allogeneicmhcgenetransferenhancesaneffectiveantitumor...

12
Allogeneic MHC Gene Transfer Enhances an Effective Antitumor Immunity in the Early Period of Autologous Hematopoietic Stem Cell Transplantation Akihiko Kobayashi, 1,3 Hidehiko Hara, 1 Masaki Ohashi, 2 Takeshi Nishimoto, 1 KimikoYoshida, 1 Nobuhiro Ohkohchi, 3 TeruhikoYoshida, 2 and Kazunori Aoki 1 Abstract Purpose: In autologous hematopoietic stem cell transplantation (HSCT), lymphopenia-induced homeostatic proliferation of T cells is driven by the recognition of self-antigens, and there is an opportunity to skew theT-cell repertoire during theT-cell recovery by engaging tumor-associated antigens, leading to a break of tolerance against tumors. However, the homeostatic proliferation ^ driven antitumor responses seem to decline rapidly in association with tumor growth.We hypoth- esized that a tumor-specific immune response induced by an immune gene therapy could enhance and sustain homeostatic proliferation ^ induced antitumor immunity. Experimental Design: The antitumor effect of allogeneic MHC (alloMHC) gene transfer was examined at the early phase of the immune reconstitution after syngeneic HSCT. Results: Syngeneic HSCTshowed significant tumor growth inhibition of syngeneic colon cancer cells within a period of 30 days; however, the tumor then resumed rapid growth and the survival of the mice was not prolonged. In contrast, when the alloMHC plasmid was intratumorally injected at the early phase after syngeneic HSCT, the established tumors were markedly regressed and the survival of recipient mice was prolonged without significant toxicities, whereas no survival advantage was recognized in recipient mice injected with a control plasmid. This tumor suppression was evident even in the other tumors that were not injected with the alloMHC plas- mid. The antitumor response was characterized by the development of tumor-specificTcell ^ and natural killer cell ^ mediated cytotoxicities. Conclusion: The results suggest the efficacy and safety of integrating intratumoral alloMHC gene transfer with an autologous HSCT for the treatment of solid cancers. The central objective of cancer immunotherapy is to induce and sustain a tumor-specific immune response; that is, an in vivo generation of a large number of highly reactive antitumor lymphocytes that are not restrained by normal or cancer- induced tolerance mechanisms. One of the most promising approaches to achieve this objective is based on the recognition of tumor-associated antigens (TAA) by T cells (1). It is well known that lymphopenia is followed by spontane- ous expansion of the remaining T cells in the periphery to restore the original T-cell pool size and maintain homeostasis. Lymphopenia-induced homeostatic proliferation of T cells is driven by the recognition of self-MHC/peptide ligands in the absence of foreign antigens or inflammatory signals (2). Although TAAs are usually a poor target under lymphocyte- sufficient conditions because they are often self-antigens with low-affinity epitopes, lymphopenic conditions seem to be able to create an environment to mount an efficient antitumor immunity through the homeostatic proliferation – induced expansion of autoreactive T cells as shown in a variety of animal tumor models (3 – 7). However, several studies have also shown that the integration of other immunotherapeutic strategies might be necessary to successfully eradicate preexist- ing malignant tumors, because homeostatic proliferation – driven antitumor responses seem to rapidly decline in association with tumor growth (8). Intratumoral transfer of an allogeneic MHC (alloMHC) gene modifies tumor cells to express the alloMHC molecule, a highly immunogenic antigen that causes an allogeneic rejection response. In the process of this response, cytolytic T-lympho- cytes are generated not only against the modified tumor cells but also against unmodified tumor cells (9). A putative mechanism for the induction of specific tumor immunity is that the allogeneic response increases local production of Cancer Therapy: Preclinical Authors’ Affiliations: 1 Section for Studies on Host-Immune Response and 2 Genetics Division, National Cancer Center Research Institute, Tokyo, Japan; and 3 Department of Surgery, University of Tsukuba,Tsukuba, Japan Received 5/11/07; revised 9/3/07; accepted 9/18/07. Grant support: Third Term Comprehensive 10-Year Strategy for Cancer Control from the Ministry of Health, Labour and Welfare of Japan; grants-in-aid for Cancer Research from the Ministry of Health, Labour and Welfare of Japan. H. Hara, M. Ohashi, and T. Nishimoto received a Research Resident Fellowship from the Foundation for Promotion of Cancer Research. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Requests for reprints: Kazunori Aoki, Section for Studies on Host-Immune Response, National Cancer Center Research Institute, 5-1-1Tsukiji, Chuo-ku,Tokyo 104-0045, Japan. Phone: 81-3-3542-2511; Fax: 81-3-3541-2685; E-mail: kaoki@ gan2.res.ncc.go.jp. F 2007 American Association for Cancer Research. doi:10.1158/1078-0432.CCR-07-1163 www.aacrjournals.org Clin Cancer Res 2007;13(24) December 15, 2007 7469 Research. on July 3, 2018. © 2007 American Association for Cancer clincancerres.aacrjournals.org Downloaded from

Upload: nguyendat

Post on 30-May-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

Allogeneic MHCGeneTransfer Enhances an EffectiveAntitumorImmunity in the Early Period of Autologous HematopoieticStem CellTransplantationAkihiko Kobayashi,1,3 Hidehiko Hara,1Masaki Ohashi,2 Takeshi Nishimoto,1KimikoYoshida,1

Nobuhiro Ohkohchi,3 TeruhikoYoshida,2 and Kazunori Aoki1

Abstract Purpose: In autologous hematopoietic stem cell transplantation (HSCT), lymphopenia-inducedhomeostatic proliferation of Tcells is driven by the recognition of self-antigens, and there is anopportunity to skew theT-cell repertoire during theT-cell recovery by engaging tumor-associatedantigens, leading to a breakof tolerance against tumors. However, the homeostatic proliferation ^driven antitumor responses seem to decline rapidly in associationwith tumor growth.Wehypoth-esized that a tumor-specific immune response inducedbyan immune gene therapycould enhanceand sustain homeostatic proliferation ^ induced antitumor immunity.Experimental Design: The antitumor effect of allogeneic MHC (alloMHC) gene transfer wasexamined at the early phase of the immune reconstitution after syngeneic HSCT.Results: Syngeneic HSCTshowed significant tumor growth inhibitionof syngeneic colon cancercells within a period of 30 days; however, the tumor then resumed rapid growth and the survivalof the mice was not prolonged. In contrast, when the alloMHC plasmid was intratumorallyinjected at the early phase after syngeneic HSCT, the established tumors were markedlyregressed and the survival of recipient micewas prolongedwithout significant toxicities,whereasno survival advantagewas recognized in recipientmice injectedwith a control plasmid.This tumorsuppression was evident even in the other tumors that were not injected with the alloMHC plas-mid.The antitumor responsewas characterized by the development of tumor-specificTcell ^ andnatural killer cell ^ mediated cytotoxicities.Conclusion:The results suggest the efficacy and safety of integrating intratumoral alloMHCgenetransfer with an autologous HSCT for the treatment of solid cancers.

The central objective of cancer immunotherapy is to induce andsustain a tumor-specific immune response; that is, anin vivo generation of a large number of highly reactive antitumorlymphocytes that are not restrained by normal or cancer-induced tolerance mechanisms. One of the most promisingapproaches to achieve this objective is based on the recognitionof tumor-associated antigens (TAA) by T cells (1).

It is well known that lymphopenia is followed by spontane-ous expansion of the remaining T cells in the periphery torestore the original T-cell pool size and maintain homeostasis.Lymphopenia-induced homeostatic proliferation of T cells isdriven by the recognition of self-MHC/peptide ligands in theabsence of foreign antigens or inflammatory signals (2).Although TAAs are usually a poor target under lymphocyte-sufficient conditions because they are often self-antigens withlow-affinity epitopes, lymphopenic conditions seem to be ableto create an environment to mount an efficient antitumorimmunity through the homeostatic proliferation– inducedexpansion of autoreactive T cells as shown in a variety ofanimal tumor models (3–7). However, several studies havealso shown that the integration of other immunotherapeuticstrategies might be necessary to successfully eradicate preexist-ing malignant tumors, because homeostatic proliferation–driven antitumor responses seem to rapidly decline inassociation with tumor growth (8).

Intratumoral transfer of an allogeneic MHC (alloMHC) genemodifies tumor cells to express the alloMHC molecule, a highlyimmunogenic antigen that causes an allogeneic rejectionresponse. In the process of this response, cytolytic T-lympho-cytes are generated not only against the modified tumor cellsbut also against unmodified tumor cells (9). A putativemechanism for the induction of specific tumor immunity isthat the allogeneic response increases local production of

Cancer Therapy: Preclinical

Authors’ Affiliations: 1Section for Studies on Host-Immune Response and2Genetics Division, National Cancer Center Research Institute,Tokyo, Japan; and3Department of Surgery, University ofTsukuba,Tsukuba, JapanReceived 5/11/07; revised 9/3/07; accepted 9/18/07.Grant support: Third Term Comprehensive 10-Year Strategy for Cancer Controlfrom the Ministry of Health, Labour andWelfare of Japan; grants-in-aid for CancerResearch from the Ministry of Health, Labour andWelfare of Japan. H. Hara, M.Ohashi, and T. Nishimoto received a Research Resident Fellowship from theFoundation for Promotion of Cancer Research.The costs of publication of this article were defrayed in part by the payment of pagecharges.This article must therefore be hereby marked advertisement in accordancewith18 U.S.C. Section1734 solely to indicate this fact.Note: Supplementary data for this article are available at Clinical Cancer ResearchOnline (http://clincancerres.aacrjournals.org/).Requests for reprints: Kazunori Aoki, Section for Studies on Host-ImmuneResponse, National Cancer Center Research Institute, 5-1-1Tsukiji, Chuo-ku,Tokyo104-0045, Japan. Phone: 81-3-3542-2511; Fax: 81-3-3541-2685; E-mail: [email protected].

F2007 American Association for Cancer Research.doi:10.1158/1078-0432.CCR-07-1163

www.aacrjournals.org Clin Cancer Res 2007;13(24) December15, 20077469

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

cytokines, facilitates antigen presentation and T-cell accumula-tion, and consequently causes sensitization to previouslyunrecognized TAAs. The first critical step of this immune genetherapy is to induce a robust immune reaction at the tumorsites. Because f1% to 10% of T cells are considered to bealloreactive and the expression of alloMHC class I gene couldinduce strong allogeneic reactions in the injected tumor site,the transgene suits the aim of this therapy. Clinical trials withdirect intratumoral injection of the human leukocyte antigen-B7 gene-expressing plasmid DNA complexed with liposomerevealed an excellent safety profile and an objective systemicresponse in a substantial fraction (10-15%) of patients withmelanoma and head and neck cancer (10–14). The treatmentthus seemed to be promising, but at the same time it turned outto have some limitations of clinical efficacy (15).

Recently, we reported that the antitumor effect of intra-tumoral alloMHC gene transfer significantly augmented thegraft-versus-tumor effect of allogeneic hematopoietic stem celltransplantation (HSCT) without exacerbating graft-versus-hostdisease. However, the graft-versus-host disease still remained asa severe adverse effect in the treated mice (16). On the otherhand, autologous HSCT has several important advantages overallogeneic HSCT, such as safety and independence of donoravailability. In this study, we examined whether homeostaticproliferation–induced antitumor activity can be enhanced by astrong exposure of TAAs to donor T cells by alloMHC genetransfer during the physiologic immune reconstitution afterlymphocyte depletion. Furthermore, tumors often establish animmunotolerant environment with immunoinhibitory cyto-kines and regulatory T cells (17). The conditioning of HSCTwith irradiation and/or immunosuppressive reagents candestroy such immunotolerance mechanisms deployed by thetumor. The ‘‘resetting’’ and rapid expansion of a fresh and re-established immune system, in which tolerance to tumor cellsis not yet induced, may be able to augment the efficacy ofimmune therapies.

Materials andMethods

Animals and transplantation. Seven to 9-week-old female BALB/c(H-2d, Ly-1.2) mice were purchased from Charles River Japan, Inc., andwere housed under sterilized conditions. Animal studies were carriedout according to the Guideline for Animal Experiments of the NationalCancer Center Research Institute and approved by the InstitutionalCommittee for Ethics in Animal Experimentation. BALB/c mice receiveda lethal dose (9 Gy) of total body irradiation on the day oftransplantation. The irradiated mice were injected i.v. with 5 � 106 ofT cell–depleted bone marrow cells and 2 � 106 splenic T cells fromdonor BALB/c mice in a total volume of 0.2 mL Dulbecco’s PBSsolution (Nissui Pharmaceutical Co.). Bone marrow cells were isolatedfrom donors by flushing each femur and tibia with RPMI 1640 (NissuiPharmaceutical Co.) supplemented with 5% fetal bovine serum (ICNBiomedicals, Inc.), and splenic cells were prepared by macerating thespleens with a pair of tweezers. After lysis of the erythrocytes, the bonemarrow and splenic cells were incubated with anti–Thy-1.2 immuno-magnetic beads (Miltenyi Biotec) at 4jC for 15 min, followed bydepletion or selection of T cells by AutoMACS (Miltenyi Biotec),respectively. More than 90% of the T cells were depleted from the bonemarrow cells.

Tumor cell lines. CT26 and Renca cells are weakly immunogenicBALB/c-derived colon and renal cancer cell lines, respectively, and wereobtained from the American Type Culture Collection. Both cell lineswere confirmed to express MHC class I molecules (H-2Kd and H-2Dd)

abundantly by flow cytometry (data not shown). Cells were maintainedin RPMI containing 10% fetal bovine serum, 2 mmol/L L-glutamine,and 0.15% sodium bicarbonate (complete RPMI). A CT26 cell line thatstably expresses the H-2Kb gene was generated by retrovirus vector-mediated transduction and designated as CT26/H-2Kb (16).

In vivo alloMHC gene transfer and preimmunization. Tumor cellswere prepared in a total volume of 50 AL PBS and injected s.c. on the

right or left leg. A plasmid DNA expressing the H-2Kb gene under the

control of the Rous sarcoma virus long terminal repeat promoter wasused for intratumoral gene transfer. The same vector plasmid DNA

without a transgene (Empty) was used as a negative control. PlasmidDNA-liposome complex per mouse was prepared by the addition of 10

Ag plasmid DNA into a total of 25 AL PBS, followed by the addition of

25 AL of 0.15 mmol/L DMRIE/DOPE [(+/-)-N-(2-hydroxyethyl)-N ,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide/dioleoyl-

phosphatidylethanolamine], which was provided from Vical, Inc. Themixture solution was incubated at room temperature for 15 min, and

then injected directly into the tumor.

A preimmunization treatment with the plasmid DNA expressingH-2Kb gene to prime the allogeneic antigen-reactive cytolytic T-

lymphocyte was done either by i.p. injection of 1 � 106 of 30 Gy-

irradiated CT26/H-2Kb cells twice before tumor inoculation (at 14 and7 days), or by i.m. injection of 10 Ag of H-2Kb –expressing plasmid

DNA suspended with 50 AL of PBS twice before tumor inoculation (atdays 14 and 7 or at days 7 and 0) or after tumor inoculation (at days 0

and 7). Based on the previous report (9), preimmunization with CT26/

H-2Kb followed by treatment with H-2Kb DNA-liposome complex wasdone in some of the transplanted mice as a positive control to discover

the best possible therapeutic effect. The shortest (r) and longest (l)tumor diameters were measured at indicated days and the tumor

volume was determined as r2l/2. Data are presented as mean F SD.

Reverse transcription-PCR of H-2Kb gene in s.c. tumors. To examinethe in vivo expression of the H-2Kb transgene, total RNA was extracted

from s.c. CT26 tumors at 3, 7, 10, and 14 days after the injection of 10 Agof H-2Kb plasmid complexed with liposome. Reverse transcription-PCR

amplification was carried out using total RNA from the tumors in a

50 AL PCR mixture with the following primer set: H-2Kb upstream(5¶-ACTATTCAGGTGATCTCTCTGG-3¶) and downstream (5¶-TTTATC-

TTCAGGTCTGCTGTGATG-3¶) primers, and b-actin upstream (5¶-GATATCGCTGCGCTGGTCGT-3¶) and downstream (5¶-GGTCATCTTTT-

CACGGTTGG-3¶) primers. In total, 35 cycles (h-actin: 25 cycles) of the

PCR were carried out at 95jC for 1 min, 60jC for 1 min, and 72jC for2 min. The PCR products were fractionated on 2.0% agarose gel.

Cytotoxic assays. Splenocytes (5 � 106/mL) were cultured for 3 dayswith 30 Gy-irradiated CT26 stimulators (2.5 � 105/mL) in a completeRPMI containing 50 Amol/L 2-mercaptoethanol. The responder cellswere then collected and used as effector cells in a 4-h chromium releaseassay against indicated target cells. Concanavalin A (ConA) lympho-blasts were prepared by stimulating the splenocytes of naBve BALB/cmice for 3 days with 5 Ag/mL concanavalin-A at 2 � 106/mL in thecomplete RPMI containing 2-mercaptoethanol. Indicated target cellswere labeled by combining 5 � 106 cells with 50 ACi 51Cr (Perkin-Elmer Japan Co.) in a total volume of 0.2 mL complete RPMI for 1 h at37jC, followed by washing thrice with plain RPMI. For the chromiumrelease assay, 6 � 105 effector cells were mixed with 2 � 104 target cellsin a total volume of 0.2 mL complete RPMI in a 96-well round-bottomplate (BD Biosciences). To evaluate the relative contributions of naturalkiller (NK) cells, CD4+ and CD8+ T cells for the tumor cell lysis, effectorcells were incubated with monoclonal antibodies (mAb) against mouseCD4 (L3T4; BD PharMingen), CD8 (Ly-2; BD PharMingen), or NK(anti-asialo GM1, Wako Pure Chemical Industries, Ltd.) for 30 min at37jC before mixing with target cells. Supernatants were harvested withthe Skatron harvesting system (Skatron) and counted in a gammacounter (Packard Bioscience Company). Percentage of cytotoxicity wascalculated as [(experimental cpm - spontaneous cpm) / (maximumcpm - spontaneous cpm)] � 100. Spontaneous cpm was obtained fromtargets cultured in the medium alone, and maximum cpm was obtained

Cancer Therapy: Preclinical

www.aacrjournals.orgClin Cancer Res 2007;13(24) December15, 2007 7470

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

from targets incubated with 1% NP40. Each assay was done intriplicate.

ELISpot assays. IFN-g ELISpot kits (BD Biosciences) were usedaccording to the manufacturer’s instructions. Briefly, splenocytes (1 �105) and 30 Gy-irradiated CT26 (5 � 104) cells were cocultured in96-well plates precoated with mouse IFN-g (BD Biosciences) for20 h at 37jC in complete RPMI medium in triplicate. After aspiratingthe cell suspension and washing wells with deionized water,biotinylated anti-mouse IFN-g antibody (2 Ag/mL) was added andincubated for 2 h at room temperature. After extensive washing, astreptavidin–horseradish peroxidase solution was added and incu-bated for 1 h at room temperature. After washing, an aminoethylcarbozole substrate solution was added, and the plate was incubatedfor 15 min. Spots were counted under a stereomicroscope afterwashing the plate.

Cell surface marker and intracellular cytokine staining. Phycoery-rythrin-conjugated mAb to identify mouse IFN-g and FITC-conjugatedmAb to CD4, CD8, and CD49b were purchased from BD PharMingen.Splenocytes (1 � 106) were incubated with medium alone (control) orCT26 (1 � 105) cells for 10 h; brefeldin-A (10 Ag/mL) was then addedfor a 2-h incubation. After washing, cells were incubated with the CD4,CD8, or CD49b mAbs in a total volume of 100 AL PBS with 5% fetalbovine serum for 30 min at 4jC, and then fixed and permeabilized witha permeabilization buffer (BD Biosciences). Cells were finally stainedwith antibody to IFN-g for 15 min at room temperature, washed again,and analyzed by FACSCalibur (BD Biosciences). Irrelevant IgG mAbswere used as a negative control. Ten thousand live events were acquiredfor analysis.

In vivo depletion of T and NK cells. To deplete specific immuneeffector cell subsets before and during treatment with H-2Kb genetransfer, the transplanted mice received i.p. injections of 0.3 mg mAbfrom the anti-CD4+ hybridoma (clone GK1.5, rat IgG2b) or anti CD8+

hybridoma (clone Lyt-2.1, mouse IgG2b; ref. 18) or 0.5 mg of anti-asialo GM1 antibody (Wako Pure Chemical Industries, Ltd). Injectionsstarted on the inoculation day of CT26 cells, and the treatment repeatedevery 5 to 6 days throughout the entire experimental period to ensuredepletion of the target cell type. The f80% of CD4+, f60% of CD8+ Tcells, and f100% of NK cells were depleted in the antibody-treatedmice by flow cytometry (data not shown).

Immunohistochemistry. Immunostaining was done using the strep-tavidin-biotin-peroxidase complex techniques (Nichirei). Consecutivecryostat tissue sections (5 Am) were mounted on glass slides and fixedin cold acetone (-20jC) for 5 min. After blocking with normal ratserum, the sections were stained with rat anti-mouse CD4 and CD8antibodies (BD PharMingen). Parallel negative controls withoutprimary antibodies were examined in all cases. The sections werecounterstained with hematoxylin.

Statistical analysis. Using statistical software StatView for Windows(SAS Institute, Inc.), comparative analyses of the data were done by theStudent’s t test and survival was analyzed using the Kaplan-Meiermethod with the log-rank test. P < 0.05 was considered as a significantdifference.

Results

Growth suppression of colon cancer in lymphopenic hosts. Wefirst examined whether homeostatic proliferation of T cellscould induce antitumor immunity in lymphopenic hosts. Onthe day of lethal (9 Gy) irradiation, BALB/c mice were injecteds.c. with CT26 colon cancer cells shortly after the irradiation,and then bone marrow and T cells were infused. The growth oftumors was significantly suppressed in the syngeneic HSCTrecipients (Fig. 1A). Although the lymphocyte infusion intononirradiated naBve mice showed some antitumor effect at day14 after the tumor injection, the tumors rapidly grew andcaught up with the tumor growth in the control mice at day 24

(Fig. 1A). Sublethal irradiation (6.5 Gy) is a common means ofinducing lymphopenia in mice. The infusion of T cells withoutmarrow cells suppressed s.c. tumor growth in sublethallyirradiated mice also (Fig. 1A), suggesting that T cells accompa-nying the graft could participate in antitumor immunity inlymphopenic recipients.

In the clinical autologous HSCT, the grafts were preparedfrom tumor-bearing patients. The influence of the tumor-bearing condition of the donor on the antitumor effect of thesyngeneic HSCT was examined by using the BALB/c donor micethat had been injected with CT26 cells 4 weeks before theHSCT. The growth of tumors was substantially suppressed inthe mice that received HSCT from a tumor-bearing donorcompared with a naBve donor (Fig. 1B), suggesting that somefraction of donor lymphocytes from tumor-bearing mice areprimed in response to TAAs and remain responsive upontransplantation.

AlloMHC gene transfer induces synergistic antitumor effect atearly phase after syngeneic HSCT. We sought to determine thetime point after HSCT when alloMHC gene transfer couldenhance the antitumor effects in syngeneic HSCT recipients.When we assessed the posttransplant immune reconstitution ofT cells in the spleen, the CD4+ T-cell number returned to anormal level by the 8th week, whereas the CD8+ cell countswere increasing but remained below normal at 8 weeks(Supplementary Fig. S1). To compare the antitumor responseby alloMHC gene transfer at various time points after thesyngeneic HSCT, transplantations were staggered at weeklyintervals, followed by the s.c. injection of 1 � 106 CT26 cells.Then, for each experiment, the H-2Kb gene–expressing plasmidDNA complexed with liposome was injected thrice into thetumor at 5, 7, and 9 days after the tumor inoculation. Thetumor volumes at day 5 were 60 to 100 mm3. A single injectionof plasmid DNA–liposome complexes showed f1% genetransduction efficiency in vivo (data not shown). The H-2Kb

mRNA was detected up to 10 days after the intratumoralinjection of H-2Kb plasmid/liposome complex by reversetranscription-PCR (Supplementary Fig. S2). The control emptyvector was not immunogenic in vivo because the tumor volumeof the negative control group without any intratumoralinjections was similar to that of the empty vector–injectedgroup (data not shown). AlloMHC gene transfer at 8 weeksafter transplantation did not enhance the antitumor immunity,whereas a substantial antitumor effect was observed by H-2Kb

gene transfer at early phases (3-5 weeks) after the transplanta-tion (Fig. 2A). These results suggested that intratumoralalloMHC gene transfer during immune reconstitution caninduce a synergistic antitumor effect.

Next, we examined whether a preimmunization with i.m.injection of H-2Kb plasmid could enhance the antitumoreffect of intratumoral H-2Kb gene transfer. In this experiment,an 8-week gene transfer model was used, because the strongantitumor effect of 3 to 5-week gene transfers would makethe effect of preimmunization unclear. Preimmunization withirradiated CT26/H-2Kb cells was also carried out as a positivecontrol, because it was expected to represent the maximumpossible effect of the H-2Kb immune gene therapy (9). Thepreimmunization with the H-2Kb plasmid DNA induced aconsiderable suppression of the tumor growth (Fig. 2B),suggesting that the priming of immune cells with analloMHC molecule augments the allogeneic rejection reaction

AllogeneicMHCGeneTransfer and Autologous HSCT

www.aacrjournals.org Clin Cancer Res 2007;13(24) December15, 20077471

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

in gene-transferred tumors, leading to a strong antitumoreffect. Because the i.m. injection of plasmid DNA istechnically much more feasible compared with the makingof alloMHC-expressing autologous tumor cells for eachpatient, this finding has important implications for theapplication of combining i.m. preimmunization and intra-tumoral gene transfer in the clinical setting.

AlloMHC gene transfer prolongs the survival of syngeneicHSCT recipients. We next examined the combined antitumoractivity of intratumoral alloMHC gene transfer with i.m.preimmunization at an early time point of immune reconsti-tution in syngeneic HSCT recipients from a tumor-bearingdonor. To avoid the influence of irradiation on tumor cells,recipient mice were inoculated s.c. with the CT26 cells shortlyafter lethal irradiation, followed by the infusion of thesyngeneic bone marrow and T cells. The H-2Kb plasmid wasi.m. injected on the left leg twice and then the H-2Kb plasmid-liposome complex was thrice injected into the tumor on theright leg (Fig. 3A). Although significant tumor suppression wasobserved up to 30 days after the tumor inoculation in thesyngeneic HSCT recipients injected with control plasmid-liposome (Fig. 3A), mouse survival was not extended due tothe subsequent rapid tumor growth (Fig. 3B). The tumorsmight establish the effective immunotolerant environmentagainst the reconstituted immune system in the absence ofthe alloMHC transfer. In contrast, intratumoral H-2Kb gene

transfer with preimmunization enhanced the antitumor effectsin syngeneic HSCT recipients and the s.c. tumors disappearedin f50% of the mice, leading to a prolongation of survival(Fig. 3A and B) without major side effects (data not shown).The disappearance of the tumors and the strong tumorsuppression due to the sustainable antitumor immune reactionafter the alloMHC gene transfer may be important factors toprolong mice survival. The antitumor activity of combinationtherapy with gene transfer and preimmunization was alsoevident against syngeneic Renca renal cancer cells in thesyngeneic recipient mice (Fig. 3C). Although we did not followthe treated mice until their final survival days, the three of threeempty plasmid–injected mice showed steady tumor growthand one mouse already died for the tumor by day 60, whereasthe s.c. tumors in two of four H-2Kb plasmid–injected micedisappeared up to the last observation day (day 96), at whichpoint all the mice of the group were alive, suggesting improvedsurvival (Fig. 3C, right).

The tumor growth suppression experiment was repeated byinoculating the CT26 cells 1 day before irradiation as apreexisting tumor model. Although the tumor growth mightbe retarded by irradiation per se, the combination therapy withgene transfer and preimmunization induced a strong suppres-sion of the growth of the CT26 tumor (Fig. 3D).

Expansion of tumor-specific T cells after intratumoral alloMHCgene transfer in syngeneic HSCT recipients. To examine the

Fig.1. Syngeneic HSCTof bonemarrow andTcells significantly suppresses s.c. tumor growth in lethally irradiatedmice. A, growth suppression of s.c. tumor in the syngeneictransplant mice.The mice were irradiated with 9 Gy (lethal) or 6.5 Gy (sublethal) and received bone marrow andTcells (9 Gy!HSCT) orTcells (6.5 Gy!T), respectively.As a control, nonirradiated mice were untreated (control) or receivedT lymphocyte infusion (LI ; n = 4-6). B, the antitumor effect in syngeneic HSCTrecipients fromtumor-bearing mice.The mice were lethally irradiated and received bone marrow transplantation andTcells from tumor-bearing mice or naI« ve mice (n = 4-8).

Cancer Therapy: Preclinical

www.aacrjournals.orgClin Cancer Res 2007;13(24) December15, 2007 7472

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

immune reaction to the alloMHC gene transfer in syngeneicHSCT recipients, the frequency of tumor-reactive T cells wasdetermined by intracellular cytokine staining and flow cytom-etry on splenocytes. In nontransplanted mice that receivedthe control vector, only 0.16% of splenocytes produced IFN-gin response to the stimulation of CT26 cells. The IFN-g–producing cells were 0.56% of the splenocytes in syngeneicHSCT recipients treated with control plasmid, and theyincreased to 1.3% in the syngeneic recipients injected withH-2Kb vector (Fig. 4A). The percentages of CD4+ and CD8+

T cells stimulated to produce IFN-g in response to CT26 cellswere also significantly higher in the recipient mice injected withthe H-2Kb vector, and there was also an increase in thepercentage of the IFN-g–producing NK cells (Fig. 4A).

The ELISpot assay also showed that average numbers of IFN-g–secreting spots were clearly increased in response to CT26stimulation but not to lymphocytes in syngeneic HSCTrecipients, and alloMHC gene transfer further increased thespot numbers for CT26 cells (Fig. 4B).

An in vitro cytotoxic assay was done to examine antitumorcytotoxic immune responses induced by alloMHC gene transferin syngeneic HSCT recipients. The splenocytes derived from thetransplanted mice recognized and lysed CT26 cells but notConA blasts, and H-2Kb gene transfer significantly enhancedthe cytolysis to CT26 cells (Fig. 4C, left) but not to ConA blasts(Fig. 4C, right). These results suggest that the alloMHC gene

transfer–mediated exposure and recognition of TAAs inducedexpansion of the tumor antigen–specific T cells in a lympho-penic host.

CD4+ T cells, CD8+ T cells, and NK cells contribute toantitumor immunity. In the in vitro blocking assays oflymphocyte cytotoxicity with anti-murine CD4, CD8, andanti-asialo GM1 antibodies, all of the CD4+, CD8+ T cells, andNK cells were shown to contribute to the tumor cell lysis insyngeneic HSCT recipients treated with H-2Kb gene transfer(Fig. 5A). The flow cytometry detected no MHC class IIexpression on CT26 cells (data not shown). Several murinebone marrow transplantation models have shown that CD4+

T cells as well as CD8+ T cells are major effectors to induce thegraft-versus-host disease and also graft versus tumor effectsagainst MHC II–negative tumor cells, and the inflammatorycytokines such as tumor necrosis factor-a, IFN-g, andinterleukin-1 produced by the T cells might contribute to theCD4-mediated killing effect in the MHC class II– independentmanner (16, 19, 20). To further explore the role of immunecells in antitumor immunity in vivo, the mice were adminis-tered with anti-CD4, anti-CD8, and anti-asialo GM1 anti-bodies to deplete these cell populations. The antitumor effectwas completely canceled in the mice with depletion of all ofthe CD4+ and CD8+ T cells and NK cells. Depletion of CD4+

T cells showed some growth advantages but still resulted insignificant tumor growth inhibition, whereas the antitumor

Fig. 2. AlloMHC gene transfer during immune reconstitution enhances antitumor effect in syngeneic transplant mice. A, AlloMHC gene transfer at early stage of immunereconstitution enhances antitumor effects. Syngeneic HSCTs to the BALB/c mice were staggered at weekly intervals, followed by a s.c. injection of1 �106 CT26 cells andintratumoral H-2Kb gene transfer at days 5, 7, and 9 (n = 6-8). B, preimmunization with alloMHC plasmid enhances the antitumor activity of intratumoral alloMHC genetransfer.The syngeneic HSCTrecipient mice were given an i.m. injection ofH-2Kb plasmid DNA as a preimmunization in the 8-wk gene transfer model, and then CT26 tumorswere injected with H-2Kb vector (n = 9).Tumor volumes were compared between groups at 28 d after the tumor inoculation.

AllogeneicMHCGeneTransfer and Autologous HSCT

www.aacrjournals.org Clin Cancer Res 2007;13(24) December15, 20077473

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

activities were inhibited in animals depleted of either CD8+

T cells or NK cells (Fig. 5B), suggesting that CD4+ T cells havea certain antitumor activity in vivo, but CD8 + cytotoxic T cellsand NK cells play a major role in the in vivo antitumorimmunity. In addition, the depletion of regulatory T cells(21–23) and making space for proliferation of CD8+ T cellsmay explain the difference in the antitumor effect of CD4+

T-cell depletion in vitro and in vivo .

Immunohistochemical staining showed that CD4+ and CD8+

T cells infiltrated into the CT26 s.c. tumors in syngeneic HSCTrecipients, and that the H-2Kb gene transfer increased the num-ber of these cells within the tumor (Supplementary Fig. S3). Theaverage counts of CD4+ and CD8+ T cells increased at 1 weekafter the H-2Kb gene transfer on the HSCT recipient mice, and asignificant number of cells were still detected until at least 3weeks after the gene transfer. On the other hand, few of these

Fig. 3. AlloMHC gene transfer during immune reconstitution prolongs the survival of transplant mice. A, preimmunization followed by intratumoral alloMHC gene transferstrongly suppresses tumor growth in syngeneic transplant mice. After the intratumoral injection ofH-2Kb or empty vector, tumor volume in themicewasmeasured (n = 6-12).B, AlloMHC gene transfer significantly extends the survival of syngeneic transplant mice. After intratumoral injection ofH-2Kb vector, survival of the mice was observed(n = 5-12). C, antitumor activity of alloMHC gene transfer with preimmunization against murine renal cancer cells.The 5 � 106 Renca cells were s.c. inoculated in syngeneictransplant mice. Left, 17 d after the intratumoral H-2Kb gene transfer (n = 4), the tumor volumes were compared with control plasmid injection in the syngeneic HSCT mice(n = 3). Right, tumor volume in each mouse was plotted on the days indicated. Pre, preimmunization. D, tumor growth suppression by alloMHC gene transfer in tumorpreexisting model.The1�106 CT26 cells were s.c. inoculated1d before transplantation. After intratumoralH-2Kb gene transfer, tumor volumes were compared with controlplasmid injection (n = 4-6).

Cancer Therapy: Preclinical

www.aacrjournals.orgClin Cancer Res 2007;13(24) December15, 2007 7474

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

cells were detected in tumors injected with the control plasmid at3 weeks after the injection (Fig. 5C). Although the H-2Kb mRNAwas not detected at day 14 after the intratumoral injection ofH-2Kb plasmid (Supplementary Fig. S2), the significant numbersof CD4+ and CD8+ T cells were detected in the tumors until atleast 3 weeks after the gene transfer (Fig. 5C), suggesting that along-term expression of the transgene does not seem to be nec-essary to induce a sustainable activation of antitumor immunity.

AlloMHC gene transfer during immune reconstitution causesgrowth suppression of both local and distant tumors. To evaluatethe therapeutic efficacy of alloMHC gene transfer for tumors at adistant site, CT26 cells were s.c. inoculated on the left leg of

syngeneic HSCT recipients 3 days after the injection of H-2Kb

vector into the tumors on the right leg. The H-2Kb gene transfersignificantly suppressed the growth of the newly inoculated,untreated tumor on the left leg, as well as the vector-injectedtumor on the right leg (Fig. 6A). The infiltration of CD4+ andCD8+ T cells was examined in the untreated tumors at theopposite site by immunohistochemical staining. Although theCD4+ and CD8+ cells infiltrated significantly into the untreatedtumors 16 days after the tumor inoculation in the HSCTrecipients with H-2Kb plasmid injection (Fig. 6B), the averagenumber of CD8+ T cells were lower in the untreated tumors thanthe H-2Kb vector–injected tumors (Fig. 5C). The intratumoral

Fig. 4. A larger number of IFN-g^producing cells are induced byH-2Kb genetransfer in the syngeneic transplant mice.A, intracellular cytokine staining of IFN-g^producing cells in response to stimulation ofCT26 cells.The splenocytes (1 �106) fromtreated mice were incubated with CT26(1�105) and stained by phycoerythrinanti-mouse IFN-g and counted (Overall).The activated cell fractions were analyzedby staining with FITC ^ anti-mouse CD4,CD8, CD49b (NK) (n = 1-4). B, ELISpotassay of IFN-g^ producing cells in responseto stimulation of CT26 cells. Splenocytes(1�105) from treated or control mice werecocultured with CT26 (5 � 104) or controllymphocytes and stained with biotinylatedanti-mouse IFN-g antibody to detectcaptured IFN-g (n = 3). C, in vitrocytotoxicity assay of transplant mice withalloMHC gene transfer. Splenocyteswere collected from each treatmentgroup15 d after H-2Kb gene transfer,and their cytotoxicity was evaluatedin a standard 4-h 51Cr release assay(effector/target ratio = 30) against CT26cells (left) and BALB/c-derived ConAlymphoblasts (right) after stimulationwith irradiated CT26 cells (n = 3).The CT26killing at various effector to target (E/T)ratio in two groups (H-2Kb plasmidEinjection with preimmunization orempty plasmidEinjection withoutpreimmunization) of HSCTrecipientswere also presented.

AllogeneicMHCGeneTransfer and Autologous HSCT

www.aacrjournals.org Clin Cancer Res 2007;13(24) December15, 20077475

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

injection of H-2Kb plasmid may induce the infiltration of bothalloreactive and tumor-reactive CD8+ T cells into the tumors,whereas the tumor-reactive T cells might mainly infiltrate intothe untreated tumors at the opposite site.

As another model of distant metastasis, CT26 cells wereinoculated into the peritoneal cavity 17 days after the H-2Kb

gene transfer into the s.c. tumors. The injection of H-2Kb vectorsignificantly improved the survival of the treated mice, whereasall the control vector–injected mice died by the 95th day afterthe i.p. injection of the tumor cells (Fig. 6C). All of the dead

animals were confirmed to have disseminated tumors in theperitoneal cavity. These data showed that an intratumoralalloMHC gene transfer can induce a systemic antitumorimmunity in the syngeneic HSCT recipients.

Tumor immunity induced by alloMHC gene transfer is long-lasting in syngeneic HSCT recipients. The CT26 s.c. tumorsdisappeared in 40% to 60% of the transplanted mice by theH-2Kb gene transfer. To examine the long-term duration of thetumor-specific immunity in vivo , a total of four mice thatsurvived the initial CT26 injection with complete tumor

Fig. 5. Antitumor immunity of alloMHC gene transfer after depletion of immune cells. A, blocking assay of in vitro cytotoxicity. Splenocytes from transplant mice treatedwithH-2Kb gene transfer were analyzed for their cytotoxicity in a 51Cr release assay (effector/target ratio, ref. 30) against CT26 cells with anti-CD4 or anti-CD8 or anti-asialoGM1antibodies. B, antitumor effect of H-2Kb gene transfer after in vivo depletion of CD4+, CD8+ Tcells, and NK cells. A group of transplant mice were treated withanti-CD4, anti-CD8, and anti ^ asialo GM1antibodies to deplete these cell populations, and the CT26 tumors were injected with H-2Kb vector (n = 7-9). Empty, miceinjected with the empty plasmid vector. Control, mice received no HSCTor gene transfer.Tumor volumes10 d after theH-2Kb gene transfer were presented. C, count ofCD4+ and CD8+ Tcells in CT26 tumors. AfterH-2Kb gene transfer, s.c. tumors were processed for immunohistochemistry stainedwith anti-murine CD4 and CD8 antibodies.CD4+ and CD8+ cells were counted under the microscope in five high-power fields (�400) until 3 wk after the H-2Kb gene transfer. Points, mean; bars, SD.Theimmunohistochemistry data in the group with preimmunization followed by intratumoral alloMHC gene transfer was not available at day 32 because the tumor volumeswere too small to process the staining.

Cancer Therapy: Preclinical

www.aacrjournals.orgClin Cancer Res 2007;13(24) December15, 2007 7476

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

regression were again inoculated with CT26 cells on the rightleg and Renca cells on the left leg. At the time of the secondtumor challenge, at least 8 weeks passed after the regression ofthe first tumor. In all the mice, Renca cells formed a tumormass, whereas the CT26 cells were rejected (SupplementaryFig. S4), suggesting that TAAs are different between CT26 andRenca cells and that a H-2Kb gene transfer to CT26 tumorincreases the recognition of CT26-specific antigens by immune

cells. The results showed that the tumor-specific immunityinduced by alloMHC gene transfer is potentially long lasting inthe syngeneic HSCT recipients.

Discussion

Low-affinity self-antigens do not induce T-cell proliferationin the periphery of normal mice, but the same antigens

Fig. 6. Suppressionof tumors at distant sites by intratumoral alloMHCgene transfer.A, suppressionof contralateral tumors.Three days after injectionofH-2Kb vector into s.c.tumor on the right leg of theHSCTrecipientmice, CT26 cells were s.c. inoculatedon the left leg (n = 4-5).B, count of CD4+ andCD8+ Tcells innontreated CT26 tumors at theopposite site by the immunohistochemical staining. At 18 d afterH-2Kb gene transfer, s.c. tumors were processed for immunohistochemistry. CD4+ and CD8+ cells werecounted under the microscope in five high-power fields (�400). Columns, mean; bars, SD. C, suppression of peritoneal dissemination. Seventeen days after injection of theH-2Kb vector into s.c. tumor on the HSCTrecipient mice, 1 �106 CT26 cells were i.p. injected, and survival of the treated mice was evaluated (n = 9-10).

AllogeneicMHCGeneTransfer and Autologous HSCT

www.aacrjournals.org Clin Cancer Res 2007;13(24) December15, 20077477

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

induce antigen-specific T-cell proliferation during immunereconstitution from lymphopenia, because the signal strengthneeded to trigger proliferation and activation of T cells isconsiderably reduced in a lymphopenia condition (8). Thisprocess could be used to break tolerance against tumorantigens, and indeed several studies in mouse models showedthat the presence of tumor antigens during homeostatic T-cellproliferation leads to effective antitumor autoimmunity withspecificity and memory (3, 24). In this study also, lympho-penia-driven T-cell proliferation significantly suppressed thegrowth of s.c. tumors. Baccala et al. hypothesized that animmunologically delicate environment decreases the activa-tion threshold of tumor-specific T cells by TAA-presentingdendritic cells in the draining lymph nodes, leading to theirpreferential engagement and expansion (25). In clinicalpractice, until recently, the aim of the autologous HSCT forsolid cancer has been to rescue the pancytopenia after high-dose chemotherapy (26–28). Although the treatment itself iswell-established and can be done safely with a mortality rateof <5%, it is by no means satisfactory in improving patientsurvival (27, 29). One of the reasons for this regrettableoutcome might be that the authors did not transfer T cellsconcomitantly with HSCs (28, 30). In addition, the antitumorimmune reactions of homeostatic lymphocyte proliferationdid not come to the front because focus was only on thetherapeutic effects of chemotherapy.

Despite significant tumor suppression in syngeneic trans-plant recipients during the early posttransplant period, thisresponse was not sustained in mice without gene transfer,because the tumor rapidly regained its growth (Fig. 3A andB). Borrello et al. (5) reported similar results: homeostaticproliferation–induced massive expansion and activation oftumor-specific T cells rapidly declines in association withtumor progression. The main reason for failure to fullyeliminate the tumor might be T-cell tolerance mechanismssuch as anergy and exhaustion, and this might be influencedby the relative quantity of antigens encountered (5, 31). Toavoid a tolerogenic reaction, attempts at sustaining the hostresponse through posttransplant immunization should bemade before the establishment of tumor-specific T-celltolerance; that is, long before full recovery of the hostimmune system (3, 5). Our data showed that the combina-tion with alloMHC gene transfer significantly enhanced theantitumor immune response and prolonged the survival oftreated mice. The alloMHC gene transfer can induce localallogeneic reactions, which consist of trafficking immunecells into a tumor, the presentation of tumor antigenicpeptides on antigen-presenting cells, and the local produc-tion of various cytokines (9). These processes could greatlyaugment the exposure of TAAs to T cells during immunereconstitution in a lymphopenic host, leading to a signifi-cant homeostatic expansion of such tumor reactive T cells.In addition, the repeated immunization may serve tomaintain the increased frequency of precursor cells and theexpanded population and activation state of tumor-specificT cells (5).

There have been several animal studies showing thepotential efficacy of gene- and cell-based immunotherapy insyngeneic transplant mice. A strong antitumor effect wasobserved for the mice vaccinated in the post–bone marrowtransplantation period with autologous tumor cells that were

retrovirally transduced with the granulocyte-macrophage colonystimulating factor gene (5). Another example of the combina-tion of the immune therapy and HSCT was an immunizationwith dendritic cells pulsed with whole tumor cell lysates in theearly phase of bone marrow transplantation, which led toefficient antitumor responses in a mouse breast tumor model(32). Adoptive transfer of tumor-specific T cells has alsoshown enhanced antitumor immune responses in lympho-penic mice compared with T-cell replete mice (8), andrecently, Morgan et al. (33) reported the efficacy of a strategycomposed of immunodepletion and adaptive cell transfer forpatients with metastatic melanoma. However, because tumor-reactive T cells are mostly polyclonal and heterogeneousexpressions of various TAAs coexist even in a tumor mass(34), the in vivo stimulation of multiple tumor-reactivelymphocytes might be of profound importance to the clinic(16). Compared with these approaches, one of the majoradvantages of the in vivo alloMHC gene transfer is that it doesnot need to manipulate and culture the immune and tumorcells ex vivo, making this strategy more feasible for manypatients with solid cancers.

In some of the experiments in this study, the CT26 tumorcells were inoculated before the transfusion of bone marrowand T cells, considering the actual clinical situations, in whichthe recipients usually harbor relapse or residual cancer cells atthe time of HSCT. This is an important point to consider in theexperimental design, but most of the previous immunothera-peutic studies used a treatment model, in which tumor cells areinoculated after bone marrow transplantation. Our result thatalloMHC gene transfer could significantly suppress the growthof preexisting tumor cells suggests that an alloMHC genetransfer with autologous HSCT is a promising therapeuticstrategy in a clinical setting. As a next step, a combination withother approaches such as immune stimulatory cytokine mightbe examined for whether they can further enhance theantitumor effects of alloMHC gene transfer in autologousHSCT recipients.

Recently, it has been reported that the extent of lympho-depletion by conditioning influences the homeostatic prolif-eration–driven antitumor immunity (8, 35). Wrzesinski et al.(8) emphasized the potential of complete immunoablationfollowed by autologous HSCT in treating solid cancer,because after the consumptive cellular cytokine ‘‘sinks’’ areremoved, adoptively transferred T cells are able to exist in aless competitive environment and to access the cytokines.Our result that lethal irradiation seems to be more effectivein tumor suppression than sublethal irradiation (Fig. 1A)might suggest the usefulness of complete lymphodepletion.On the other hand, high-intensity myeloablation can beassociated with multiple toxicities, including mucositis, graftfailure, engraftment syndrome, prolonged neutropenia-asso-ciated risk of infection, and early and late adverse effects ofirradiation (27, 28, 36). Although there was little radiation-related mortality during the observation period in this study,a safer lymphodepleting regimen for future trials should bedesigned.

In conclusion, the combination of intratumoral alloMHCgene transfer with the autologous HSCT in the period ofimmune reconstitution showed significant enhancement inthe cytotoxicity, tumor-specific release of cytokines, and aninduction of long-lasting systemic tumor immunity without

Cancer Therapy: Preclinical

www.aacrjournals.orgClin Cancer Res 2007;13(24) December15, 2007 7478

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

major toxicity. The combination therapy–mediated completeeradication of some tumors and the overall survival of theanimals was prolonged. The therapeutic strategy deserves anevaluation in future clinical trials for solid cancers.

Acknowledgments

We thank Miwa Kushida for her technical support and Vical Incorporated forproviding the DMRIE/DOPE liposome.

AllogeneicMHCGeneTransfer and Autologous HSCT

www.aacrjournals.org Clin Cancer Res 2007;13(24) December15, 20077479

References1. Nagorsen D, Scheibenbogen C, Marincola FM,Letsch A, Keilholz U. Natural T cell immunity againstcancer. Clin Cancer Res 2003;9:4296^303.

2. Surh CD, Sprent J. Homeostatic T cell proliferation:how far canTcells be activated to self-ligands? JExpMed 2000;192:F9^14.

3. Hu HM, Poehlein CH, UrbaWJ, Fox BA. Develop-ment of antitumor immune responses in reconstitutedlymphopenic hosts. Cancer Res 2002;62:3914^9.

4. DummerW, Niethammer AG, Baccala R, et al. T cellhomeostatic proliferation elicits effective antitumorautoimmunity. JClin Invest 2002;110:185^92.

5. Borrello I, Sotomayor EM, Rattis FM, Cooke SK,Gu L, Levitsky HI. Sustaining the graft-versus-tumoreffect through posttransplant immunization withgranulocyte-macrophage colony-stimulating factor(GM-CSF)-producing tumor vaccines. Blood 2000;95:3011^9.

6. MaJ, UrbaWJ, Si L,WangY, Fox BA, Hu HM. Anti-tumorTcell response and protective immunity in micethat received sublethal irradiation and immune recon-stitution. EurJ Immunol 2003;33:2123^32.

7. Mackall CL, Bare CV, Granger LA, Sharrow SO,Titus JA, Gress RE. Thymic-independent Tcell regen-eration occurs via antigen-driven expansion of pe-ripheral T cells resulting in a repertoire that is limitedin diversity and prone to skewing. J Immunol 1996;156:4609^16.

8.Wrzesinski C, Restifo NP. Less is more: lymphodeple-tion followed by hematopoietic stem cell transplantaugments adoptiveT-cell-based anti-tumor immuno-therapy. Curr Opin Immunol 2005;17:195^201.

9. Plautz GE,Yang ZY,Wu BY, Gao X, Huang L, NabelGJ. Immunotherapy of malignancy by in vivo genetransfer into tumors. Proc Natl Acad Sci U S A1993;90:4645^9.

10. Nabel GJ, Nabel EG, Yang ZY, et al. Direct genetransfer with DNA-liposome complexes in melano-ma: expression, biologic activity, and lack of toxicityin humans. Proc Natl Acad Sci U S A 1993;90:11307^11.

11. Rini BI, Selk LM,Vogelzang NJ. Phase I study ofdirect intralesional gene transfer of HLA-B7 intometa-static renal carcinoma lesions. Clin Cancer Res 1999;5:2766^72.

12. Gleich LL, GluckmanJL, Armstrong S, et al. Alloan-tigen gene therapy for squamous cell carcinoma of thehead and neck: results of a phase-1trial. Arch Otolar-yngol Head Neck Surg1998;124:1097^104.

13. Gonzalez R, Hutchins L, Nemunaitis J, Atkins M,Schwarzenberger PO. Phase 2 trial of Allovectin-7 in

advanced metastatic melanoma. Melanoma Res2006;16:521^6.

14. Stopeck AT, Jones A, Hersh EM, et al. Phase II studyof direct intralesional gene transfer of allovectin-7, anHLA-B7/h2-microglobulin DNA-liposome complex,in patients with metastatic melanoma. Clin CancerRes 2001;7:2285^91.

15. OverwijkWW,Theoret MR, Finkelstein SE, et al. Tu-mor regression and autoimmunity after reversal of afunctionally tolerant state of self-reactive CD8+ T cells.JExp Med 2003;198:569^80.

16. Ohashi M, Kobayashi A, Hara H, et al. AllogeneicMHC gene transfer enhances antitumor activity ofallogeneic hematopoietic stem cell transplantationwithout exacerbating graft-versus-host disease. ClinCancer Res 2006;12:2208^15.

17. Khong HT, Restifo NP. Natural selection of tumorvariants in the generation of ‘‘tumor escape’’ pheno-types. Nat Immunol 2002;3:999^1005.

18.Nakayama E, Uenaka A. Effect of in vivo administra-tion of Lyt antibodies. Lyt phenotype ofTcells in lym-phoid tissues and blocking of tumor rejection. J ExpMed1985;161:345^55.

19.TeshimaT, Hill GR, Pan L, et al. IL-11separates graft-versus-leukemia effects from graft-versus-host dis-ease after bone marrow transplantation. J Clin Invest1999;104:317^25.

20. Homma S, Komita H, Sagawa Y, OhnoT, Toda G.Antitumour activity mediated by CD4+ cytotoxic Tlymphocytes against MHC class II-negative mousehepatocellular carcinoma induced by dendritic cellvaccine and interleukin-12. Immunology 2005;115:451^61.

21. Dannull J, Su Z, Rizzieri D, et al. Enhancement ofvaccine-mediated antitumor immunity in cancerpatients after depletion of regulatory T cells. J ClinInvest 2005;115:3623^33.

2 2 . Cla r ke SL , Be t t s GJ, P l an t A , e t a l .CD4+CD25+FOXP3+ regulatoryTcells suppress anti-tumor immune responses in patients with colorectalcancer. PLoSONE 2006;1:e129.

23. Imai H, Saio M, Nonaka K, et al. Depletion ofCD4+CD25+ regulatoryT cells enhances interleukin-2-induced antitumor immunity in a mouse model ofcolon adenocarcinoma. Cancer Sci 2007;98:416^23.

24. Grossman Z, Paul WE. Self-tolerance: context de-pendent tuning of T cell antigen recognition. SeminImmunol 2000;12:197^203.

25. Baccala R, Gonzalez-Quintial R, Dummer W,Theofilopoulos AN. Tumor immunity via homeostaticT cell proliferation: mechanistic aspects and clinical

perspectives. Springer Semin Immunopathol 2005;27:75^85.

26.Tallman MS, Gray R, Robert NJ, et al. Conventionaladjuvant chemotherapy with or without high-dosechemotherapy and autologous stem-cell transplanta-tion in high-risk breast cancer. N Engl J Med 2003;349:17^26.

27. Zander AR, Kroger N, Schmoor C, et al. High-dosechemotherapy with autologous hematopoietic stem-cell support compared with standard-dose chemo-therapy in breast cancer patients with 10 or morepositive lymph nodes: first results of a randomizedtrial. J Clin Oncol 2004;22:2273^83.

28. Rodenhuis S, Bontenbal M, Beex LV, et al. High-dose chemotherapy with hematopoietic stem-cellrescue for high-risk breast cancer. N Engl J Med2003;349:7^16.

29. ChengYC, Rondon G,YangY, et al. The use of high-dose cyclophosphamide, carmustine, and thiotepaplus autologous hematopoietic stem cell transplanta-tion as consolidation therapy for high-risk primarybreast cancer after primary surgery or neoadjuvantchemotherapy. Biol Blood MarrowTransplant 2004;10:794^804.

30. Durando X, Lemaire JJ, Tortochaux J, et al.High-dose BCNU followed by autologous hemato-poietic stem cell transplantation in supratentorialhigh-grade malignant gliomas: a retrospective anal-ysis of 114 patients. Bone Marrow Transplant 2003;31:559^64.

31. Gilboa E. The risk of autoimmunity associated withtumor immunotherapy. Nat Immunol 2001;2:789^92.

32. AsavaroengchaiW, KoteraY, MuleJJ.Tumor lysate-pulsed dendritic cells can elicit an effective antitumorimmune response during early lymphoid recovery.Proc Natl Acad Sci US A 2002;99:931^6.

33.Morgan RA, Dudley ME,Wunderlich JR, et al. Can-cer regression in patients after transfer of geneticallyengineered lymphocytes. Science 2006;314:126^9.

34. Juretic A, Spagnoli GC, Schultz-Thater E, SarcevicB. Cancer/testis tumour-associated antigens: immu-nohistochemical detection with monoclonal antibod-ies. Lancet Oncol 2003;4:104^9.

35. Muranski P, Boni A,Wrzesinski C, et al. Increasedintensity lymphodepletion and adoptive immunother-apyEhow far can we go? Nat Clin Pract Oncol2006;3:668^81.

36. Bhatia S, Robison LL, Francisco L, et al. Late mor-tality in survivors of autologous hematopoietic-celltransplantation: report from the Bone Marrow Trans-plant Survivor Study. Blood 2005;105:4215^22.

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

2007;13:7469-7479. Clin Cancer Res   Akihiko Kobayashi, Hidehiko Hara, Masaki Ohashi, et al.   Hematopoietic Stem Cell TransplantationAntitumor Immunity in the Early Period of Autologous Allogeneic MHC Gene Transfer Enhances an Effective

  Updated version

  http://clincancerres.aacrjournals.org/content/13/24/7469

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2007/12/14/13.24.7469.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/13/24/7469.full#ref-list-1

This article cites 36 articles, 16 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/13/24/7469.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://clincancerres.aacrjournals.org/content/13/24/7469To request permission to re-use all or part of this article, use this link

Research. on July 3, 2018. © 2007 American Association for Cancerclincancerres.aacrjournals.org Downloaded from