You are on page 1of 7

Clin Exp Immunol 1999; 116:388–394

Studies of the mechanism of cytolysis by tumour-infiltrating lymphocytes

M. HISHII, J. T. KURNICK, T. RAMIREZ-MONTAGUT & F. PANDOLFI* Pathology Research Laboratory,


Massachusetts General Hospital, Boston, MA, USA, and *Chair of Semeiotica Medica, Catholic University, Rome, Italy

(Accepted for publication 8 January 1999)

SUMMARY
In order to determine the mechanism of tumour destruction by tumour-infiltrating lymphocytes (TIL),
we examined the ability of both CD4þ and CD8þ effector TIL, and TIL clones, to manifest granzyme-
mediated and Fas-mediated destruction of tumour targets. In many in vitro studies TIL have been shown
to manifest anti-tumour reactivity, yet many tumours escape immunological destruction. To investigate
the role of Fas expression and the concomitant sensitivity to the inducibility of apoptotic death, we
derived TIL from four melanomas and one glioma. The glioma, and all but one of the melanomas,
expressed Fas, but Fas-mediated apoptosis could only be detected if the targets were treated with
cyclohexamide. The melanomas and the glioma all expressed detectable cytoplasmic Bcl-2 protein,
known to exert anti-apoptotic activity. Lysis of tumours by CD8-enriched cultures and CD8þ clones was
Ca2þ-dependent and could not be modified by an anti-Fas MoAb. In CD4-enriched cultures or CD4þ
clones with cytotoxic potential against tumour cells, cytotoxicity was also Ca2þ-dependent. As Ca2þ-
dependent cytotoxicity is usually the result of secretion of perforin/granzyme-B, we investigated the
presence of perforin in cytotoxic CD4þ clones and demonstrated the presence of granular deposits of
this enzyme in some of the CD4þ clones. Although an anti-Fas MoAb did not block the lysis of
melanoma targets by CD4þ clones, the examination of Fas-dependent targets demonstrated that these
clones also had the potential to kill by the Fas/Fas ligand system. These data suggest that the
predominant mechanism in tumour killing by TIL appears to be perforin–granzyme-dependent, and that
the solid tumour cell lines we studied are less susceptible to Fas-mediated apoptosis. As non-apoptotic
pathways may enhance tumour immunogenicity, exploitation of the perforin–granzyme-dependent
cytotoxic T lymphocyte (CTL) pathways may be important for achieving successful anti-tumour
responses.

Keywords tumour-infiltrating lymphocytes cytotoxicity perforin Fas/CD95 FasL Bcl-2


apoptosis

INTRODUCTION particular lytic pathways, may heavily influence the outcome of


the immune response to tumours and our ability to modulate this
Despite numerous investigations, the role of tumour-infiltrating
response. In this study we evaluate the contribution of perforin and
lymphocytes (TIL) in controlling tumour growth remains unclear.
Fas-mediated lysis to the recognition of autologous human tumours
The ability of some TIL cultures to exert strong cytotoxic activity
by the T lymphocytes which infiltrate them.
against autologous tumour cells suggests that such cells should be
Although the literature is replete with articles using murine
able to suppress tumour growth, but the ability of some tumours to
models, including knock-out and transgenic animals, which
progress, even when infiltrated with tumour-specific TIL, suggests
describe the contributions of perforin and Fas-mediated lysis in
that the mechanism of killing may be impaired in vivo. The
the rejection of allografts [2], in the pathogenesis of viral [3,4] and
question of the mechanism of killing manifested in vivo may
autoimmune diseases [5], and in a few instances against leukaemic
have particular importance. Melcher et al. [1] have recently
malignant cells [6,7], the question of which lytic pathways are
demonstrated that apoptotic killing may result in reduced tumour
taken by TIL in the destruction of solid tumours has rarely been
immunogenicity compared with lytic mechanisms which induce
addressed [8]. Thus, although there is much data to suggest that
heat shock protein and produce inflammation. Thus, the question of
tumours may escape from immune recognition by a variety of
how tumours are lysed, and whether there is impairment of
pathways, there is no evaluation of the contribution of the Fas
Correspondence: Professor F. Pandolfi, Clinical Immunology, Viale versus the perforin and granzyme pathways in the lysis of solid
dell’ Universita’ 37, 00185 Rome, Italy. tumours by human TIL. In the light of tumour progression taking
E-mail: pandolfi@axrma.uniroma1.it place even in the presence of a lymphoid infiltrate with

388 q 1999 Blackwell Science


Mechanism of cytotoxicity by TIL 389
tumour-specific cells, a better understanding of these mechanisms [45–48]. A few studies have pointed out, however, that CD4þ
could help explain how tumours escape from immune recognition. cells may also lyse targets via a Ca2þ-dependent mechanism
Since the Fas–FasL mechanism appears to be more sensitive to [49–51]. Importantly, it has been demonstrated that some tumours
activation-induced regulation, we reasoned that down-regulation may be resistant to Fas-mediated killing, by virtue of Bcl-2 expression,
of this mechanism could be involved in the down-regulation of but remain susceptible to perforin-mediated lysis [8,10,11]
cytotoxicity. We therefore investigated Fas expression, and the In this study, we have assessed the ability of TIL to mediate
susceptibility to Fas-mediated apoptosis in several tumour cell tumour destruction by both Ca2þ-dependent and -independent
lines, including the tumour cells’ expression of the anti-apoptotic mechanisms. Although our data demonstrate the ability of many
protein Bcl-2 [8–11]. In addition, we studied the mechanisms of TIL to manifest Fas-mediated target destruction, in fact the tumour
killing involved in both CD8þ and CD4þ effector TIL mediating cells from which they were derived were relatively insusceptible to
anti-tumour cytotoxicity. Ca2þ-independent lysis, indicating that such mechanisms are
Several studies have correlated the presence of TIL with a unlikely to play a major role in tumour destruction in vivo.
relatively better prognosis in melanomas and gliomas [12–15]. In However, both CD8þ TIL, and some CD4þ TIL, were able to
malignant melanoma, it has been shown that TIL are able to manifest strong Ca2þ-dependent lysis of tumour targets, and
recognize tumour-associated antigens, and in some cases to lyse perforin-containing granules were detected in these lytic cells,
the malignant cells [16–30]. These findings provided a basis for the indicating that this is probably the predominant tumour lytic
use of in vitro cultured TIL in adoptive immunotherapy in an effort activity among TIL.
to enhance the in vivo accumulation of tumour-lytic lymphocytes
[29,31]. However, there is considerable doubt concerning the
MATERIALS AND METHODS
efficacy of TIL in vivo, even in cases in which it is possible to
demonstrate the accumulation in vivo of clones capable of exerting Cell lines
strong anti-tumour cytotoxicity in vitro. For example, our observa- Five pairs of autologous tumour cell lines and TIL were derived:
tion of a predominant tumour-specific cytotoxic T lymphocyte four melanoma cell lines (M-1, M-3, M-5 and Mel-EW), one
(CTL) cell clone in multiple metastatic lesions from the same glioma cell line (Gli-PI) and the corresponding TIL. M-1, M-3 and
patient indicated that such cells were able to propagate, circulate, M-5 melanomas and TIL have been previously characterized
home and accumulate within tumour deposits. However, these [52,53]. Mel-EW was derived from a metastatic melanoma patient
tumour-specific CTL clones were found in a patient with progres- and Gli-PI from a glioma. Cultures supplemented with IL-2
sive melanoma, indicating that they were not sufficient to contain demonstrated propagation of activated T cells in those specimens
tumour growth [32]. Thus, the discrepancy between a demon- containing lymphocytic infiltrates. These cultures were kept in IL-2
strated in vitro tumour lytic activity, and the in vivo tumour and restimulated with phytohaemagglutinin (PHA) and irradiated
progression in the presence of this lymphoid infiltrate, remains to feeder cells every 2–4 weeks to enhance proliferation, as pre-
be clarified. viously described [54]. Other cell lines used included Jurkat
Several mechanisms of lymphocyte-mediated cytotoxicity (derived from a T cell leukaemia), U-937 (derived from a promo-
have been described, including specific cytotoxicity mediated by nocytic leukaemia) and K562 (from an erythroleukaemia). These
CTL, as well as natural killer (NK) and lymphokine-associated cell lines were obtained from ATCC (Rockville, MD).
killer (LAK) activities, antibody-dependent cell-mediated cyto-
toxicity, and cytokine-mediated tumour destruction [8,33–35]. Cloning of TIL cultures
Specific cytotoxicity requires T cell receptor (TCR) recognition T cell clones were obtained by limiting dilution as previously
of tumour-associated antigens presented in the context of MHC reported [27,55]. Limiting dilution of IL-2-propagated TIL was
molecules as well as an array of accessory molecules which performed within 4 weeks from original establishment of the
mediate both target binding and the delivery of regulatory culture.
‘second signals’. Current models indicated that CTL-mediated
lysis usually occurs through the release of cytolytic proteins Antibodies
(perforin, granzymes) accumulated in the granules abundantly TIL (bulk cultures and clones) were studied with a panel of MoAbs
present in the cytoplasm of CD8þ cells. Cell lysis then occurs in which included anti-CD3, CD4, CD8 (Ortho Diagnostic System,
a Ca2þ-dependent fashion [36]. Raritan, NJ). MoAbs were used to stain cells in an indirect
More recently, however, it has been shown that CTL can also immunofluorescence test followed by analysis using a Becton
kill targets via a Ca2þ-independent mechanism. This event is Dickinson FACScan (Palo Alto, CA). Fas expression was detected
mediated by the engagement of a surface molecule (Fas or by anti-Fas CH-11 MoAb obtained from UBI (Lake Placid, NY).
CD95) capable of delivering a lethal signal. Cross-linking of Fas, Fas expression was also studied after pre-incubation of tumour
physiologically mediated by cells expressing FasL or artificially cells with interferon-gamma (IFN-g) as previously described in
mediated by the addition of anti-Fas MoAbs or soluble FasL, can detail [53]. FasL expression was detected on TIL activated for 3 h
be shown to induce apoptotic death [37,38]. Cell death via Fas- in culture with PHA, anti-CD3 (purified OKT3 from Ortho, used at
binding is independent of extracellular Ca2þ [39] and does not appropriate dilutions) or phorbol myristate acetate (PMA) and
require active macromolecular synthesis [40,41]. Ca-ionophore (ionomycin, obtained from Sigma (St Louis, MO)
FasL is expressed by activated CD8þ and CD4þ cells, thus and used at 3 mg/ml), with a polyclonal rabbit anti-FasL (N-20)
providing a basis for the assumption that CD8þ cells can kill by serum (Santa Cruz Biotechnology, Santa Cruz, CA), in an indirect
both Ca2þ-dependent and -independent mechanisms. In contrast, immunofluorescence test with an anti-rabbit immunoglobulin FITC
among CD4þ cells, most Th1 cells lack perforins [42–44] and thus serum.
presumably kill primarily through the Fas-based mechanism. Anti-Fas IgM CH-11 antibody was also used to test
CD4þ cells of the Th2 subset have little killing potential susceptibility to Fas-mediated lysis by tumour cell lines at the
q 1999 Blackwell Science Ltd, Clinical and Experimental Immunology, 116:388–394
390 M. Hishii et al.
concentration of 0·5 mg/ml. Anti-Fas MoAb (IgG ZB4; Immuno- RESULTS
tech, Westbrook, ME) was used at a concentration of 1 mg/ml to
Fas and Bcl-2 expression on tumour cell line
inhibit Fas-mediated lysis.
We evaluated Fas expression on eight tumour cell lines. These
Perforin accumulations were detected in the cytoplasm of
included Jurkat (derived from a T cell leukaemia), U-937 (derived
cytocentrifuged smears by immunofluorescence with an anti-
from a promonocytic leukaemia), K562 (from erythroleukaemia),
perforin antibody (obtained from Kamiya Biomedical Co.,
four melanoma cell lines (M-1, M-3, M-5 and Mel-EW) and one
Thousand Oaks, CA), and used at a concentration of 150 mg/ml.
glioma cell line (Gli-PI). Our data confirmed Fas expression on the
The percentage of stained cells was calculated from a minimum of
Jurkat and U937 cell lines, while K562 was negative. In addition,
200 cells counted, with cells scored as positive if they contained
we observed Fas expression on three melanoma cell lines (M-3, M-
distinctly fluorescent granules.
5, Mel-EW) and the Gli-PI glioma cell line, while M-1 cells were
Cytoplasmic staining with Bcl-2 was assessed using anti-Bcl-2
negative. Pre-incubation of the tumour cells with IFN-g had only
(Novacastra Labs, Newcastle upon Tyne, UK) using cells which
insignificant effects on Fas expression in all the cell lines tested
were first fixed for 10 min in 1% paraformaldehyde, followed by
(not shown). All of the melanomas and the glioma cell lines
membrane permeabilization with 0·1% saponin for 10 min prior to
showed detectable cytoplasmic Bcl-2, although levels were gen-
addition of the anti-Bcl-2 antibody for 45 min. Following two
erally low.
washes, FITC-labelled goat anti-mouse IgG was added for 30 min.
All steps were performed at room temperature (228C). Assessment
Fas-mediated apoptosis in tumour cells lines
of staining was made by FACScan flow cytometry (Becton
We then tested the ability of an anti-Fas MoAb (clone CH-11) to
Dickinson).
induce apoptosis of target cells. Tumour cell lines, incubated with
the anti-Fas antibody CH-11, were evaluated for apoptosis using PI
Cytotoxicity
and flow cytometry and by MTT assays. As shown in Table 1, the
Cytotoxic activity was assessed by the lysis of 51Cr-labelled target
MTT assay (data shown are representative of four separate assays)
cells in a 4-h Cr-release assay, as previously reported [56]. Per cent
revealed that Fas ligation was able to induce cell death in Jurkat
specific lysis was calculated as: (experimental ¹spontaneous) ×
(80%) and, to a lesser extent, in U-937 (32%) cells. Of the other
100/(maximum ¹ spontaneous), where all experimental points
cell lines tested, only 19% of the M-3 cell line population was
were calculated as the average of triplicate cultures, and the
killed, while the other cells tested showed very little susceptibility
spontaneous and maximum release by the target cells was the
to cytotoxicity (# 12%). PI staining confirmed that the cell death
average of six wells each. Cytotoxicity was determined at serial
observed in these cultures was due to apoptosis (data not shown).
dilutions of effector cells at ratios of 50:1, 25:1, 12·5:1 and 6·25:1
As it has been shown that Fas-induced apoptosis in certain
to determine the level at which maximum lysis could be achieved
tumour cells may be inhibited by short lived cytoplasmic proteins
for each target/effector pair. As maximum release was achieved
[59], we also evaluated the ability of CH-11 to kill tumour targets
using 25 effectors per target, the data shown are for this ratio only
after pre-incubation with CHX (an inhibitor of protein synthesis).
in order to simplify data presentation. In all cases the ‘spontaneous
Data show that, under these conditions, Fas was able to trigger
release’ (background) was < 15% of the maximum, including
increased apoptotic death in some of the cells lines (Fig. 1). (Data
cultures in which antibodies and cyclohexamide were added.
shown are representative of three separate assays).
Target cells included autologous tumour cell lines, as well as the
series of tumour lines described above. Effector cells were TIL
Mechanisms of killing involved in TIL-mediated cytotoxicity of
maintained in IL-2 (100 U/ml) for between 8 and 20 days after the
tumours. Ca2þ-dependent killing by CD8þ and CD4þ cells
previous restimulation with PHA and irradiated feeder cells. Assays
We first investigated the mechanisms involved in cell-mediated
were performed at multiple dilutions resulting in effector:target
cytotoxicity by CD8þ TIL. We studied a previously described
ratios of 50, 25, 12 and 6:1. As maximal lysis was apparent at ratios
of 25:1, results are reported as percentage of specific lysis at this
ratio. Effects on cytotoxicity by a calcium chelant were measured
after adding 2 mM EGTA (Sigma) to the cultures.
Jurkat
Detection of Fas-induced cell death
After treatment of the tumour cell lines with anti-Fas (CH-11, at
1 mg/ml), apoptosis was evaluated by flow cytometry as previously
described [57] with minor modifications. Briefly, the cells were
centrifuged and resuspended in 0·6 ml of propidium iodide (PI) M-3
hypotonic fluorochrome solution (PI 50 mg/ml in 0·1% sodium
citrate plus 0·1% NP-40; Sigma). The tubes were incubated over-
night at 48C in the dark before the flow cytometric analysis. The
PI fluorescence was measured using an Ortho Absolute flow
M-5
cytometer.
Assessment of cell death was also evaluated by the reduction of
tetrazolium salt, MTT, by living cells to form a blue formazan 0 20 40 60 80 100
product as previously reported [58].
Fas-induced apoptosis was also evaluated after coincubation of Fig. 1. Mortality rate (expressed as percentage of dead cells in the MTT
tumour cells with CH11 and cyclohexamide (CHX; used at a test) of tumour cell lines incubated with anti-Fas MoAb (CH-11) with or
concentration of 1 mg/ml) as previously reported [59]. without cyclohexamide (CHX). A, CH-11; B, CH-11 þ CHX.
q 1999 Blackwell Science Ltd, Clinical and Experimental Immunology, 116:388–394
Mechanism of cytotoxicity by TIL 391
Table 1. Fas and Bcl-2 expression (as determined by flow cytometry), and Table 2. Killing (51Cr-release assay) of autologous tumour by CD8þ
mortality rate (expressed of percentage of dead cells in the MTT test) of tumour-infiltrating lymphocytes (TIL) (bulk culture and three clones)
tumour cell lines incubated with anti-Fas MoAb (CH-11)

Effector cells Percent killing þ EGTA þ anti-Fas (ZB-4)


Cell lines Fas expression Fas-induced mortality Bcl-2 expression
MU-bulk 34 6 6 13 6 4 32 6 6
Jurkat þ 80 6 6 NT* Clone MU-63 32 6 8 15 6 3 38 6 8
U937 – 32 6 3 NT* Clone MU-79 27 6 5 20 6 6 29 6 5
K562 – 261 NT* Clone MU-115 27 6 6 11 6 4 32 6 6
M-1 – 861 þ
M-3 þ 19 6 2 þ
M-5 þ 961 þ
Mel EW þ 12 6 1 þ
Therefore, we evaluated the presence of perforin in the cytoplasm
Gli PI þ 561 þ
of six CD4þ clones (three from the glioma and three from
melanoma patient Mel-EW), by indirect immunofluorescence. In
*These cell lines were not tested (NT) in our assays, but these cell lines three out of six CD4þ clones examined, we observed the presence
have been reported to have minimal, or absent Bcl-2 expression. of scattered perforin staining in the cytoplasm in a granular
distribution (Fig. 2). The positive clones were glioma clones PI-
A and PI-H, and melanoma clone EW-2E5. This clone showed
CD8þ TIL cell line, derived from a melanoma patient, with strong 43% killing of the autologous tumour cells, but killing was not
cytotoxic capacity against autologous tumour cells (M-1 cell line). significantly inhibited by either EGTA or ZB-4. In the three
Some clones derived from this bulk culture were also studied (MU- positive CD4þ clones, perforin staining was strong in only 15–
63, MU-79, MU-115). These CD8þ clones kill the autologous 50% of the cells (scored þ or stronger (out of þ þ þ þ) among 200
tumours through the recognition of the Melan A/MART-1 cells counted per sample). In contrast, in CD8þ cultures the
presented on HLA-A2 (not shown). As expected, killing by the number of strongly perforin-positive cells was > 80% (most
bulk culture and the three derived clones was dependent on þ þ þ or þ þ þ þ). In addition, Giemsa staining revealed the
the presence of Ca2þ, as shown by the inhibition of cytotoxicity presence of the characteristic azurophilic cytoplasmic granules in
in the presence of the Ca2þ chelant EGTA, whereas it could not be the CD8þ cells, while in the CD4þ clones azurophilic granules
blocked by the addition of the anti-Fas MoAb ZB-4 (Table 2). were not detected. Glioma clones PI-D and melanoma clones
(Data shown are representative of three experiments.) EW-2E10 and 3F8 did not show detectable perforin.
The mechanisms involved in killing by CD4þ cells were
investigated in TIL bulk cultures with a CD4þ phenotype and in Fas-mediated killing by CD4 TIL
CD4þ clones derived from both a melanoma and a glioma patient. To determine if the CD4þ effectors derived from tumour lesions
TIL derived from a glioma patient, for which an autologous tumour also had the potential to kill by Fas engagement, we first evaluated
cell line (Gli-PI) was available, were shown to be predominantly the ability of TIL cell lines to express FasL. Bulk cultures,
CD4þ (63%). This TIL cell line showed only marginal lytic
capacity against the autologous tumour (5%, 10% and 15% in
three different experiments). Three CD4þ clones (PI-A, PI-D and
PI-H) derived from the glioma TIL also showed little cytotoxicity
(not shown).
The expression of MHC class II antigens on glioma tumour
cells was undetectable by staining with an anti-DR antibody and
class II expression could not be induced by pretreatment of the
glioma cells with IFN-g (data not shown). As HLA class II
molecules were not expressed on tumour cells, this precluded
efficient recognition of the target by CD4 effectors. Therefore,
we cross-linked effectors and target cells with PHA before per-
forming the cytotoxic assay. Under these conditions, both the bulk
culture and the three clones showed killing of the autologous
tumour. In clones PI-A, -D and -H killing was 57%, 35% and
87%, respectively. Killing was Ca2þ-dependent, as shown by
> 90% inhibition observed with EGTA, while marginal (< 10%)
effects were seen with the addition of the ZB-4 anti-Fas antibody
(representative of three separate assays).

Expression of perforin on CD4þ cells


Ca2þ-dependent killing manifested by CD4þ cells in the experi- Fig. 2. Expression of perforin by tumour-infiltrating lymphocytes (TIL) by
ments with PHA-cross-linked targets suggested that Fas–FasL- indirect immunofluorescence on cytocentrifuged preparations. CD4þ clone
mediated killing of tumour cells may not have been involved in the PI-H (derived from a glioma lesion) showed bright cytoplasmic staining in
mechanism of lysis by these cells. Instead, the Ca2þ-dependent approximately half of the cells; at higher magnification (inset) the cyto-
killing observed is more consistent with perforin-mediated lysis. plasmic stain shows a granular pattern.

q 1999 Blackwell Science Ltd, Clinical and Experimental Immunology, 116:388–394


392 M. Hishii et al.
maintained in IL-2 for 6 or more days after restimulation with PHA observed with both EGTA and anti-Fas (Fig. 4a). Lysis of this
and feeder cells, showed little expression of FasL. However, when target by clone 2E10 could not be blocked by either EGTA or anti-
we tested the same cell lines 3 h after stimulation with either PMA Fas, thus precluding a delineation of the mechanism involved in
and ionomycin, anti-CD3 or PHA (without feeder cells), we this interaction. Killing of the Fas-resistant K562 target by M-3
observed increased FasL expression in all the tested cell lines, bulk could be inhibited only by EGTA, and not by anti-Fas,
suggesting transient expression after stimulation (Fig. 3). confirming that killing of this target occurred in a Ca2þ-dependent,
To assess the ability of CD4þ TIL cells to kill via Fas–FasL Fas-independent fashion.
interactions, we developed a model using a Fas-sensitive target. As
shown in Table 1, among the tumour cell lines derived in our DISCUSSION
laboratory from fresh tumours, only the M-3 targets were suscep-
tible to some Fas-mediated apoptosis (19%). We therefore tested In this study we investigated the cytotoxic mechanisms involved in
the ability of the melanoma-derived TIL CD4þ cell line M-3 and tumour killing by TIL derived from melanoma or glioma patients.
some of the Mel-EW CD4þ clones to induce killing of the Fas- Some TIL cultures or clones (expressing either the CD8þ or the
sensitive target cell lines Jurkat and M-3. The K562 cell line was CD4þ phenotype) were able to exert cytotoxic activity against
used as a Fas-insensitive control. FasL expression on effector cells autologous targets. The CD8þ TIL, and some of the CD4þ TIL
was enhanced by pre-incubation with anti-CD3 antibodies for 3 h showed cytoplasmic perforin-containing granules and lysed their
before performing the cytotoxic assay (representative of duplicate targets in a Ca2þ-dependent fashion. In contrast, our tumour lines
experiments). As shown in Fig. 4, the M3 (Fig. 4a) and Jurkat were generally insensitive to Fas-mediated lysis, although several
targets (Fig. 4b) were lysed in the presence of anti-CD3 antibody of our fresh tumour-derived cell lines expressed Fas. As these
with or without anti-Fas or EDTA (P < 0·05 versus IL-2 alone). In tumours also expressed cytoplasmic Bcl-2, it is possible that their
contrast, K562 cells (Fig. 4c) were not lysed significantly by the insensitivity to Fas-mediated destruction was due to the anti-
EW clones, even in the presence of anti-CD3 antibody and added apoptotic activity of this protein.
anti-Fas and EGTA. The M-3 TIL bulk had anti-K562 activity even It is not surprising that the cytotoxicity manifested by the
in IL-2, which was not enhanced significantly by anti-CD3 or anti- CD8þ TIL was shown to be Ca2þ-dependent. However, most of the
Fas antibody, indicating the Fas-independent lysis of this target.
Cells cultured in IL-2 alone (which expressed low levels of (a) Target: M3 tumour
FasL, see Fig. 3) showed no killing of the M-3 target (Fig. 4a).
With Jurkat (Fig. 4b) and K562 (Fig. 4c) targets, the M-3 TIL bulk Anti-CD3+anti-Fas
Clone EW-2E10 Anti-CD3+EGTA
cultured in IL-2 alone showed some cytotoxic activity. After
Anti-CD3
activation with anti-CD3, M-3 cells and two EW clones (EW- IL-2 alone
2E5 and EW-2E10) could lyse the Jurkat (Fig. 4b) and (to a lesser Clone EW-2E5
degree), the M-3 targets (Fig. 4a). Killing was consistent with the
differential Fas sensitivity of Jurkat and M3 cell lines (Table 1).
M-3 TIL bulk
Killing of Jurkat by M-3 bulk and EW clones was inhibited by
anti-Fas antibody, but EGTA induced little or no inhibition 0 10 20 30 40 50 60
(Fig. 4b). On the other hand, lysis of the M-3 target by M-3 bulk
(b) Target: Jurkat
and clone EW-2E5 appeared to be mediated by both Ca2þ- and
Fas-dependent mechanisms, since some inhibition could be
Clone EW-2E10

80 Clone EW-2E5

IL-2
PMA and M-3 TIL bulk
Percent FasL+ cells

60 ionophore
anti-CD3 0 10 20 30 40 50 60
PHA (c) Target: K-562
40
Clone EW-2E10
20
Clone EW-2E5

0
EW-TIL PI-TIL MU-TIL M-3 TIL bulk
(CD4+) (CD4+) (CD8+)
Fig. 3. FasL expression by tumour-infiltrating lymphocytes (TIL). Sample 0 10 20 30 40 50 60
1 is a CD4þ bulk population derived from a melanoma lesion (EW). Sample Fig. 4. Killing of the Fas-sensitive targets (Jurkat and M-3) and Fas-
2 is a CD4þ bulk population derived from a glioma lesion (PI). Sample 3 is resistant target (K-562) by CD4þ tumour-infiltrating lymphocyte (TIL)
a CD8þ bulk population derived from a melanoma lesion (MU). Cells in IL- effectors kept in IL-2 at least 6 days after restimulation (IL-2 alone), or
2 were stained 6 days after restimulation with phytohaemagglutinin (PHA) preactivated with anti-CD3 for 3 h. Anti-Fas (ZB-4) or EGTA were added
and feeder. For other conditions, cells were stained 3 h after restimulation to these CD3-stimulated cultures to determine the effect of these agents on
with phorbol myristate acetate (PMA)/ionophore, anti-CD3, or PHA. tumour lysis.
q 1999 Blackwell Science Ltd, Clinical and Experimental Immunology, 116:388–394
Mechanism of cytotoxicity by TIL 393
available literature argues that CD4þ cells, which are commonly REFERENCES
devoid of perforin-containing cytoplasmic granules, kill targets 1 Melcher A, Todryk S, Hardwick N, Ford M, Jacobson M, Vile R. Tumor
through a Ca2þ-independent, Fas-mediated mechanism [42–48]. immunogenicity is determined by the mechanism of cell death via
Our data demonstrate that killing by CD4þ bulk cultures and induction of heat shock protein expression. Nature Med 1998; 4:581–7.
clones (derived from both a melanoma and a glioma) could 2 Muller K, Mariani S, Matiba B, Kyewski B, Krammer P. Clonal
occur in a Ca2þ-dependent fashion, and was not affected by anti- deletion of major histocompatibility complex class I-restricted
Fas antibody. In fact, the demonstration of perforin-containing CD4þCD8þ thymocytes in vitro is independent of the CD95 (APO-
granules in a proportion of these CD4þ clones adds a novel model 1/Fas) ligand. Eur J Immunol 1995; 25:2996–9.
for TIL/tumour interaction, in which CD4þ cells kill in a Fas- 3 Lowin B, Hahne M, Mattmann C, Tschopp J. Cytolytic T-cell
independent fashion. cytotoxicity is mediated through perforin and Fas lytic pathways.
Nature 1994; 370:650–2.
Although our data suggest that CD4þ clones do have the
4 Bachmann M, Ohteki T, Faienza K, Zakarian A, Kagi D, Speiser D,
potential to kill by the Fas/FasL mechanisms, it appears that the Ohashi P. Altered peptide ligands trigger perforin- rather than Fas-
autologous tumour targets we tested are not susceptible to this lytic dependent cell lysis. J Immunol 1997; 159:4165–70.
activity. CD4þ TIL can easily express FasL upon activation and 5 Fukuyama H, Adachi M, Suematsu S, Miwa K, Suda T, Yoshida N,
can kill Fas-sensitive targets (such as the Jurkat cell line) by a Nagata S. Transgenic expression of Fas in T cells blocks lymphopro-
mechanism which can be inhibited by anti-Fas antibodies. How- liferation but not autoimmune disease in MRL-lpr mice. J Immunol
ever, this potential activity did not take place in the interactions of 1998; 160:3805–11.
TIL with the autologous tumours we investigated. This conclusion 6 Dilloo D, Brown M, Roskrow M, Zhong W, Holladay M, Holden W,
was further supported by the fact that the melanoma and glioma Brenner M. CD40 ligand induces an antileukemia immune response in
tumour cell lines we derived were also resistant to Fas-mediated vivo. Blood 1997; 90:1927–33.
7 Tsujimura K, Takahashi T, Iwase S, Matsudaira Y, Kaneko Y, Yagita
killing by the apoptosis-inducing antibody, CH-11. This antibody
H, Obata Y. Two types of anti-TL (thymus leukemia) CTL clones with
had activity only when the target cells were treated with CHX. The distinct target specificities: differences in cytotoxic mechanisms and
staining of these tumours with Bcl-2 antibody offers a likely accessory molecule requirements. J Immunol 1998; 160:5253–61.
explanation for this lack of apoptosis induction [11], further 8 Jaattela Benedict M, Tewari M, Shayman J, Dixit V. Bcl-x and Bcl-2
emphasizing the need for Ca2þ-dependent mechanisms of inhibit TNF and Fas-induced apoptosis and activation of phospholipase
tumour lysis to effect tumour destruction. A2 in breast carcinoma cells. Oncogene 1995; 10:2297–305.
In conclusion, our data suggest that the Ca2þ-dependent, 9 Panayiotidis P, Ganeshaguru K, Foroni L, Hoffbrand A. Expression and
perforin-associated mechanism appears to be the principle function of the Fas antigen in B chronic lymphocytic leukemia and
cytotoxic pathway used by TIL-tumour pairs, although we hairy cell leukemia. Leukemia 1995; 9:1227–32.
cannot exclude other lytic mechanisms, including cytokine- 10 Lee R, Spielman J, Podack ER. Bcl-2 protects against Fas-based but not
perforin-based T cell-mediated cytolysis. Int Immunol 1996; 8:991–
induced tumour destruction [34], which could also play a role in
1000.
TIL–tumour interactions. However, it is noteworthy that when 11 Schroter M, Lowin B, Borner C, Tschopp J. Regulation of Fas (Apo-1/
Kuwano et al. blocked cytokine-induced killing, the ability to lyse CD95)- and perforin-mediated lytic pathways of primary cytotoxic T
by a perforin–granzyme mechanism was retained [35]. The lack lymphocytes by the protooncogene bcl-2. Eur J Immunol 1995;
of Fas-dependent lysis in our assays suggests that this is unlikely 25:3509–13.
to play a major role in tumour destruction, but does not preclude 12 Clark WH, Elder DE, Guerry D et al. Model predicting survival in stage
another, perhaps counterproductive role for Fas. For example, I melanoma based on tumor progression. J Natl Cancer Inst 1989;
it has been observed that melanoma cell lines can express FasL, 81:1893–904.
and it has been proposed that they can destroy TIL by this mech- 13 Day C, Lew R, Mihm M et al. A multivariate analysis of prognostic
anism, contributing to the immune privilege of tumours [60,61]. factors for melanoma patients with lesions >3·65 mm in thickness. Ann
Surg 1982; 195:44.
However, when we tested three available melanoma cell lines
14 Mihm M, Clemente C, Cascinelli N. Tumor infiltrating lymphocytes in
(M-1, M-5, Mel-EW), all were FasL¹. Thus, the killing of TIL by lymph node melanoma metastases: a histopathologic prognostic
FasL-expressing tumour cells did not play a role in the tumour– indicator and an expression of local immune response. Lab Invest
TIL interactions we studied, as TIL were not destroyed by contact 1996; 74:43–47.
with tumour cells, and were able to deliver an efficient cytotoxic 15 Ridley A, Cavanagh JB. Lymphocytic infiltration in gliomas: evidence
signal when the TCR was engaged and perforin-mediated lysis of possible host resistance. Brain 1971; 94:117–24.
could occur. With the recent demonstration by Melcher [1] that 16 Mukherji B, Guha A, Chakraborty G, Sivanandham M, Nashed A,
apoptotic killing of tumour cells may render them less immuno- Sporn J, Ergin M. Clonal analysis of cytotoxic and regulatory T cell
genic, it is ironic that the tumour killing we have observed was defi- responses against human melanoma. J Exp Med 1989; 169:1961–76.
cient in Fas-mediated lysis, indicating that any killing which does 17 Itoh K, Platsoucas CD, Balch CM. Autologous tumor-specific cytotoxic
T lymphocytes in the infiltrate of human metastatic melanomas.
occur is more likely to be via conventional cell lysis. The fact that
Activation by interleukin 2 and autologous tumor cells, and
we were able to isolate tumour-specific CTL from these tumours involvement of the T cell receptor. J Exp Med 1988; 168:1419–41.
perhaps reflects the fact that these tumours are indeed, at least 18 Sensi M, Salvi S, Castelli C et al. T cell receptor (TCR) structure of
partially, immunogenic. However, the ultimate failure of these autologous melanoma-reactive cytotoxic T lymphocyte (CTL) clones:
CTL to control tumour growth remains a hurdle to be surmounted. tumor-infiltrating lymphocytes overexpress in vivo the TCR beta chain
sequence used by an HLA-A2-restricted and melanocyte-lineage-
specific CTL clone. J Exp Med 1993; 178:1231–46.
19 Sensi M, Traversari C, Radrizzani M et al. Cytotoxic T-lymphocyte
ACKNOWLEDGMENTS
clones from different patients display limited T-cell-receptor variable-
This work was supported in part by NIH (grants HL-43793 and CA51345), region gene usage in HLA-A2-restricted recognition of the melanoma
NATO (grant CRG940029), AIRC (Milan) and MURST-COFIN (Italy). antigen Melan-A/MART-1. Proc Natl Acad Sci USA 1995; 92:5674–8.

q 1999 Blackwell Science Ltd, Clinical and Experimental Immunology, 116:388–394


394 M. Hishii et al.
20 Kurnick J, Kradin R, Blumberg R, Schneeberger E, Boyle L. Functional 42 Strack P, Martin C, Saito S, Dekruyff R, Ju S. Metabolic inhibitors
characterization of T lymphocytes propagated from human lung distinguish cytolytic activity of CD4 and CD8 clones. Eur J Immunol
carcinomas. Clin Immunol Immunopathol 1986; 38:367–80. 1990; 20:179–83.
21 Platsoucas C. Human autologous tumor-specific T cells in malignant 43 Lancki D, Hsieh C, Fitch F. Mechanisms of lysis by cytotoxic T
melanoma. Cancer Metastasis Rev 1991; 10:151–76. lymphocyte clones. J Immunol 1991; 146:3242–9.
22 Caignard A, Dietrich P, Morand V et al. Evidence for T-cell clonal 44 Ozdemirli M,El-Khatib M, Bastiani L, Akdeniz H, Kuchroo V, Ju S. The
expansion in a patient with squamous cell carcinoma of the head and cytotoxic process of CD4 Th1 clones. J Immunol 1992; 149:1889–95.
neck. Cancer Res 1994; 54:1292–7. 45 Ju ST, Cui H, Panka DJ, Ettinger R, Marshak RA. Participation of target
23 Gervois N, Heuze F, Diez E, Jotereau F. Selective expansion of a Fas protein in apoptosis pathway induced by CD4þ Th1 and CD8þ
specific anti-tumor CD8þ cytotoxic T lymphocyte clone in the bulk cytotoxic T cells. Proc Natl Acad Sci USA 1994; 91:4185–9.
culture of tumor infiltrating lymphocytes from a melanoma patient: 46 Ramsdell F, Seaman MS, Miller RE, Picha KS, Kennedy MK, Lynch
cytotoxic activity and T cell receptor gene rearrangements. Eur J DH. Differential ability of Th1 and Th2 T cells to express Fas ligand
Immunol 1990; 20:825–31. and to undergo activation-induced cell death. Intern Immunol 1994;
24 Ioannides C, Freedman R. Selective usage of TCR V beta in tumor- 6:1545–53.
specific CTL lines isolated from ovarian tumor-associated lymphocytes. 47 Vignaux F, Vivier E, Malissen B, Depraetere V, Nagata S, Golstein P.
Anticancer Res 1991; 11:1919–25. TCR/CD3 coupling to Fas-based cytotoxicity. J Exp Med 1995;
25 Mackensen A, Ferradini L, Carcelain G, Triebel F, Faure F, Viel S, 181:781–6.
Hercend T. Evidence for in situ amplification of cytotoxic T-lympho- 48 Anel A, Buferne M, Boyer C, Schmitt VA, Golstein P. T cell receptor-
cytes with antitumor activity in a human regressive melanoma. Cancer induced Fas ligand expression in cytotoxic T lymphocyte clones is
Res 1993; 53:3569–73. blocked by protein tyrosine kinase inhibitors and cyclosporin A. Eur J
26 Miescher S, Whiteside T, Moretta L, VonFliedner V. Clonal and Immunol 1994; 24:2469–76.
frequency analyses of tumor-infiltrating T lymphocytes from human 49 Grogg D, Hahn S, Erb P. CD4þ T cell mediated killing of major
solid tumors. J Immunol 1987; 138:4004. histocompatibility complex class II-positive antigen-presenting cells.
27 Pandolfi F, Boyle L, Trentin L, Oliva A, Kurnick J. T cell receptor gene III. CD4þ cytotoxic T cells induce apoptosis of APC. J Immunol 1992;
rearrangement and cytotoxic activities of clones isolated from tumor 22:267–79.
infiltrating lymphocytes from melanoma patients. Clin Exp Immunol 50 Miskovsky E, Liu A, Pavlat W et al. Studies of the mechanism of
1994; 95:141–7. cytolysis by HIV-1 specific CD4þ human CTL clones induced by
28 Peoples GE, Schoof DD, Andrews JV, Goedegebuure PS, Eberlein TJ. candidate AIDS vaccines. J Immunol 1994; 153:2787–99.
T-cell recognition of ovarian cancer. Surg 1993; 114:227–34. 51 Susskind B, Shornick M, Iannotti M, Duffy B, nee Tanden P, Siegel J,
29 Rosenberg S, Packard B, Aebersold P. Use of tumor-infiltrating Mohanakumar T. Cytolytic effector mechanisms of human CD4þ
lymphocytes and interleukin 2 in the immunotherapy of patients with cytotoxic T lymphocytes. Human Immunol 1996; 45:64–75.
metastatic melanoma. N Eng J Med 1988; 25:1676–80. 52 Pandolfi F, Boyle LA, Trentin L, Kurnick JT, Isselbacher KJ, Gattoni
30 Wong JT, Pinto CE, Gifford JD, Kurnick JT, Kradin RL. Charac- Celli S. Expression of HLA-A2 antigen in human melanoma cell lines
terization of the CD4þ and CD8þ tumor infiltrating lymphocytes and its role in T-cell recognition. Cancer Res 1991; 51:3164–70.
propagated with bispecific monoclonal antibodies. J Immunol 1989; 53 Pandolfi F, Trentin L, Boyle LA, Stamenkovic I, Byers HR, Colvin RB,
143:3404–11. Kurnick JT. Expression of cell adhesion molecules in human melanoma
31 Kradin R, Kurnick J, Lazarus D et al. Tumour-infiltrating lymphocytes cell lines and their role in lymphocyte mediated cytotoxicity. Cancer
andinterleukin-2in treatment ofadvanced cancer. Lancet 1989;i:577–80. 1992; 69:1165–73.
32 Hishii M, Andrews D, Boyle L, Wong J, Pandolfi F, van den Elsen P, 54 Kurnick J, Kradin R, Blumberg R, Schneeberger E, Boyle L.
Kurnick J. In vivo accumulation of the same anti-melanoma T cell clone Functional characterization of T lymphocytes propagated from
in two different metastatic sites. Proc Natl Acad Sci USA 1997; human lung carcinomas. Clin Immunol Immunopathol 1986; 38:367–
94:1378–83. 80.
33 Berke G. The binding and lysis of target cells by cytotoxic lymphocytes. 55 Kurnick J, Hayward A, Altevogt P. Helper and suppressor/inducer
Annu Rev Immunol 1994; 12:735–73. activity of human T cells and their cloned progeny maintained in long-
34 Hehner S, Hofmann T, Ratter F, Dumont A, Droge W, Schmitz M. term culture. J Immunol 1981; 126:1307.
Tumor necrosis factor-alpha-induced cell killing and activation of 56 Pandolfi F, Strong DM, Slease RB, Smith ML, Ortaldo JR, Herberman
transcription factor NF-kappaB are uncoupled in L929 cells. J Biol RB. Characterization of a suppressor T-cell chronic lymphocytic
Chem 1998; 273:18117–21. leukemia with ADCC but not NK activity. Blood 1980; 56:653–60.
35 Kuwano K, Akashi A, Arai S. An anergic cytotoxic T lymphocyte clone 57 Nicoletti I, Migliorati G, Pagliacci MC, Grignani F, Riccardi C. A rapid
exhibits granule exocytosis-mediated cytotoxicity. Cell Immunol 1998; and simple method for measuring thymocyte apoptosis by propidium
185:114–22. iodide staining and flow cytometry. J Immunol Methods 1991;
36 Henkart P. Lymphocyte-mediated cytotoxicity: two pathways and 139:271–9.
multiple effector molecules. Immunity 1994; 1:343–6. 58 Denizot F, Lang R. Rapid colorimetric assay for cell growth and survival.
37 Suda T, Nagata S. Purification and characterization of the Fas-ligand Modifications to the tetrazolium dye procedure giving improved
that induces apoptosis. J Exp Med 1994; 179:873–9. sensitivity and reliability. J Immunol Methods 1986; 89:271–7.
38 Owen JJ, Jenkinson EJ. Apoptosis and T-cell repertoire selection in the 59 Weller M, Frei K, Groscurth P, Krammer P, Yonekawa Y, Fontana A.
thymus. Ann NY Acad Sci 1992; 663:305–10. Anti-Fas/Apo-1 antibody-mediated apoptosis of cultured human glioma
39 Rouvier E, Luciani MF, Golstein P. Fas involvement in Ca(2þ)- cells. J Clin Invest 1994; 94:954–64.
independent T cell-mediated cytotoxicity. J Exp Med 1993; 177:195– 60 Hahne M, Rimoldi D, Schroter M et al. Melanoma cell expression of
200. Fas (Apo-1/CD95) ligand: implication for tumor escape. Science 1996;
40 Itoh N, Yonehara S, Ishii A et al. The polypeptide encoded by the 274:1363–6.
cDNA for human cell surface antigen Fas can mediate apoptosis. Cell 61 Yamauchi A, Taga K, Mostowski HS, Bloom ET. Target cell-
1991; 66:233–43. induced apoptosis of interleukin-2-activated human natural killer
41 Stalder T, Hahn S, Erb P. Fas antigen is the major target molecule for cells: roles of cell surface molecules and intracellular events. Blood
CD4þ T cell-mediated cytotoxicity. J Immunol 1994; 152:1127–33. 1996; 87:5127–35.

q 1999 Blackwell Science Ltd, Clinical and Experimental Immunology, 116:388–394

You might also like