaction. Proc. Natl. Acad. Sci. U. S. A.86, 5620–5624

22 Uchiyama, T. (1997) Human T cell leukemia virus type I (HTLV-I) andhuman diseases. Annu. Rev. Immunol. 15, 15–37

23 Kannagi, M. et al. (1994) Cytotoxic T cell response and expression ofthe target antigen in HTLV-I infection. Leukemia 8, S54–S59

24 Hanon, E. et al. (2000) Fratricide among CD8(þ) T lymphocytesnaturally infected with human T cell lymphotropic virus type I.Immunity 13, 657–664

25 Mori, N. et al. (1999) Constitutive activation of NF-kappaB in primaryadult T-cell leukemia cells. Blood 93, 2360–2368

26 Takemoto, S. et al. (1997) Proliferation of adult T cell leukemia/lymphoma cells is associated with the constitutive activation of JAK/STAT proteins. Proc. Natl. Acad. Sci. U. S. A. 94, 13897–13902

27 Daenke, S. et al. (1990) Sequence variants of human T-cell lympho-tropic virus type I from patients with tropical spastic paraparesis andadult T-cell leukemia do not distinguish neurological from leukemicisolates. J. Virol. 64, 1278–1282

28 Kinoshita, T. et al. (1991) Sequence variations in LTR and env regionsof HTLV-I do not discriminate between the virus from patients withHTLV-I-associated myelopathy and adult T-cell leukemia. Int.J. Cancer 47, 491–495

29 Kannagi, M. et al. (1983) Establishment of human cytotoxic Tcell linesspecific for human adult T cell leukemia virus-bearing cells.J. Immunol. 130, 2942–2946

30 Kannagi, M. et al. (1984) Specific cytolysis of fresh tumor cells by anautologous killer T cell line derived from an adult T cell leukemia/lymphoma patient. J. Immunol. 133, 1037–1041

31 Jacobson, S. et al. (1990) Circulating CD8 þ cytotoxic T lymphocytesspecific for HTLV-I pX in patients with HTLV-I associated neurologicaldisease. Nature 348, 245–248

32 Parker, C.E. et al. (1992) Activated, HTLV-1-specific cytotoxicT-lymphocytes are found in healthy seropositives as well as in patientswith tropical spastic paraparesis. Virology 188, 628–636

33 Kannagi, M. et al. (1993) Expression of the target antigen for cytotoxicT lymphocytes on adult T-cell leukemia cells. Int. J. Cancer 54,582–588

34 Arnulf, B. et al. (2004) Loss of the ex vivo but not the reinducibleCD8 þ T-cell response to Tax in human T-cell leukemia virus type1-infected patients with adult T-cell leukemia/lymphoma. Leukemia18, 126–132

35 Lehky, T.J. et al. (1995) Detection of human T-lymphotropic virus typeI (HTLV-I) tax RNA in the central nervous system of HTLV-I-associated myelopathy/tropical spastic paraparesis patients by in situhybridization. Ann. Neurol. 37, 167–175

36 Levin, M.C. et al. (1997) Immunologic analysis of a spinal cord-biopsyspecimen from a patient with human T-cell lymphotropic virus typeI-associated neurologic disease. N. Engl. J. Med. 336, 839–845

37 Osame, M. et al. (1987) Chronic progressive myelopathy associatedwith elevated antibodies to human T-lymphotropic virus type I andadult T-cell leukemia-like cells. Ann. Neurol. 21, 117–122

38 Taylor, G.P. et al. (1999) Effect of lamivudine on human T-cell leukemiavirus type 1 (HTLV-1) DNA copy number, T-cell phenotype, and anti-tax cytotoxic T-cell frequency in patients with HTLV-1-associatedmyelopathy. J. Virol. 73, 10289–10295

39 Kannagi, M. et al. (1991) Predominant recognition of human T cellleukemia virus type I (HTLV-I) pX gene products by human CD8 þ

cytotoxic T cells directed against HTLV-I-infected cells. Int. Immunol.3, 761–767

40 Elovaara, I. et al. (1993) High human T cell lymphotropic virus type 1(HTLV-1)-specific precursor cytotoxic T lymphocyte frequencies inpatients with HTLV-1-associated neurological disease. J. Exp. Med.

177, 1567–157341 Pique, C. et al. (2000) Evidence for the chronic in vivo production of

human T cell leukemia virus type I Rof and Tof proteins from cytotoxicT lymphocytes directed against viral peptides. J. Exp. Med. 191,567–572

42 Niewiesk, S. et al. (1994) The transactivator gene of human T-cellleukemia virus type I is more variable within and between healthycarriers than patients with tropical spastic paraparesis. J. Virol. 68,6778–6781

43 Kato, H. et al. (1998) Oral administration of human T-cell leukemiavirus type 1 induces immune unresponsiveness with persistentinfection in adult rats. J. Virol. 72, 7289–7293

44 Hasegawa, A. et al. (2003) Expansion of human T-cell leukemia virustype 1 (HTLV-1) reservoir in orally infected rats: inverse correlation

Review TRENDS in Microbiology Vol.12 No.7 July 2004 351

www.sciencedirect.com

Page 7: Immunological risks of adult T-cell leukemia at primary HTLV-I infection

with HTLV-1-specific cellular immune response. J. Virol. 77,2956–2963

45 Hanabuchi, S. et al. (2001) Regression of human T-cell leukemia virustype I (HTLV-I)-associated lymphomas in a rat model: peptide-inducedT-cell immunity. J. Natl. Cancer Inst. 93, 1775–1783

46 Ohashi, T. et al. (2000) Prevention of adult T-cell leukemia-likelymphoproliferative disease in rats by adoptively transferred T cellsfrom a donor immunized with human T-cell leukemia virus type 1Tax-coding DNA vaccine. J. Virol. 74, 9610–9616

47 Harashima, N. et al. (2004) Graft-versus-Tax response in adult T-cellleukemia patients after hematopoietic stem cell transplantation.Cancer Res. 64, 391–399

48 Hisada, M. et al. (1998) Predictors of level of circulating abnormallymphocytes among human T-lymphotropic virus type I carriers inJapan. Int. J. Cancer 77, 188–192

49 Hisada, M. et al. (2001) Sex-specific mortality from adult T-cellleukemia among carriers of human T-lymphotropic virus type I. Int.J. Cancer 91, 497–499

50 Usuku, K. et al. (1988) HLA haplotype-linked high immune respon-siveness against HTLV-I in HTLV-I-associated myelopathy: compari-son with adult T-cell leukemia/lymphoma. Ann. Neurol. 23 (Suppl.),S143–S150

51 Yashiki, S. et al. (2001) HLA-A*26, HLA-B*4002, HLA-B*4006, andHLA-B*4801 alleles predispose to adult T cell leukemia: the limitedrecognition of HTLV type 1 tax peptide anchor motifs and epitopes togenerate anti-HTLV type 1 tax CD8(þ) cytotoxic T lymphocytes. AIDSRes. Hum. Retroviruses 17, 1047–1061

52 Ishiguro, N. et al. (1992) A rat model of human T lymphocyte virus typeI (HTLV-I) infection. 1. Humoral antibody response, provirusintegration, and HTLV-I-associated myelopathy/tropical spastic para-paresis-like myelopathy in seronegative HTLV-I carrier rats. J. Exp.Med. 176, 981–989

53 MacDonald, T.T. (1998) T cell immunity to oral allergens. Curr. Opin.Immunol. 10, 620–627

54 Weiner, H.L. (2001) Oral tolerance: immune mechanisms and thegeneration of Th3-type TGF-beta-secreting regulatory cells. MicrobesInfect. 3, 947–954

55 Hino, S. (2003) Primary prevention of adult T cell leukemia (ATL) inNagasaki, Japan by refraining from breast-feeding. In Gann mono-graph on Cancer Research (Vol. 50) (Sugamura, K. et al., eds),pp. 241–251, Japan Science Societies Press

56 Tateno, M. et al. (1984) Rat lymphoid cell lines with human T cellleukemia virus production. I. Biological and serological characteriz-ation. J. Exp. Med. 159, 1105–1116

57 Simpson, R.M. et al. (1996) Experimental acute adult T cell leukemia-lymphoma is associated with thymic atrophy in human Tcell leukemiavirus type I infection. Lab. Invest. 74, 696–710

58 Kondo, A. et al. (1993) A model of in vivo cell proliferation of adult T-cellleukemia. Blood 82, 2501–2509

59 Ohashi, T. et al. (1999) Induction of adult T-cell leukemia-likelymphoproliferative disease and its inhibition by adoptive immu-notherapy in T-cell-deficient nude rats inoculated with syngeneichuman T-cell leukemia virus type 1-immortalized cells. J. Virol. 73,6031–6040

60 Nomura, M. et al. Repression of Tax expression is associated both withresistance of human T-cell leukemia virus type-1-infected T cells toTax-specific cytotoxic T lymphocytes killing and with impairedtumorigenicity in a rat model. J. Virol. (in press)

61 Furuta, R.A. et al. (2002) Mouse model for the equilibration interactionbetween the host immune system and human T-cell leukemia virustype 1 gene expression. J. Virol. 76, 2703–2713

62 Kannagi, M. et al. (1992) Target epitope in the Tax protein of humanT-cell leukemia virus type I recognized by class I major histocompati-bility complex-restricted cytotoxic T cells. J. Virol. 66, 2928–2933

63 Kinoshita, K. et al. (1985) Oral infection of a common marmoset withhuman T-cell leukemia virus type-I (HTLV-I) by inoculating freshhuman milk of HTLV-I carrier mothers. Jpn. J. Cancer Res. 76,1147–1153

64 Nakano, S. et al. (1986) Primary infection of Japanese infants withadult T-cell leukaemia-associated retrovirus (ATLV): evidence for viraltransmission from mothers to children. J. Infect. 12, 205–212

65 Kusuhara, K. et al. (1987) Mother-to-child transmission of humanT-cell leukemia virus type I (HTLV-I): a fifteen-year follow-up studyin Okinawa, Japan. Int. J. Cancer 40, 755–757

66 Kubota, R. et al. (2000) HTLV-I specific IFN-gamma þ CD8 þ

lymphocytes correlate with the proviral load in peripheral blood ofinfected individuals. J. Neuroimmunol. 102, 208–215

67 Wodarz, D. et al. (2001) Cytotoxic T-cell abundance and virus load inhuman immunodeficiency virus type 1 and human T-cell leukaemiavirus type 1. Proc. R. Soc. Lond. B. Biol. Sci. 268, 1215–1221

68 Bangham, C.R. (2000) The immune response to HTLV-I. Curr. Opin.Immunol. 12, 397–402

Free journals for developing countries

The WHO and six medical journal publishers have launched the Access to Research Initiative, which enables nearly 70 of the world’s

poorest countries to gain free access to biomedical literature through the Internet.

The science publishers, Blackwell, Elsevier, the Harcourt Worldwide STM group, Wolters Kluwer International Health and Science,

Springer-Verlag and JohnWiley,were approachedby theWHOand theBritishMedical Journal in 2001. Initially,more than 1000 journals

will be available for free or at significantly reduced prices to universities,medical schools, research and public institutions in developing

countries. The second stage involves extending this initiative to institutions in other countries.

Gro HarlemBrundtland, director-general for theWHO, said that this initiativewas ’perhaps the biggest step ever taken towards reducing

the health information gap between rich and poor countries’.

See http://www.healthinternetwork.net for more information.

Review TRENDS in Microbiology Vol.12 No.7 July 2004352

www.sciencedirect.com