25.01.2013 Views

Cancer Immune Therapy Edited by G. Stuhler and P. Walden ...

Cancer Immune Therapy Edited by G. Stuhler and P. Walden ...

Cancer Immune Therapy Edited by G. Stuhler and P. Walden ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

16.5 Melanoma<br />

The remaining animals exhibited a 5-fold reduction in tumor growth compared to<br />

controls receiving PBS, the empty vector, or the DNA vaccine lacking ubiquitin. All animals<br />

in the three control groups grew large tumors (800±900 mm 3 ) <strong>and</strong> failed to reject<br />

the wild-type tumor cell challenge [75].<br />

Two lines of evidence indicated that the tumor-protective immunity induced <strong>by</strong> the<br />

DNA vaccine was mediated <strong>by</strong> MHC class I antigen-restricted CD8 + T cells secreting<br />

increased amounts of the pro-inflammatory cytokine IFN-g. First, adoptive transfer<br />

of CD8 + T cells from mice successfully immunized <strong>by</strong> the DNA vaccine to syngeneic<br />

scid/scid mice resulted in more than 60% reduction in s.c. tumor growth after challenge<br />

with wild-type B16 melanoma cells when compared with control animals that<br />

received CD8 + T cells from mice vaccinated with only the empty ubiquitin vector.<br />

Second, only CD8 + T cells isolated from splenocytes of successfully vaccinated mice<br />

induced MHC class I antigen-restricted CTL-mediated cytolytic killing of melanoma<br />

target cells in vitro. Significantly, the presence of ubiquitin upstream of the minigene<br />

proved to be essential for achieving this tumor-protective immunity, suggesting that<br />

more effective antigen processing in the proteasome <strong>and</strong> presentation <strong>by</strong> antigenpresenting<br />

cells made it possible to break peripheral T cell tolerance to tumor selfantigens<br />

[76, 77].<br />

Although these findings obtained with the DNA vaccine encoding melanoma minigenes<br />

were encouraging, we attempted to improve its efficacy <strong>by</strong> combining it with<br />

small boosts <strong>by</strong> sub-optimal doses of ch14.18±IL-2 immunocytokine. Actually, three<br />

lines of prior evidence provided the rationale for this approach. First, we demonstrate<br />

previously that immunocytokines targeted to the tumor microenvironment where<br />

they actively activated <strong>and</strong> exp<strong>and</strong>ed CD8 + T cell. In fact, combined with CD4 + T cell<br />

help, this approach induced effective antitumor responses <strong>and</strong> eradicated established<br />

metastases of murine colon carcinoma [11, 12] <strong>and</strong> murine melanoma [61, 62]. Second,<br />

we already demonstrated that the humanized huKS1/4±IL-2 immunocytokine<br />

(huKS1/4±IL-2) elicited a long-lived cellular-mediated memory immune response<br />

against murine colon carcinoma which, importantly, was substantially amplified <strong>by</strong><br />

additional boosts with non-curative, suboptimal doses of this same immunocytokine<br />

targeted to the tumor microenvironment. Third, systemically administered IL-2 was<br />

shown to be of key importance for boosting the efficacy of a variety of malignancies<br />

tested in the clinic [78, 79]. In fact, modified gp100209±217 peptide-induced T cell responses<br />

resulted in a striking objective clinical response rate of 42% among melanoma<br />

patients only when these individuals received concurrent boosts with high systemic<br />

doses of IL-2 [79]. Thus, IL-2 may actually be essential for breaking peripheral<br />

T cell tolerance against melanoma self-antigens.<br />

To prove this point, C57BL6/J mice immunized three times with the DNA vaccine<br />

encoding ubiquitin <strong>and</strong> the gp10025±53 <strong>and</strong> TRP-2181±189 minigenes, were challenged<br />

s.c. with wild-type murine melanoma cells, 1 week after the last immunization <strong>and</strong><br />

24 h later received i.v. boosts with 5 mg ch14.18±IL-2 for 4 consecutive days. This resulted<br />

in complete tumor rejection in 75% of experimental animals with the remaining<br />

mice revealing an almost 10-fold reduction in tumor growth when compared<br />

with control animals. These fusion-protein boosts markedly enhanced tumor-protective<br />

immunity when compared to the vaccine alone (P > 0.01), which resulted in tu-<br />

335

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!