ArticlePDF Available

Can immunotherapy by gene transfer tip the balance against colorectal cancer?

Authors:

Abstract

Gene therapy, in particular the transfer of genes encoding immunostimulatory molecules (cytokines and costimulatory molecules) as well as selectively cytotoxic enzymes and DNA vaccination, has the potential of enhancing cell mediated immune responses against tumours including those of colorectal origin. Genes can be transferred using viral vectors either to cultured tumour cells in vitro that can be returned to the patient as a "cancer vaccine", or directly to tumour cells in vivo. Vaccination with DNA constructs expressing specific tumour antigens characteristic of colorectal neoplasia can trigger immune recognition and destruction of tumour cells. The aim is to tip the balance from protumour to antitumour mechanisms by generating a local immune response and systemic antitumour immune memory to destroy metastases. Studies in murine models, combined with human studies, show that such approaches could become an adjunct to current treatments for human colorectal cancer in the near future.
Leading article
Can immunotherapy by gene transfer tip the balance against
colorectal cancer?
Summary
Gene therapy, in particular the transfer of genes encoding
immunostimulatory molecules (cytokines and costi-
mulatory molecules) as well as selectively cytotoxic
enzymes and DNA vaccination, has the potential of
enhancing cell mediated immune responses against
tumours including those of colorectal origin. Genes can be
transferred using viral vectors either to cultured tumour
cells in vitro that can be returned to the patient as a “can-
cer vaccine”, or directly to tumour cells in vivo.
Vaccination with DNA constructs expressing specific
tumour antigens characteristic of colorectal neoplasia can
trigger immune recognition and destruction of tumour
cells. The aim is to tip the balance from protumour to
antitumour mechanisms by generating a local immune
response and systemic antitumour immune memory to
destroy metastases. Studies in murine models, combined
with human studies, show that such approaches could
become an adjunct to current treatments for human
colorectal cancer in the near future.
Introduction
Colorectal cancer comprises 10–15% of deaths from
cancer in industrial nations, second only to lung cancer.1
Survival rates (40% >5 years) have remained stable over
the past 20 years and so a number of treatments to supple-
ment surgical resection and chemotherapy are under
investigation, including enhancement of the immune
response. This article considers gene therapy, in particular
the transfer of immunomodulatory genes and selectively
cytotoxic enzymes to tumour cells as well as DNA vaccina-
tion, as a means of enhancing cell mediated immunity spe-
cifically for the treatment of colorectal cancer.
The current model for colorectal tumorigenesis postu-
lates a multi-stage progression involving an accumulation
of gene mutations (APC, K-ras, p53, DNA mismatch
repair genes), alterations in gene expression (c-myc,
MHC) and chromosome losses, during which regulation
of cell growth is disrupted.2Dietary and inherited genetic
factors predispose to such changes. The majority of deaths
from colorectal cancer follow tumour metastasis to the
liver and treatment must be aimed at controlling local
regrowth after resection and distant metastases. Cell
mediated immunity (mainly CD8+ cytotoxic T lym-
phocytes (CTL)) is potentially the most eVective arm of
the immune response as CTL can recognise epitopes
processed and presented from any protein synthesised
within the tumour cell and can kill the cell specifically and
also anamnestically (memory cells). Cytokines from
CD4+ helper T cells (Th) are also required to activate not
only CTL, but also natural killer (NK) cells and antigen
presenting cells and other inflammatory cells at the
tumour site. Although lessons can be learned from gene
therapy approaches against other tumours, mainly
melanoma,3colorectal cancers have characteristic features
which require separate consideration.
The immune response to colorectal tumours and
reasons for its failure
The emergence of a tumour may be the result of an inad-
equate immune response on two fronts: poor or lack of
immunogenicity of the tumour cells and low eYciency of
the immune response against the tumour. However, colo-
rectal tumours do not develop more frequently in
immunodeficient individuals, unlike some other
tumours—for example, lymphomas in patients with AIDS,
skin tumours in transplant recipients. This suggests that
the tumour itself has immunomodulatory or immuno-
evasive, or both, properties.
Tumour cells often fail to present antigen due to the total
loss (in around 20% of colorectal neoplasia) or reduction
in expression of MHC class I molecules.45 Mutations in
peptide transporting molecules (TAP) may also aVect
presentation of T cell epitopes.6The genetic changes
occurring during tumour development frequently lead to
the expression of oncogenic and neo-antigens (tumour
specific) or aberrant expression of normal or fetal antigens,
which are potential targets for immune attack of the cancer
cells. Antigens recognised by T cells in colorectal cancer
include mutated p21 ras78 cell surface associated mucin9
and an annexin-like molecule.10 However, for an eVective
antitumour response T cell specificities may need to be
directed towards subdominant or cryptic epitopes of
unmutated self molecules as dominant epitopes may have
induced thymic depletion or peripheral anergy of epitope
specific T cells.11 A precedent for this is seen in melanoma
where a number of self antigens are associated with protec-
tive immunity to tumours.12
The total or partial loss of MHC class I molecules means
that tumour antigens may not be presented to CTL if a
particular MHC class I allele is required for peptide pres-
entation. This provides a selective advantage for the
tumour cells and a problem for the immune response. In
addition, the presence of MHC class I alleles can inhibit
Leading articles express the views of the author and not those of the editor and editorial board.
Gut 1998;43:445–449 445
the non-specific cytotoxic activities of NK cells. Thus, cer-
tain phenotypes of MHC expression can render the
tumour cells non-susceptible to direct cellular cytotoxicity
by CTL or NK cells. Loss of polarisation at the luminal
membrane of epithelial tumour cells gives rise to aberrant
expression of mucin molecules (such as MUC-1).13
Expression of mucin all over the cell membrane can mask
surface immunoregulatory molecules and inhibit interac-
tion between tumour and immune cells.14 However,
changes in mucin glycosylation may make the mucin a tar-
get for CTL activity and Th cell recognition without MHC
restriction due to its repeating and possible TcR
cross-linking properties.15 An additional paradox in the
importance of mucin is the fact that MUC-1 expression on
adenocarcinomas (or shed from them) can cause apoptosis
of T cells,16 which is another way in which the tumour may
evade the immune response. A number of tumour infiltrat-
ing lymphocyte (TIL) populations have been identified in
colorectal tumours, which may have an association with
increased patient survival.17 18 Indeed, TIL cultured in vitro
and adoptively returned to patients have resulted in varying
degrees of protection in other cancers.12 NK cells are par-
ticularly abundant in colorectal TIL19 as are CD4+ T cells
which outnumber CD8+.20 21 Colorectal tumour infiltrat-
ing T cells with a limited repertoire of T cell receptor vari-
able regions suggests tumour specific clonal expansion22 23
as does the possession of activation markers.24 T cells with
the ãä T cell receptor, which have been identified in
neoplastic as well as in healthy intestinal mucosa,25 have
specific killing activity towards epithelial-derived tumours
in a non-MHC restricted manner26 and may provide
another antitumour mechanism. Furthermore, both ãä T
cells,27 and NK cells28 seem to recognise heat shock
proteins (hsp), molecules which are constitutively ex-
pressed by colorectal neoplasia29 (S Todryk, unpublished
data). The upregulation of hsp by heating30 or gene
transfer30a could therefore be another means of improving
immune recognition of colorectal cancer.
Cell mediated immunity tends to be down regulated in
environments such as the gut in order to minimise damage
caused by excessive inflammation in response to the
barrage of antigens encountered. Indeed, colon adenomas
and carcinomas produce transforming growth factor
(TGF) â31 32 and interleukin (IL) 10,33 cytokines known to
suppress cell mediated responses, an eVect that may be
more pronounced within larger, established tumours.
Secretion by colorectal tumours of factors such as leukae-
mia inhibitory factor and prostaglandins may also have
immunosuppressive eVects.34 35 This could, in part, explain
why these tumours tend to develop and persist, despite the
presence of TIL. This suppression may also occur in gut
associated lymphoid tissue, mediated by T cells.36 Recent
evidence, however, has shown that colorectal tumour cells
secrete IL-7, a cytokine that can cause TIL to proliferate,
secrete tumour necrosis factor (TNF) áand lyse
autologous tumour cells.37
The absence of costimulation (e.g. by helper cytokines or
B7 binding) during recognition of tumour cells by T cells
results in anergy of tumour specific T cells,38 rendering
them ineVective. Such anergy may need to be reversed in
immunotherapy. In humans with colorectal cancer the
functional suppression of T cells in the TIL and periphery
seems to coincide with alterations in the T cell receptor
signal transduction mechanism,39 40 but these may be
reversible by cytokines such as IL-2.41 Finally, colorectal
tumours express not only functional Fas ligand, which can
induce apoptosis in tumour infiltrating T cells bearing Fas,
but also Fas itself, which although expressed at lower levels
than in normal colon epithelium may make the tumour
cells susceptible to apoptosis.42 43
In conclusion, the fact that it is possible to detect cellu-
lar immune responses specific for colorectal tumours in
vitro,44–48 albeit at low levels, suggests that the immune
defect could be reversed in vivo by immunotherapy.
Gene transfer mediated immunotherapy of
colorectal cancer
IMMUNOSTIMULATORY GENES
Immunostimulatory gene transfer is a potentially powerful
therapeutic approach for treating colorectal cancer that
aims to mobilise the immune response to recognise and
destroy tumour cells (box). Gene transfer therapy usually
involves the resection of tumour and then infection in vitro
of tumour cells with retro-, adeno- or herpes viruses49 con-
taining genes for cytokines and/or costimulatory mol-
ecules. This is followed by reinjection of the irradiated or
unirradiated cells as a “cancer vaccine”. When cytokine
genes are transferred, tumour cells will secrete the cytokine
and stimulate immune responses and inflammation by a
local paracrine eVect. When genes for costimulatory
molecules (e.g. B7) are transferred the molecule will be
expressed on the tumour cell surface and stimulate
lymphocytes by direct contact. Local production of
cytokines also avoids toxic eVects of systemic cytokine
administration. Together with the elimination of the
inoculating tumour cells, this approach aims to elicit
systemic immune memory and protection against second-
ary contact with parental tumour (subcutaneous or in the
liver in the mouse model), which represents tumour
regrowth and metastasis in humans. A number of
transferred cytokines have shown varying degrees of
protection in tumour models,50 with IL-2,51 GM-CSF52–54
and IL-1255 56 being most eVective and consistent at induc-
ing protective immunity in murine colorectal tumour
models. We have found IL-12 and B7.1 to be a
combination that elicits the greatest degree of protection54
and IL-12 can also give rise to CTL that successfully treat
colorectal tumour “metastases” in the lung.57 The local
release of these cytokines induces a cell mediated Th type
1 response (IL-12) or the stimulation of dendritic cell pre-
cursors (GM-CSF), which take up tumour antigens,
migrate to the lymph nodes and prime T cells giving rise to
eVector and memory T cells.58 CTL and/or NK cells medi-
ate the tumour rejection in these models, with51 or
without55 T cell help. The inflammatory environment cre-
ated by the transferred cytokines also enhances the expres-
sion (together with MHC molecules) and recognition of
less dominant self-antigen T cell epitopes which can then
become targets for CTL.11 In comparison to such murine
models, humans may have carried their tumours for long
periods prior to gene therapy, and both priming of naive
cells and reversal of anergy will be required. In addition,
selective pressure over many years will have caused the
tumour to adapt to, and evade, the immune response. In
human studies, the transfer of the B7 costimulatory
molecule to colorectal tumour cell lines did not cause acti-
vation of T cells in vitro,59 whereas the transfer of IL-2
Aims of immunostimulatory gene transfer
therapy against colorectal cancer
+Induce an immunostimulatory environment in the
vicinity of the tumour/vaccine
+Induce direct or cross-priming of cytotoxic and
helper T cells against tumour antigens
+Overcome immunosuppression and/or T cell anergy
+Generate immune memory against tumour regrowth
and metastasis
446 Todryk, Chong, Pandha, et al
stimulated NK cells in vitro but not tumour specific CTL.60
In human melanoma, exogenous IL-12 and IL-2 in
combination, but not B7, yielded the best in vitro CTL
responses.61
In vivo infection of tumour cells with tumour targeting
viruses (systemic or local administration) may also be fea-
sible in gene therapy62 and avoids in vitro cell manipulation
for each patient, and the production of a “personal”
vaccine. Liver metastases could—for example, be targeted
by perfusion of the liver with viruses via the hepatic portal
vein or by intratumoural injection. Carcinoembryonic
antigen (CEA) expressing tumours could be targeted by
engineering proteins within the viral envelopes that bind
specifically to surface CEA, or by incorporating the CEA
promotor.63 This in vivo gene delivery approach should
aVect the growth of the targeted tumour and elicit protec-
tive immunity against spread of the tumour. Alternatively,
the possibility exists of using allogeneic tumour cells, with
antigens in common with the patient’s tumour, which will
be rapidly destroyed and these antigens released, resulting
in T cell cross-priming against the antigen. Established
tumours, or tumour cells ex vivo, can also be made alloge-
neic by transfer of allo-MHC genes.64 In addition to
tumour cell modification, transfer of the gene for TNF-áto
TIL from melanomas has been achieved,65 and may
provide another means of enhancing cell mediated immu-
nity against colorectal neoplasia using gene therapy.
SUICIDE GENES
Another form of gene therapy involves the in vitro
(followed by injection) or in vivo infection of tumour cells
with viruses carrying “suicide” genes which encode
enzymes (e.g. herpes simplex virus thymidine kinase (tk)
and Escherichia coli cytosine deaminase (CD)) that convert
prodrugs (ganciclovir and 5-fluorocytosine, respectively)
into toxic forms that kill the tumour cells in vivo. This
inflammatory process rapidly releases antigens that stimu-
late memory immune responses resulting in the killing of
parental tumour cells distal from the initial tumours (e.g.
metastases).62 66 This approach has been successful at
reducing growth of a tumour challenge in a mouse
colorectal tumour model67 (and our unpublished data).
Moreover, cotransfection of tk with GM-CSF, adminis-
tered in adenovirus in vivo, was able to increase survival of
mice with liver metastases.68 The use of suicide genes may
also reduce the need for tumour cell irradiation, which
could adversely aVect the vaccine’s eYciency.69 The
tumour cells would be killed when the prodrug is adminis-
tered. In a comparison between tk and CD gene transfer,
CD was more eVective than tk at killing a human colorec-
tal tumour line in nude mice using in vitro,70 or in vivo
delivery (adenovirus).71 However, colorectal tumour lines
passaged over many years may not provide the most accu-
rate model for colorectal cancer therapy. For this reason we
are currently studying tumour cells freshly isolated from
patients.
DNA VACCINATION
The administration of genes encoding tumour associated
antigens provides another potential route of immuno-
therapy against colorectal cancer. Antigen encoding
plasmid DNA can be given in its naked form by intrader-
mal or intramuscular routes, and by injection or “gene
gun”. Alternatively, vehicles for DNA vaccination include
liposomes, viral vectors and protein carriers. The pro-
longed antigen expression that is obtained can induce CTL
and Th responses.72 Even though relatively few antigen
specific T cell responses have been identified for colorectal
tumours, as previously mentioned, there are a number of
candidate proteins that could be exploited as DNA
vaccines (table 1). Potential tumour specific antigens are
those expressed uniquely by the tumour, or in greater
abundance than normal tissue. In addition, as T cells rec-
ognise peptide epitopes of around eight to 20 amino acids,
MHC restriction of peptide recognition by the heterog-
enous human population may necessitate the use of larger
antigenic fragments that encompass many epitopes.
Mutations in oncogenes may be single amino acid
changes, as at codons 12, 13, and 61 in p21 ras. These
mutations disrupt normal ras signalling function and are
not expressed in normal tissue. Human CTL that
recognise a single ras mutation at residue 13 and are capa-
ble of killing tumour cells harbouring the same mutation
have been isolated from a patient with colon carcinoma.73
Peptides from this region of ras also bind MHC class II
molecules with high promiscuity,74 which is a desirable
attribute of a vaccine. Mutations in the p53 tumour
suppressor protein can give rise to multiple amino acid
substitutions. Such changes mean that cell growth is
unchecked and further gene mutations and chromosomal
rearrangements can accumulate. Murine CTL have been
raised to p53 mutated at codon 135.75 Carcinoembryonic
antigen is a glycosylated single-chain peptide overex-
pressed in carcinomas of the colon, breast, stomach,
pancreas, and lung. Promising CEA DNA vaccination
studies in mice76 77 using naked DNA or a vaccinia virus
vector are beginning to translate to human studies where
CTL generated by vaccinia-CEA immunisation could lyse
CEA+ tumour cells.78 Polymorphic epithelial mucin
encoded by the MUC-1 gene is overexpressed in a number
of adenocarcinomas. The mucin expression is no longer
only associated with the apical surface of ductal epithelial
cells and aberrant mucin glycosylation on tumour cells
results in exposure of the polypeptide core and unmasking
of otherwise cryptic epitopes. Immunisation with MUC-1
DNA, again naked or in vaccinia virus, has shown protec-
tion against tumour growth in mice.79 80 Although most
frequently associated with melanoma, the MAGE family of
genes has also been found in colorectal neoplasia81 and so
represent another potential candidate for DNA vaccina-
tion. Finally, mutational frameshifts such as those associ-
ated with APC gene expression can result in a stretch of
unique protein sequence containing potential T cell
epitopes.82
Recently, a potentially eVective route of DNA vaccina-
tion has emerged in which dendritic cells are pulsed or
transduced with tumour antigen encoding DNA and can
eYciently prime T cells.83 84 In light of the identification of
a number of tumour-regression antigens in melanoma and
Table 1 Candidate antigens for DNA vaccination against colorectal cancer
Antigen Class of antigen T cell responses Antitumour eVect/association
p21 ras Mutated oncogene product Human CTL, Th1 No
p53 Mutated tumour suppressor Murine CTL No
CEA Embryonic gene product Human CTL In mice
MUC-1 Epithelial mucin Non-MHC CTL In mice
MAGE Melanoma associated antigen Human CTL in melanoma In melanoma
GA733 Surface molecule Murine CTL, Th In mice88
Annexin-like molecules Placental/structural protein Th No
Treatment of colorectal cancer by gene transfer 447
other tumours, a requirement exists for the identification of
further tumour antigens in colorectal cancer.
Conclusion and future prospects
A number of cells of the immune system may be manipu-
lated in the gene therapy of colorectal cancer in order to tip
the balance from protumour to antitumour mechanisms
(fig 1). One of the challenges is to stimulate an eVective
immune response towards the various tumour phenotypes
and locations by transferring genes encoding the appropri-
ate immunostimulatory or cytotoxic molecules, or by
immunising with the appropriate tumour antigen encoding
DNA. EVective immune responses to colorectal neoplasia
that express or fail to express MHC class I molecules—for
example, may require diVerent immunostimulatory mol-
ecules to activate diVerent eVector cells.85 Tumour burden,
and in particular the size of the tumour, may very much
determine the success of such therapies as an immunosup-
pressive environment may be created and the tumour may
simply be proliferating too rapidly for the immune system
to contain. Previous studies immunising patients with
colorectal cancer with autologous tumour cells and bacillus
Calmette-Guerin (BCG) have shown some improvements
in survival rates86 and current gene transfer trials involve
transfer of allo-MHC molecules (HLA-B7)87 and cytosine
deaminase to colorectal tumours.3Genes encoding IL-12,
GM-CSF, B7, cytosine deaminase, and thymidine kinase
have shown therapeutic eYcacy in murine models of
colorectal cancer. DNA vaccination studies in murine
models that are currently translating to human studies
include CEA.
Colorectal cancer is amenable to gene therapy as
patients can be returned to a state of minimal residual dis-
ease following resection of the primary tumour. Latent
micrometastses will be a more controllable target. To put
these principles into practice we are currently working
towards clinical trials comprising in vitro transfection of
colorectal tumour cells with adenovirus encoding genes for
tk and GM-CSF, followed by reinjection of the cells as a
“vaccine”.89 These gene therapy approaches have the
potential to be useful adjuvants to conventional treatments
with potential advantages of being physiologically less toxic
and providing systemic vigilance against tumour regrowth
and metastasis.
This work is supported by the Lewis Family Charitable Trust (SMT) and the
Imperial Cancer Research Fund.
S M TODRYK
H CHONG
R G VILE
Laboratory of Molecular Therapy,
Imperial Cancer Research Fund Molecular Oncology Unit,
Imperial College School of Medicine,
Hammersmith Hospital, London W12 0NN, UK
H PANDHA
N R LEMOINE
Laboratory of Molecular Pathology,
Imperial Cancer Research Fund Molecular Oncology Unit,
Imperial College School of Medicine,
Hammersmith Hospital, London W12 0NN, UK
Correspondence to: Dr Stephen Todryk (email: s.todryk@icrf.icnet.uk).
1 Beart RW. Colon and rectum. In: AbeloVMD, ed. Clinical oncology.
Edinburgh: Churchill Livingstone, 1995:1267–86.
2 Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell
1996;87:159–70.
3 Roth JA, Cristiano RJ. Gene therapy for cancer: what have we done and
where are we going? J Natl Cancer Inst 1997;89:21–39.
4 Smith ME, Marsh SG, Bodmer JG, et al. Loss of HLA-A,B,C allele products
and lymphocyte function-associated antigen 3 in colorectal neoplasia. Proc
Natl Acad Sci USA 1989;86:5557–61.
5 Browning M, Petronzelli F, Bicknell D, et al. Mechanisms of loss of HLA
class I expression on colorectal tumor cells. Tissue Antigens 1996;47:364–
71.
6 Kaklamanis L, Townsend A, Doussis-Anagnostopoulou IA, et al. Loss of
major histocompatibility complex-encoded transporter associated with
antigen presentation (TAP) in colorectal cancer. Am J Pathol 1994;145:
505–9.
7 Fossum B, Gedde DT, Breivik J, et al. p21-ras-peptide-specific T-cell
responses in a patient with colorectal cancer. CD4+ and CD8+ T cells rec-
ognize a peptide corresponding to a common mutation (13GlyAsp). Int
J Cancer 1994;56:40–5.
8 Qin H, Chen W, Takahashi M, et al. CD4+ T-cell immunity to mutated ras
protein in pancreatic and colon cancer patients. Cancer Res 1995;55:2984–
7.
9 Kim JA, Martin E Jr, Morgan CJ, et al. Expansion of mucin-reactive
T-helper lymphocytes from patients with colorectal cancer. Cancer Biother
1995;10:115–23.
10 Ransom JH, Pelle BA, Hubers H, et al. Identification of colon-tumor-
associated antigens by T-cell lines derived from tumor-infiltrating
lymphocytes and peripheral-blood lymphocytes from patients immunized
with an autologous tumor-cell/bacillus Calmette-Guerin vaccine. Int J
Cancer 1993;54:734–40.
11 Nanda NK, Sercarz EE. Induction of anti-self-immunity to cure cancer.Cell
1995;82:13–17.
12 Rosenberg SA. Cancer vaccines based on the identification of genes encod-
ing cancer regression antigens. Immunol Today 1997;18:175–82.
13 Finn OJ, Jerome KR, Henderson RA, et al. MUC-1 epithelial tumor mucin-
based immunity and cancer vaccines. Immunol Rev 1995;145:61–89.
14 van-de-Wiel-van-Kemenade E, Ligtenberg MJ, de-Boer AJ, et al. Episialin
(MUC1) inhibits cytotoxic lymphocyte-target cell interaction. J Immunol
1993;151:767–76.
15 Barnd DL, Lan, MS, Metzgar RS, et al. Specific, major histocompatibility
complex-unrestricted recognition of tumor-associated mucins by human
cytotoxic T cells. Proc Natl Acad Sci USA 1989;86:7159–63.
16 Gimmi CD, Morrison BW, Mainprice BA, et al. Breast cancer-associated
antigen, DF3/MUC1, induces apoptosis of activated human T cells. Nat
Med 1996;2:1367–70.
17 Svennevig JL, Lunde OC, Holter J,et al. Lymphoid infiltration and progno-
sis in colorectal carcinoma. Br J Cancer 1984;49:375–7.
18 Di Giorgio A, Botti C, Tocchi A, et al. The influence of tumor lymphocytic
infltration on long term survival of surgically treated colorectal cancer
patients. Int Surg 1992;77:256–60.
19 Takii Y, Hashimoto S, Iiai T, et al. Increase in the proportion of granulated
CD56+ T cells in patients with malignancy. Clin Exp Immunol
1994;97:522–7.
20 Jackson PA, Green MA, Marks CG, et al. Lymphocyte subset infiltration
patterns and HLA antigen status in colorectal carcinomas and adenomas.
Gut 1996;38:85–9.
21 Balch CM, Riley LB, Bae YJ, et al. Patterns of human tumor-infiltrating
lymphocytes in 120 human cancers. Arch Surg 1990;125:200–5.
22 Ostenstad B, Sioud M, Lea T, et al. Limited heterogeneity in the T-cell
receptor V-gene usage in lymphocytes infiltrating human colorectal
tumours. Br J Cancer 1994;69:1078–82.
23 Sensi M, Parmiani G. Analysis of TCR usage in human tumors: a new tool
for assessing tumor-specific immune responses. Immunol Today 1995;16:
588–95.
24 Ostenstad B, Lea T, Schlichting E, et al. Human colorectal tumour infiltrat-
ing lymphocytes express activation markers and the CD45RO molecule,
showing a primed population of lymphocytes in the tumour area. Gut
1994;35:382–7.
25 Watanabe N, Hizuta A, Tanaka N, et al. Localization of T cell receptor
(TCR)-gamma delta + T cells into human colorectal cancer: flow cytomet-
ric analysis of TCR-gamma delta expression in tumour-infiltrating
lymphocytes. Clin Exp Immunol 1995;102:167–73.
26 Maeurer MJ, Martin D, Walter W, et al. Human intestinal Vdeltal+
lymphocytes recognize tumor cells of epithelial origin. J Exp Med
1996;183:1681–96.
27 Rajasekar R, Sim GK, Augustin A. Self heat shock and gamma delta T-cell
reactivity. Proc Natl Acad Sci USA 1990;87:1767–71.
28 MulthoVG, Botzler C, Jennen L, et al. Heat shock protein 72 on tumour
cells: a recognition structure for natural killer cells. J Immunol
1997;158:4341–50.
Figure 1 Tipping the balance in colorectal cancer.A number of
protumour mechanisms outweigh antitumour mechanisms and allow
colorectal tumours to survive and proliferate. Gene therapy with cytokine,
immunostimulatory or suicide (prodrug activating) gene transfer to
tumour cells, or DNA vaccination, aims to tip the balance towards
antitumour mechanisms and tumour rejection by enhancing antitumour
cellular immune responses. Correction of immune deficiencies associated
with the tumour could work in synergy with enhancement of antitumour
immunity and many of the genes shown could apply to both sides of the
balance.
Protumour mechanisms
Loss or partial loss of MHC molecules
FasL mediated apoptosis of T cells
MUC-1 mediated apoptosis of T cells
Immunosuppressive cytokines
No tumour antigen release
Antitumour mechanisms
MHC restricted CTL
MHC non-restricted NK cells
Fas mediated apoptosis of tumour cells
γδ T cell recognition of hsp
CTL recognition of MUC-1
Tumour antigen release and cross-priming
IL-12
GM-CSF
Prodrug activating
genes
DNA vaccines
IL-2
B7
Tumour
persistence
Gene therapy to counter
protumour factors
Gene therapy to increase
antitumour factors
Tumour
rejection
448 Todryk, Chong, Pandha, et al
29 Lazaris AC, Theodoropoulos GE, Davaris PS, et al. Heat shock protein 70
and HLA-DR molecules tissue expression. Prognostic implications in
colorectal cancer. Dis Colon Rectum 1995;38:739–45.
30 Wei Y, Zhao X, Kariya Y, et al. Induction of autologous tumor killing by heat
treatment of fresh human tumor cells: involvement of gamma delta T cells
and heat shock protein 70. Cancer Res 1996;56:1104–10.
30a Melcher A, Todryk S, Hardwick M, et al. Tumour immunogenicity is
determined by the mechanism of cell death via induction of heat shock
protein. Nat Med 1998:4:581–7.
31 Avery A, Paraskeva C, Hall P, et al. TGF-beta expression in the human
colon: diVerential immunostaining along crypt epithelium. Br J Cancer
1993;68:137–9.
32 Tsushima H, Kawata S,Tamura S, et al. High levels of transforming growth
factor beta 1 in patients with colorectal cancer: association with disease
progression. Gastroenterology 1996;110:375–82.
33 Gastl GA, Abrams JS, Nanus DM, et al. Interleukin-10 production by
human carcinoma cell lines and its relationship to interleukin-6 expression.
Int J Cancer 1993;55:96–101.
34 Burg C, Patry Y, Le-Pendu J, et al. Leukaemia inhibitory factor derived from
rat colon carcinoma cells increases host susceptibility to tumour growth.
Cytokine 1995;7:784–92.
35 Kubota Y, Sunouchi K, Ono M, et al. Local immunity and metastasis of
colorectal carcinoma. Dis Colon Rectum 1992;35:645–50.
36 Harada M, Matsunaga K, Oguchi Y, et al.The involvement of transforming
growth factor beta in the impaired antitumor T-cell response at the
gut-associated lymphoid tissue (GALT). Cancer Res 1995;55:6146–51.
37 Maeurer MJ, Walter W, Martin D, et al. Interleukin-7 (IL-7) in colorectal
cancer: IL-7 is produced by tissues from colorectal cancer and promotes
preferential expansion of tumour infiltrating lymphocytes. Scand J Immunol
1997;45:182–92.
38 Harding FA, McArthur JG, Gross JA, et al. CD28-mediated signalling
co-stimulates murine T cells and prevents induction of energy in T-cell
clones. Nature 1992;356:607–9.
39 Nakagomi H, Petersson M, Magnusson I, et al. Decreased expression of the
signal-transducing zeta chains in tumor-infiltrating T-cells and NK cells of
patients with colorectal carcinoma. Cancer Res 1993;53:5610–12.
40 Matsuda M, Petersson M, Lenkei R, et al. Alterations in the signal-
transducing molecules of T cells and NK cells in colorectal tumor-
infiltrating, gut mucosal and peripheral lymphocytes: correlation with the
stage of the disease. Int J Cancer 1995;61:765–72.
41 Zier K, Gansbacher B, Salvadori S.Preventing abnormalities in signal trans-
duction of T cells in cancer: the promise of cytokine gene therpay.Immunol
Today 1996;17:39–45.
42 O’Connell J, O’Sullivan GC, Collins JK, et al. The Fas counterattack: Fas-
mediated T cell killing by colon cancer cells expressing Fas ligand. JExp
Med 1996;184:1075–82.
43 Moller P, Koretz K, Leithauser F, et al. Expression of APO-1 (CD95), a
member of the NGF/TNF receptor superfamily, in normal and neoplastic
colon epithelium. Int J Cancer 1994;57:371–7.
44 Shimizu Y, Iwatsuki S, Herberman RB, et al.EVects of cytokines on in vitro
growth of tumor-infiltrating lymphocytes obtained from human primary
and metastatic liver tumors. Cancer Immunol Immunother 1991;32:280–8.
45 Yoo YK, Heo DS, Hata K, et al. Tumor-infiltrating lymphocytes from
human colon carcinomas. Functional and phenotypic characteristics after
long-term culture in recombinant interleukin 2 [see comments]. Gastroen-
terology 1990;98:259–68.
46 Hom SS, Rosenberg SA, Topalian SL. Specific immune recognition of
autologous tumor by lymphocytes infiltrating colon carcinomas: analysis by
cytokine secretion. Cancer Immunol Immunother 1993;36:1–8.
47 Bateman WJ, Donnellan I, Fraser IA, et al. Lymphocytes infiltrating color-
ectal cancer have low proliferative capacity but can secrete normal levels of
interferon gamma. Cancer Immunol Immunother 1995;41:61–7.
48 Jacob L, Somasundaram R, Smith W, et al. Cytotoxic T-cell clone against
rectal carcinoma induced by stimulation of a patient’s peripheral blood
mononuclear cells with autologous cultured tumour cells. Int J Cancer
1997;71:325–32.
49 Vile R, Russell SJ. Gene transfer technologies for the gene therapy of cancer.
Gene Ther 1994;1:88–98.
50 Tepper RI, Mule JJ. Experimental and clinical studies of cytokine
gene-modified tumor cells. Hum Gene Ther 1994;5:153–64.
51 Fearon ER, Pardoll DM, Itaya T, et al. Interleukin-2 production by tumor
cells bypasses T helper function in the generation of an antitumor response.
Cell 1990;60:397–403.
52 DranoVG, JaVee E, Lazenby A,et al. Vaccination with irradiated tumor cells
engineered to secrete murine granulocyte-macrophage colony-stimulating
factor stimulates potent, specific, and long-lasting anti-tumor immunity.
Proc Natl Acad Sci USA 1993;90:3539–43.
53 Gunji Y, Tagawa M, Matsubara H, et al. Antitumor eVect induced by the
expression of granulocyte macrophage-colony stimulating factor gene in
murine colon carcinoma cells. Cancer Lett 1996;101:257–61.
54 Chong H, Todryk S, Hutchinson G, et al. Tumour cell expression of B7
costimulatory molecules and interleukin-12 or granulocyte-macrophage
colony-stimulating factor induces a local anti-tumour response and may
generate systemic protective immunity.Gene Ther 1998:5:223–32.
55 MartinoYA, Stoppacciaro A, Vagliani M, et al. CD4 T cells inhibit in vivo
the CD8-mediated immune response against murine colon carcinoma cells
transduced with interleukin-12 genes. Eur J Immunol 1995;25:137–46.
56 Colombo MP, Vagliani M, Spreafico F, et al. Amount of interleukin 12 avail-
able at the tumor site is critical for tumor regression. Cancer Res 1996;56:
2531–4.
57 Rodolfo M, Zilocchi C, Melani C, et al. Immunotherapy of experimental
metastases by vaccination with interleukin gene-transduced adenocarci-
noma cells sharing tumor-associated antigens. Comparison between IL-12
and IL-2 gene-transduced tumor cell vaccines. J Immunol 1996;157:5536–
42.
58 Huang AY, Golumbek P, Ahmadzadeh M, et al. Role of bone marrow-
derived cells in presenting MHC class I-restricted tumor antigens. Science
1994;264:961–5.
59 Habicht A, Lindauer M, Galmbacher P, et al. Development of immunogenic
colorectal cancer cell lines for vaccination: expression of CD80 (B7.1) is
not suYcient to restore impaired primary T cell activation in vitro. Eur J
Cancer 1995;2:396–402.
60 Lindauer M, Schackert HK, Geber t J, et al. Immune reactions induced by
interleukin-2 transfected colorectal cancer cells in vitro: predominant
induction of lymphokine-activated killer cells. J Mol Med 1996;74:43–9.
61 De Wit D, Flemming CL, Harris JD, et al. K. IL-12 stimulation but not B7
expression increases melanoma killing by patient cytotoxic T lymphocytes
(CTL). Clin Exp Immunol 1996;105:353–9.
62 Vile RG, Nelson JA, Castleden S, et al. Systemic gene therapy of murine
melanoma using tissue specific expression of the HSVtk gene involves an
immune component. Cancer Res 1994;54:6228–34.
63 Tanaka T, Kanai F, Okabe S, et al. Adenovirus-mediated prodrug gene
therapy for carcinoembryonic antigen-producing human gastric carcinoma
cells in vitro. Cancer Res 1996;56:1341–5.
64 Nabel GJ, Gordon D, Bishop DK, et al. Immune response in human
melanoma after transfer of an allogeneic class I major histocompatibility
complex gene with DNA-liposome complexes. Proc Natl Acad Sci USA
1996;93:15388–93.
65 Hwu P, Yannelli J, Kriegler M, et al. Functional and molecular characteriza-
tion of tumor-infiltrating lymphocytes transduced with tumor necrosis
factor-alpha cDNA for the gene therapy of cancer in humans. J Immunol
1993;150:4104–15.
66 Vile RG, Castleden J, Marshall R, et al Generation of an anti-tumour
immune response in a non-immunogenic tumour: HSVtk killing stimulates
a mononuclear cell infiltrate and a Th1-like profile of intratumoural cyto-
kine expression. Int J Cancer 1997:71:267–74.
67 Caruso M, Pham-Nguyen K, Kwong YL, et al. Adenovirus-mediated
interleukin-12 gene therapy for metastatic colon carcinoma. Proc Natl Acad
Sci USA 1996;93:11302–6.
68 Chen SH, Kosai K, Xu B, et al. Combination suicide and cytokine gene
therapy for hepatic metastases of colon carcinoma: sustained antitumor
immunity prolongs animal survival. Cancer Res 1996;56:3758–62.
69 Allione A, Consalvo M, Nanni P, et al. Immunizing and curative potential of
replicating and nonreplicating murine adenocarcinoma cells engineered
with interleukin (IL)-2, IL-4, IL-6, IL-7, IL-10, tumor necrosis factor
alpha, granulocyte-macrophage colony-stimulating factor, and gamma-
interferon gene admixed with conventional adjuvants. Cancer Res 1994;54:
6022–6.
70 Trinh QT, Austin EA, Murray DM, et al. Enzyme/prodrug gene therapy:
comparison of cytosine deaminase/5-fluorocytosine versus thymidine
kinase/ganciclovir enzyme/prodrug systems in a human colorectal carci-
noma cell line. Cancer Res 1995;55:4808–12.
71 Ohwada A, Hirschowitz EA, Crystal RG. Regional delivery of an adenovirus
vector containing the Escherichia coli cytosine deaminase gene to provide
local activation of 5-fluorocytosine to suppress the growth of colon
carcinoma metastatic to liver. Hum Gene Ther 1996;7:1567–76.
72 Corr M, Lee D, Carson D, et al. Gene vaccination with naked plasmid DNA:
mechanism of CTL priming. J Exp Med 1996;184:1555–60.
73 Fossum B, Olsen AC, Thorsby E, et al. CD8+ T cells from a patient with
colon carcinoma, specific for a mutant p21-ras-derived peptide (Gly13-
Asp), are cytotoxic towards a carcinoma cell line harbouring the same
mutation. Cancer Immunol Immunother 1995;40:165–72.
74 Fossum B, Gedde-Dahl T, Hansen T, et al. Overlapping epitopes
encompassing a point mutation (12 Gly-Arg) in p21 ras can be recognized
by HLA-DR, -DP and -DQ restricted T cells. Eur J Immunol
1993;23:2687–91.
75 Yanuck M, Carbone DP, Pendleton CD, et al. A mutant p53 tumor suppres-
sor protein is a target for peptide-induced CD8+ cytotoxic T-cells. Cancer
Res 1993;53:3257–61.
76 Kantor J, Irvine K, Abrams S, et al. Antitumor activity and immune
responses induced by a recombinant carcinoembryonic antigen-vaccinia
vaccine. J Natl Cancer Inst 1992;84:1084–91.
77 Conry RM, LoBuglio AF, Loechel F, et al. A carcinoembryonic antigen
polynucleotide vaccine has in vivo antitumour activity. Gene Ther
1995;2:59–65.
78 Tsang KY, Zaremba S, Nieroda CA, et al.Generation of human cytotoxic T
cells specific for human carcinoembryonic antigen epitopes from patients
immunized with recombinant vaccinia-CEA vaccine. J Natl Cancer Inst
1995;87:982–90.
79 Acres RB, Hareuveni M, Balloul JM, et al. Vaccinia virus MUC1 immuniza-
tion of mice: immune responses and protection against the growth of
murine tumors bearing the MUC1 antigen. J Immunother 1993;14:136–43.
80 Graham R, Burchell J. Intramuscular immunization with MUC-1 cDNA
can protect C57 mice challenged with MUC-1-expressing syngeneic tumor
cells. Int J Cancer 1996;65:664–70.
81 Mori M, Inoue H, Mimori K, et al. Expression of MAGE genes in human
colorectal carcinoma. Ann Surg 1996;224:183–8.
82 Townsend A, Ohlen C, Rogers M, et al. Source of unique antigens. Nature
1994;371:662.
83 Henderson RA, Nimgaonkar MT, Watkins SC, et al. Human dendritic cells
genetically engineered to express high levels of the human epithelial tumor
antigen mucin (MUC-1). Cancer Res 1996;56:3763–70.
84 Reeves M, Royal R, Lam J, et al. Retroviral transduction of human dendritic
cells with a tumor-associated antigen gene. Cancer Res 1996;56:5672–7.
85 Levitsky HI, Lazenby A, Hayashi RJ, et al. In vivo priming of two distinct
antitumor eVector populations: the role of MHC class I expression. JExp
Med 1994;179:1215–24.
86 Hoover H Jr, Brandhorst JS, Peters LC, et al. Adjuvant active specific immu-
notherapy for human colorectal cancer: 6.5-year median follow-up of a
phase III prospectively randomized trial. J Clin Oncol 1993;11:390–9.
87 Rubin J, Galanis E, Pitot HC, et al. Phase I study of immunotherapy of
hepatic metastases of colorectal carcinoma by direct gene transfer of an
allogeneic histocompatibility antigen, HLA-B7. Gene Ther 1997;4:419–25.
88 Li W, Berencsi K, Basak S, et al. Human colorectal cancer (CRC) antigen
CO17–1A/GA733 encoded by adenovirus inhibits growth of established
CRC cells in mice. J Immunol 1997;159:763–9.
89 Diaz RM, Todryk S, Chong H, et al. Rapid adenoviral transduction of
freshly resected tumour explants with therapeutically useful genes provides
a rationale for genetic immunotherapy for colorectal cancer. Gene Ther
1998;5:869–79.
Treatment of colorectal cancer by gene transfer 449
... 30 However, the presence of DC in tumours and the surrounding areas per se is not always sufficient for tumour rejection.The development of a tumour may result from either poor immunogenicity of the tumour cells or low efficiency of the immune response to the tumour. In colorectal cancer, it is believed that the tumour itself has either or both immunomodulatory and immunoevasive properties, 31 probably through the inhibition of development and maturation of functional DC and their recruitment into tumours. Thus, colorectal cancer cells have been shown to release soluble factors (IL-10, transforming growth factor-b, and vascular endothelial growth factor), [32][33][34] which can inhibit the maturation of DC. 35 Troy et al. found that there is no active recruitment of DC into cancer tissues and that those present are only minimally activated. ...
Article
Human tumours including those of the gastrointestinal tract express a number of specific antigens that can be recognized by T cells, thus providing potential targets for cancer immunotherapy. Dendritic cells (DC) are rare leucocytes that are uniquely potent in their ability to capture, process and present antigens to T cells, and so selectively migrate through tissues to reach lymph nodes and spleen where initiation of immune responses takes place. Studies in murine tumour models have shown clearly that DC are capable of presenting tumour antigens to initiate tumour-specific cytotoxic T cell responses, and DC vaccination can induce anti-tumour activity against both primary tumours and pre-established tumour metastases. These findings together with the ability to culture sufficient numbers of DC from human bone marrow or blood progenitors have prompted the current major interest in their potential use in human tumour vaccination. Vaccine production involves harvesting autologous DC from cultured peripheral blood mononuclear cells in the presence of a cocktail of cytokines, ex vivo exposure of the DC to tumour antigens and return of pulsed DC to the patient to induce tumour immunity. Reports from Phase I/II clinical trials indicate that DC vaccines are safe with little or no side effect, and are capable of initiating antigen-specific T cell responses. Furthermore, defined tumour antigens are not necessarily required, which may make the process more applicable to human cancers, including many gastrointestinal cancers that lack well-characterized tumour-specific antigens. Additional trials of DC vaccination for a variety of human cancers including colorectal cancers are under way, and refinement of vaccine protocols and methods for targeting tumour antigens to DC in vivo are also being explored. There is reason to believe that DC-based vaccination could become an adjunct to current treatments for human cancers including colorectal cancer in the foreseeable future.
Article
Colorectal cancer accounts for 10% of all cancer-related deaths. The overall 5-year survival rate is still around 50%. This article reviews actual problems, open questions, and potential future treatment modalities of colorectal carcinoma. The value of the sentinel node and of micrometastases is discussed, pros and contras of technical aspects are elucidated. An outlook to future gene therapy perspectives concludes this review.
Article
A unique, comprehensive reference that integrates the molecular, cellular, physiological, pathological, and engineering aspects of regenerative processes. Bioregenerative engineering is an emerging discipline based on applying engineering principles and technologies to regenerative medicine. It induces, modulates, enhances, and/or controls regenerative processes by using engineering approaches to improve the restoration of the structure and function of disordered or lost molecules, cells, tissues, and organs. This reference systematically summarizes bioregenerative engineering principles, technologies, and current research to help scientists understand biological regeneration and design new therapeutic strategies. Succinct and well-organized with a detailed table of contents to help readers pinpoint information, this reference: Provides the fundamental theory and principles of molecular, cellular, and tissue regenerative engineering concurrently with experimental approaches. Presents the foundations of bioregenerative engineering, encompassing the molecular basis, the regulatory mechanism of regeneration, and the developmental aspects. Combines molecular and cell biology with potential applications. Addresses experimental design, methods, and modeling at the molecular/cellular/tissue levels. Covers the general mechanisms and technologies of bioregenerative engineering, as well as its application to the treatment of human disorders. Discusses the engineering tests and therapies for major organ systems. Presenting an in-depth introduction to the biological and engineering aspects of the field and an up-to-date overview of current research, this is a one-of-a-kind resource for scientific researchers and medical practitioners, as well as for graduate and undergraduate students in biomedical engineering, bioengineering, chemical engineering, molecular biology, and cell biology.
Article
Colorectal cancer is a leading cause of cancer mortality in Western countries. Gene therapy has been proposed as a potential novel treatment modality for colorectal cancer, but it is still in an early stage of development. The preclinical data have been promising and numerous clinical trials are underway. This brief review aims to summarise the current status of clinical trials of different gene therapy strategies, including immune stimulation, mutant gene correction, prodrug activation and oncolytic virus therapy, for patients with colorectal cancer. Data from phase I trials have proven the safety of the reagents but have not yet demonstrated significant therapeutic benefit. In order to achieve this and extend the scope of the treatment, continuing efforts should be made to improve the antitumour potency, efficiency of gene delivery and accuracy of gene targeting.
Article
Full-text available
The expression of immunosuppressive molecules influences in the prognosis, metastasis appearance and cytostatic treatment response. Among these molecules, HLA-G and indoleamine 2,3-dioxygenase (IDO) are sobreexpressed in cancer, such as in gastrointestinal tumours, and they could contribute to its development. Their expression by tumor cells or by immune system cells facilitates that tumour cells scape from immune attack and could induce immunotolerance in other body sites, and also influence in treatment effectiveness. The knowledge of tolerogenic capacity of tumours helps not only in the understanding of cancer growth but also in developing new therapeutic approaches.
Article
New modalities of treatment for colorectal cancer are required to support and improve those currently available. One such approach is immunotherapy by transfer of immunostimulatory genes to tumor cells. Here, we report the use of a herpes simplex virus (HSV) vector that is capable of a single round of infection (disabled infectious single-cycle [DISC]-HSV) as a gene transfer vehicle for colorectal cancer. This vector has potential advantages over other vectors for cancer immunotherapy in that it lyses infected tumor cells. Infection with DISC-HSV inhibited tumor cell growth both in vitro and in vivo. In addition, DISC-HSV-mediated cell killing occurs by both apoptotic and necrotic mechanisms. A range of colorectal tumor cell lines could be rapidly transduced with DISC-HSV/lacZ (14-90% in 4 hr). Both tumor prevention and tumor therapy protocols showed clear antitumor effects with DISC-HSV/mGM-CSF. In the prophylactic approach, an infected/irradiated whole cell vaccine protected up to 80% of mice from rechallenge. In addition, intratumoral injection of established tumors with DISC-HSV/GM-CSF caused rejection in 40% of mice and generated some protection from subsequent rechallenge. In both cases, however, it is clear that a dominant therapeutic effect of the DISC-HSV vector derives from its oncolytic properties, irrespective of the transduced gene. As a prelude to taking these studies forward to human clinical trials, we demonstrate that tumor cells could be successfully grown from freshly obtained human colorectal cancer resections (within 1 week of surgery), were transduced with DISC-HSV/hGM-CSF, and secreted the cytokine. This study provides the preclinical basis for trials of immunotherapy of colorectal cancer using DISC-HSV.
Article
Colorectal cancer is the second most common cause of cancer mortality in Western countries. Gene therapy represents a novel approach to the treatment of colorectal cancer, and this review addresses the current strategies and ongoing clinical trials, including gene correction, immunomodulatory approaches and virus-directed enzyme-prodrug systems. Although the pre-clinical results for these strategies have been encouraging, clinical trials have not yet reflected these data. However, gene therapy for colorectal cancer is still in the early stages of development, and its potential, particularly in combination with conventional cancer therapies, warrants further investigation.
Article
The elderly population has much to gain from the advances of molecular medicine, although at present genetic pharmacology remains mostly at the conceptual level. Cancer, in particular, is an increasing health burden and the majority (over 70%) of gene therapy trials are aimed at tackling this problem. Available strategies employ both viral and synthetic vectors with the selective delivery and expression of therapeutic genes a pivotal requirement. Clinical trials are now in progress with a view to modulating disease at many different levels, including the direct replacement of abnormal genes. suicide-gene formulations, and the delivery of 'gain of function' genes, which seek to alter the malignant phenotype by indirect means, such as, immunopotentiation and stromal reorganisation. Early data from these studies is tantalising and we must remain optimistic that gene therapy will benefit the patient with cancer by both reducing morbidity and extending life.
Article
An ability to induce a specific immune response to cancer would provide an important new dimension in its management. We report our initial work investigating the safety and efficacy of a dendritic cell vaccine in patients with colorectal cancer. Fifteen (15) patients with advanced colorectal cancer had vaccines prepared from autologous dendritic cells pulsed with tumor RNA and keyhole limpet hemocyanin. Vaccines were administered intravenously and patients were observed in hospital for 2 days. Thereafter, consultations were at monthly intervals at which time booster doses were given to a total of 4. Patients were monitored with weekly blood tests, including carcinoembryonic antigen, and 3-monthly computed tomography scans. Flow cytometry confirmed dendritic cell phenotype and in vitro function was confirmed by mixed lymphocyte reaction. No major adverse effects were observed. Eleven of 13 patients tested developed a positive keyhole limpet hemocyanin skin test and in 7 the carcinoembryonic antigen fell suggesting some in vivo anticancer effect. To date no dramatic clinical responses have been observed but follow-up is very short. The therapy was well tolerated. Dendritic cells were verified by phenotype and in vitro function. The positive keyhole limpet hemocyanin skin test confirms in vivo function by effective vaccination to keyhole limpet hemocyanin. Demonstration of any anticancer efficacy will require further follow-up.
Article
Genetic prodrug activation therapy (GPAT) is a form of cancer gene therapy that has potential use against tumours such as colorectal malignancy. The characterization of such therapies using laboratory models provides a basis for clinical trials. In this study the gene encoding Herpes Simplex Virus thymidine kinase (HSVtk) was delivered to colorectal tumour cells using an Adenoviral (Ad) vector in vitro. In this way the cells were made susceptible to killing with the prodrug ganciclovir to various degrees depending on cell infectability with Ad. Bystander killing effect appeared minimal both in vitro and when transduced cells were injected in vivo. Mechanisms of cell death, measured in vitro using anti-BrDU (DNA-break labelling) and propidium iodide staining variously showed a combination of apoptosis in the G1 cell cycle phase and late apoptotic or necrotic sub-G1 DNA fragmentation, depending on the tumour cell line. These findings suggest that gene therapy of colorectal cancer by GPAT gives rise to therapeutic forms of direct cell death, but requires improvements in transduction, and possibly immune augmentation.
Article
Full-text available
Sllmm~ Downregulation of major histocompatibility complex (MHC) class I expression is an important mechanism by which tumors evade classical T cell-dependent immune responses. Therefore, a system was designed to evaluate parameters for active immunization against MHC class I-tumors. Mice were capable of rejecting a MHC class I-tumor challenge after immunization with an irradiated granulocyte/macrophage colony-stimulating factor (GM-CSF) transduced MHC class I-tumor vaccine. This response was critically dependent on CD4 + T cells and natural killer (NK) cells, but minimally on CD8 + T cells. A strong protective response against MHC class I + variants of the tumor could be elicited when mice were immunized with irradiated MHC class I + GM-CSF-secreting tumor cells. This response required CD4 + and CD8 + T cells, and in addition, elimination of NK cells resulted in outgrowth of tumors that had lost expression of at least one MHC class I gene. Finally, class I MHC expression on the vaccinating cells inhibited the response generated against a MHC class I-tumor challenge. These results demonstrate that the host is capable of being immunized against a tumor that has lost MHC class I expression and reveal conditions under which distinct effector cells play a role in the systemic antitumor immune response.
Article
Full-text available
Cytotoxic T-lymphocytes (CTL) recognize processed peptide fragments of any endogenous protein, after these peptides are carried to the cell surface by class I major histocompatibility molecules. Thus, a tumor antigen does not have to be expressed as an intact protein on the cell surface to be recognizable by CTL. However, mutant oncogene products have not yet been shown to be targets of CD8+ CTL. Here, we generate p53-specific CD«1CTL by immunizing BÂLB/c mice with spleen cells pulsed with a peptide, corresponding to a 21-amino acid sequence encom passing a point mutation (135 Cys to Tyr) in the mutant p53 gene product from a human lung carcinoma. The mutation created a new A'''class I molecule binding motif sequence, and the determinant recognized was mapped to this motif and presented by the Kd class I molecule. The wild type peptide, without the mutation, was not recognized. Importantly, the CTL killed specifically BALB/c fibroblasts transfected with the mutant p53 gene and endogenously expressing the mutant protein, but not control fibroblasts or ones transfected with a different human mutant p53 gene. Thus, endogenously synthesized mutantp53, at levels found in tumors, can render cells targets for specific CTL, and these CTL can be generated by peptide immunization. These findings point the way toward an approach to selective immunotherapy against tumors.
Article
Full-text available
To evaluate the efficacy of vaccinations with cytokine-gene-trans duced tumor cells, BALB/c mice were challenged with 1 x 10' parental cells of a syngeneic adenocarclnoma cell line (TSA-pc). No protection was observed in mice Immunized 30 days earlier with 1 X 10' nonrep licatingmltomycin-C.treated TSA-pcalone,or with Co,ynebacterium parvum or Complete Freund Adjuvant (CFA). Ten to 30% of mice Immunized with nonreplicating cells engineered to produce Interleukin (IL)-2,IL-4, IL-6,IL-7, IL-b, tumornecrosisfactora, granulocyte macrophage colony-stimulating factor, and i-interferon gene were protected. Fifty % of mice immunized with replicating TSA-pc ad mixedwith C.parvumand 80â€"100% of miceimmunizedwith replicat ing tumor cells transduced with IL-2, IL-4, IL-7, IL-b, or y-interferon gene were protected. No cure was afforded by TSA cells admixed with C.parvumor CFA,nor by TSA cellsengineeredwith IL-6,granulocyte macrophagecolony-stimulating factor, and tumornecrosisfactor a geneInjectedstarting1 day after TSA-pcchallenge.Completetumor regression, however,wasobtainedIn10â€"20% ofmicetreatedwithTSA cells transducedwith IL-2, IL-4, IL-7,or IL-lOand In 30%of those treated with TSA cells transduced with @-lnterferon gene.
Article
Full-text available
Occupancy of the T-cell antigen receptor is insufficient to induce T-cell activation optimally; a second co-stimulatory signal is required. Exposure of T-cell clones to complexes of antigen with major histocompatibility complex molecules in the absence of the co-stimulatory signal induces a state of clonal anergy. This requirement for two stimuli for T-cell activation could have an important role in vivo in establishing peripheral tolerance to antigens not encountered in the thymus. The receptor on T cells required for the co-stimulatory stimulus involved in the prevention of anergy has not been identified. The human T-cell antigen CD28 provides a signal that can synergize with T-cell antigen receptor stimulation in activating T cells to proliferate and secrete lymphokines. Here we report that a monoclonal antibody against the murine homologue of CD28 (ref. 7; J.A.G. et al., manuscript in preparation) can provide a co-stimulatory signal to naive CD4+ T cells and to T-cell clones. Moreover, we demonstrate that this co-stimulatory signal can block the induction of anergy in T-cell clones.
Article
The injection of naked plasmid DNA directly into the muscle cells of mice has been shown to induce potent humoral and cellular immune responses. The generation of a cytotoxic T lymphocyte (CTL) response after plasmid DNA injection may involve the presentation of the expressed antigen in the context of the injected myocytes' endogenous major histocompatibility (MHC)-encoded class I molecules or may use the MHC molecules of bone marrow-derived antigen presenting cells (APC) which are capable of providing co-stimulation as well. To resolve which cell type provides the specific restricting element for this method of vaccination we generated parent-->F1 bone marrow chimeras in which H-2bxd recipient mice received bone marrow that expressed only H-2b or H-2d MHC molecules. These mice were injected intramuscularly with naked plasmid DNA that encoded the nucleoprotein from the A/PR/8/34 influenza strain, which as a single antigen has epitopes for both H-2Db and H-2Kd. The resulting CTL responses were restricted to the MHC haplotype of the bone marrow alone and not to the second haplotype expressed by the recipient's myocytes. The role of somatic tissues that express protein from injected plasmids may be to serve as a reservoir for that antigen which is then transferred to the APC. Consequently, our data show that the mechanism of priming in this novel method for vaccination uses the MHC from bone marrow-derived APC, which are efficient at providing all of the necessary signals for priming the T cell.
Article
Recent data indicate a major role for IL-10 in suppressing immune and inflammatory reactions. To date, expression of human IL-10 has been attributed primarily to helper T lymphocytes, activated monocytes, and neoplastic B cells, and was often found to be associated with IL-6 expression. In this study we sought to determine whether non-hematopoietic human tumor cell lines produce IL-10 and, if so, what is the relationship between IL-10 and IL-6. Using ELISA, we determined IL-10 and IL-6 levels in culture supernatants of 48 cell lines established from carcinomas of the kidney, colon, breast and pancreas, malignant melanomas and neuroblastomas. IL-6 protein was secreted by 28 of the tumor ceil lines; IL-10 was measurable in 15 cell lines. IL-6 secretion was maximal and most frequent in renal-cancer cell lines, while IL-10 production was found to be highest and most common among cell lines derived from colon carcinomas. IL-10 in conditioned medium of one of the colon carcinoma cell lines (CCL222) was bio-active, as demonstrated in the mouse MC/9 mast-cell-line assay and in human mixed-lymphocyte reactions. In both assays, IL-10 bio-activity was neutralized by an anti-IL-10 monoclonal antibody. Expression of IL-6 and IL-10 was confirmed by RNA analysis using message amplification by PCR and sequencing of amplified cDNA. LPS, IL-1 alpha, and TNF-alpha strongly enhanced the release of IL-6 by RCC cells, but only marginally affected IL-10 production in colon-carcinoma cells. IL-10 secretion by colon-carcinoma cells was moderately stimulated by IFN-gamma and IL-4. Dexamethasone suppressed the release of IL-6, but had no inhibitory effect on IL-10 secretion. Our results demonstrate that tumor cell lines established from certain types of human carcinomas are capable of expressing and releasing IL-6 and/or IL-10, suggesting a role of these cytokines in solid-tumor development and anti-tumor immunity. © 1993 Wiley-Liss, Inc.
Article
APO-I is a 48-kDa cell-membrane protein identical to the Fas antigen now designated CD95. It is a member of the NGF/TNF receptor superfamily. Anti-APO-I monoclonal antibody induces apoptosis in a variety of cell types expressing this antigen. We immunohistochemically investigated APO-I expression in normal colon mucosa, 20 adenomas, 258 colon carcinomas and 10 liver metastases and carried out in vitro studies using a panel of colon-carcinoma cell lines. Immunohistochemically, APO-I was regularly expressed at the basolateral membrane of normal colon epithelia. In a minor fraction of colon adenomas and in 39.1% of colon carcinomas APO-I expression was diminished and in 48.1% of carcinomas, predominantly of the non-mucinous type, APO-I expression was completely abrogated. The normal level of APO-I in carcinomas was correlated with the mucinous type. Reduced/lost APO-I expression was more frequent in rectal carcinomas. Complete loss of APO-I was more frequent in tumors that had already metastasized. APO-I expression in liver metastases essentially corresponded to that of the primary tumors. Comparative analysis with data from previous studies revealed that the mode of APO-I expression is correlated with that of HLA-A,B,C./β2m, HLA-DR, HLA-D-associated invariant chain and of the secretory component. Surface expression of APO-I was heterogeneous in colon-carcinoma cell lines; SW480 expressed considerable amounts of APO-I on all cells, while HT-29 constitutively did less so and only in a minority of cells. Surface density of APO-I and the fraction of positive cells in HT-29 was enhanced by interferon-gamma (IFN-γ) and, additively, by tumor necrosis factor-alpha (TNF-α), whereas in SW480 APO-I expression was not modulated by these cytokines. We conclude that neoplastic transformation of colon epithelium often leads to a loss of the physiologic, high level of surface APO-I by giving rise either to a stable lack of APO-I or to an IFN-γ/TNF-α-sensitive phenotype of inducible APO-I expression. © 1994 Wiley-Liss, Inc.
Article
Retroviral-mediated cytokine gene transfer into tumor cells is a highly effective way of inducing tumor inhibition and immunity. We analyzed the tumorigenicity of C-26 murine colon carcinoma cells transduced with genes encoding the two subunits of murine interleukin-12 (IL-12) in a polycistronic retroviral vector and selected for resistance to G418 and for IL-12 production (30–80 pg/ml). BALB/c mice injected s.c., i.v. and intrasplenically with C-26/IL-12 cells from three different IL-12-producing clones showed delayed tumor onset as compared with mice injected with control NeoR-transduced or parental tumor cells. Although C-26/IL-12 tumor-bearing mice eventually died of lung metastasis, their survival time was twice as long as that of mice injected with control cells. In experiments with mice selectively depleted of natural killer (NK) cells before tumor cell injection, the time of tumor onset and survival of mice injected with C-26/IL-12 s.c. and i.v., respectively, was reduced. CD8+ T cell depletion had no effect on latency or survival, whereas removal of CD4+ T cells led to C-26/IL-12 tumor regression in about 40% of mice. Histological and immunocytochemical characterization of leukocytes infiltrating C-26/IL-12 tumors showed only slight infiltration with few T cells in non-depleted mice but abundant infiltration by CD8+ T cells and asialo-GM1+ NK cells in tumors of mice depleted of CD4+ T cells. The lack of CD8+ T cell infiltration is not due to a CD4-mediated suppression of their activation because irradiated C-26/IL-12 cells primed for the induction of a strong cytotoxic T lymphocyte response against C-26 parental cells and induced CD8+ effector cells that protected against C-26/IL-12 in a Winn assay. Rather, the results suggest that, although C-26/IL-12 cells injected in vivo stimulate both NK and CD8+ T cells, tumor infiltration by the latter is inhibited by CD4+ T cells.
Article
PURPOSE: The expression of 70,000-Da heat shock protein (HSP 70) and HLA-DR molecules on cancer cells influences immunologic mechanisms that may be of some prognostic significance. The purpose of this study was to examine the relationship among immunohistochemical HSP 70, HLA-DR expression, and clinicopathologic tumor variables, as well as patient survival in a series of 128 colorectal carcinomas. METHOD: A three-step immunoperoxidase staining technique was undertaken for detection of both markers. RESULTS: Of the examined carcinomas 77.3 percent were HSP 70-positive and 74.2 percent were HLA-DR-positive. Increased HSP 70-positive expression correlated significantly with low differentiation (P0.05), showed a tendency to characterize advanced stages of disease, and was clearly associated with worse overall survival (P0.05). The highest rate of HLA-DR positivity was demonstrated in early stages and was significantly associated with more favorable prognosis (P0.001). HSP 70-positive/HLA-DR-negative patients had worse overall survival compared with the rest (P0.001). CONCLUSIONS: The resulting opposite effects on prognosis of examined markers seem to be related to different pathophysiologic functional roles on tumor immunology.
Article
Tumor-infiltrating lymphocytes (TILs), through displaying a T-cell receptor (TCR) repertoire as heterogeneous as that of normal peripheral blood T cells, show overexpression of TCR variable-gene segments that include clonally expanded TCR sequences. Here, Marialuisa Sensi and Giorgio Parmiani analyze the available information on TCR usage by T cells present in the infiltrate of histologically different tumors and suggest that the analysis of TCR sequences represent a powerful new tool to assess thein vivo immune response to growing tumors. Ultimately, this strategy may lead to the identification and manipulation of T-cell populations endowed with antitumor reactivity.