Polymorphonuclear neutrophils (PMNs) are the most abundant circulating blood leukocytes. They provide the first-line defense against infection and are potent effectors of inflammation. In addition, their release of soluble chemotactic factors guides the recruitment of both nonspecific and specific immune effector cells.1 Finally, since they both respond to and produce cytokines,2,3 they also modulate the balance between humoral and cell-mediated immunity by contributing to the promotion of a TH1 or TH2 response.4 In these ways, PMNs are engaged in a complex cross-talk with immune and endothelial cells that bridges innate and adaptive immunity.5 

Even though many facets of their biology have been thoroughly investigated,6 PMNs still have every reason to complain of the disdain with which they are regarded by oncologists and immunologists.2 So widespread is T-cell chauvinism7 that the antitumor potential of PMNs continues to receive little attention, and researchers have not yet fully considered the possibility of exploiting their functions as effective weapons against cancer.

The attempts of leukocytes to respond to cancer are suggested by their systemic, regional, and intratumoral activation.8-10Infiltration of tumors by leukocytes has been associated with a favorable prognosis in some studies in humans.11-13However, for individual patients, there is no predictable relationship between leukocyte composition and the prognosis of their disease.

PMNs are usually a scarce reactive component of both human and animal tumors. In animal models, their presence may sometimes be detrimental by favoring malignant growth and progression.14Nevertheless, recent studies have suggested that they are active in immunosurveillance against several tumors.15-18 These intriguing outcomes are probably related to the result of the interplay between (1) the kind and amount of cytokines and chemotactic factors naturally released by tumor cells19 and (2) the degree of recruitment and activation of the intermingled PMNs.

Over the last decade, cytokine gene transfer strategies in animal models have provided a tool with which to dramatically increase intratumoral cytokine availability, avoid the side effects of systemic administrations, and evaluate the antineoplastic potential of locally recruited PMNs.

The natural tumor-PMN balance can be markedly altered by engineering tumors to release interleukins20 or chemokines21 in their microenvironment. Although the amount released is usually small, it may be gigantic when compared with wild-type tumors and produce dramatic effects.

Almost all the cytokines sustainedly released by engineered tumor cells, namely interleukin-1 (IL-1), IL-2, IL-3, IL-4, IL-7, IL-10, IL-12, interferon-α (IFN-α), IFN-β, IFN-γ, granulocyte-colony stimulating factor (G-CSF), and tumor necrosis factor-α (TNF-α),22,23 quickly recruit a massive local reaction that leads to the rejection of engineered tumor cells and the establishment of a significant immunity against the wild-type parental tumor. PMNs play a key role in all of these cytokine-induced tumor rejections, often in cooperation with CD8+ T lymphocytes.23 As circulating granulocytes of tumor patients have impaired cytotoxic activity,24 which is further decreased by most chemotherapeutic agents,25 the elaboration of systems capable of guiding the recruitment of PMNs and their activation within a tumor microenvironment can be put forward as a fresh therapeutic approach.

Extravasation from the blood into a tumor is a regulated multistep process involving a series of coordinated interactions between PMNs and endothelial cells.26 Several molecular regulator families, namely selectins, integrins, and cytokines, are thought to control the steps of this process. It is well known that P-, E-, and L-selectin adhesion molecules (also known as GMP 140, ELAM-1, and LAM-1, respectively) initially tether free-flowing neutrophils to the endothelium of postcapillary venules and mediate transient interactions by causing them to roll much more slowly. Slowly progressing PMNs pick up the signals delivered by interleukins, chemokines, and other mediators released by the endothelium and become firmly attached to it via β2 integrin (MAC-1, ie, CD11b/CD18)–intercellular adhesion molecule-1 (ICAM-1) recognition.27 The endothelium is thus the most active controller of leukocyte traffic and behavior through its display of specific signals.

Some proinflammatory mediators or other factors directly secreted by engineered tumor cells, or elicited as downstream mediators by the released cytokine, increase the endothelial expression of several leukocyte adhesion and activation molecules. IL-1β and TNF-α induce and/or up-regulate ELAM-1, P-selectin, ICAM-1, and vascular cell adhesion molecule-1 expression in endothelial cells, whereas IFN-γ mainly promotes ICAM-1 expression.28-31 This cytokine-endothelium cross-talk is thus the first step in regulating PMN intratumoral accumulation (Figure 1).

Fig. 1.

Recruitment of neutrophils into cytokine-transfected tumor and their involvement in its destruction.

Local release of the cytokine from transfected tumor cells acts on the host endothelium and reactive leukocytes, mainly tumor-associated macrophages. The cytokine and chemokine cascade and endothelial adhesion molecule expression thus induced results in PMN recruitment and activation. Tumor destruction on the part of activated PMNs is achieved through their release of a variety of factors (cytokines, enzymes, chlorinated oxidants, etc) whose effects include direct tumor killing, extracellular lysis, inhibition of angiogenesis and activation of other reactive cells, resulting in NK cell, T cell, and antibody-dependent cytotoxicity.

Fig. 1.

Recruitment of neutrophils into cytokine-transfected tumor and their involvement in its destruction.

Local release of the cytokine from transfected tumor cells acts on the host endothelium and reactive leukocytes, mainly tumor-associated macrophages. The cytokine and chemokine cascade and endothelial adhesion molecule expression thus induced results in PMN recruitment and activation. Tumor destruction on the part of activated PMNs is achieved through their release of a variety of factors (cytokines, enzymes, chlorinated oxidants, etc) whose effects include direct tumor killing, extracellular lysis, inhibition of angiogenesis and activation of other reactive cells, resulting in NK cell, T cell, and antibody-dependent cytotoxicity.

Close modal

PMN intratumoral accumulation is also attained when IL-10, a cytokine typically regarded as an anti-inflammatory mediator because it inhibits the release of other interleukins and chemokines,32-34 is present in the microenvironment. We have demonstrated that local release of high levels of IL-10 by IL-10 gene transfected mammary carcinoma cells (TSA–IL-10) in a syngeneic host results in both an anti- and a pro-inflammatory activity, through strong endothelial ELAM-1 expression in peripheral tumor microvessels.35 This expression and subsequent intratumoral accumulation of PMNs were directly attributable to IL-10, since no secondary mediators were detected.35,36 Moreover, it is likely that IL-10 attenuates the constitutive endothelial cell release of nitric oxide37 and contributes to the adhesion of PMNs to microvessels38,39 and their intratumoral accumulation.

It has been recently reported that IL-10 up-regulates the expression of the liver-expressed chemokine (LEC), a human β chemokine also known as NCC-4, HCC-4, and LMC.40 LEC is chemotactic for human monocytes and dendritic cells, but not for neutrophils.41,42 Nevertheless, local accumulation of LEC secreted by engineered TSA cells (TSA-LEC) quickly induces their rejection in association with an impressive recruitment of antigen- presenting cells, lymphocytes, and particularly PMNs.21 

PMNs intratumorally recruited by highly expressed endothelial adhesion molecules also play a key role in mounting a strong antitumor response.21 

Induction of ELAM-1 and up-regulation of ICAM-1 in the blood vessels, even in the case of tumors formed by cells engineered to release G-CSF, IL-2, IL-4, and IL-12, are attributable to both the cytokine-released and downstream-induced secondary mediators, such as CXC chemokines.43,44 Experiments with tumor cells transfected to release G-CSF, IL-2, and IL-12 as well as TNF-α,45-47 have disclosed an evident production of macrophage inflammatory protein-2 (MIP-2), also known as growth-related oncogene/cytokine–induced neutrophil chemoattractant (GRO/KC).48-50 This molecule is the murine functional homologue of human IL-8, which up-regulates the binding affinity of a family of adhesion molecules called integrins on PMNs and of its counter-receptor, ICAM-1, on endothelial cells.51Integrin-mediated adhesion leads to extravasation of PMNs highly attracted to the tumor site by MIP-2, which binds to the PMNs' CXCR1 or CXCR2 counterreceptor (see review in Luster52). MIP-2, together with other factors belonging to the IL-8 granulocyte chemoattractant family, is often released by tumor-associated macrophages under the stimulus of the cytokines released by tumor cells or the secondary mediators they induce.53 We found that macrophages and endothelial cells were clearly stained by anti–MIP-2 antibody when TNF-α as well as IL-1β were present in the tumor microenvironment.44,46 In all these cases, MIP-2 expression was associated with marked recruitment of PMNs, whose accumulation was further enhanced by the further release of MIP-2 produced by PMNs themselves in response to stimulation by TNF-α in the tumor microenvironment.

Recruited PMNs produce several cytotoxic mediators, including reactive oxygen species, proteases, membrane-perforating agents, and soluble mediators of cell killing, such as TNF-α, IL-1β, and IFNs (Figure 1).

Oxidants employ 2 mechanisms to injure tumor cells. They act synergically with protease and other agents, and inactivate plasma antiproteases to allow proteases to operate.54 

Recent dissection of the cytolytic armamentarium of PMNs has suggested a primary role for hypochlorous acid (HOCl) in mediating tumor cell lysis by activated PMNs after their leukocyte function–associated antigen 1–dependent recognition of the target cell surface.55 Furthermore, a distinct adhesion pathway, mediated by CD11b/CD18 up-regulation on activated PMNs, enables these cells to adhere to the vascular endothelium and create a subjacent microenvironment, allowing accumulation of oxidants and proteolytic enzymes at local concentrations sufficient to cause endothelial damage and matrix degradation.56 In addition, PMN-released HOCl reacts with primary amines to form relatively stable chloramines with immunostimulatory properties.57 

Although reactive oxygen and reactive nitrogen intermediates are toxic molecules that contribute to the control of tumors, they also mediate inhibition of T-cell proliferation by suppressing macrophage functions. This mechanism accounts, at least in part, for the immunosuppressed state seen in certain infectious diseases, malignancies, and graft-versus-host reactions (see review in Bogdan et al58).

A further mechanism of PMN-mediated tumor cell killing is antibody-dependent cell-mediated cytotoxicity (ADCC).59The role of antibody-independent recognition of tumor cells by cytotoxic T cells has been extensively researched, whereas only a few recent works have shown that in vivo tumor ADCC also exists.60-63 Granulocyte-macrophage–CSF (GM-CSF) augments the normal PMN ADCC of melanoma, neuroblastoma, and colorectal cancer cells.64-66 

Recent preclinical studies of the treatment of advanced renal cell carcinoma by combining bispecific antibodies (with one specificity against the epidermal growth factor–receptor (EGF-R) overexpressed on the majority of renal cell carcinomas, and another specificity against Fc receptors on human leukocytes) with G-CSF or GM-CSF therapy have demonstrated that granulocytes are the most active effector cell population.67 Although systemic application of bispecific antibodies is suitable at present only for adjuvant treatment of minimal residual disease due to poor tumor cell accessibility, local administration, either alone or in combination with autologous effector cells, is highly effective in eradicating tumor cells.68 

In our murine model, IL-10–releasing TSA cells initially grow and are then rejected by the combined action of CD8+ lymphocytes, natural killer (NK) cells, and PMNs.35,69 As already mentioned, the marked anti-TSA antibody response that follows this rejection may be responsible for PMN-mediated tumor ADCC.

A markedly high titer of anti-TSA antibodies is also elicited during the early phases of LEC-releasing TSA tumor cell rejection. Here, too, antibodies may provide further guidance for the PMN-dependent tumor rejection.21 

A family of antimicrobicidal peptides called defensins has been described in humans.70 Defensins are the most abundant component of the azurophil granules of PMNs and are highly toxic against several types of tumor cells.71 

These granules also contain elastase and cathepsin G, 2 proteases particularly injurious to endothelial cells.72 It is still uncertain whether adhesion of PMNs to tumor cells is required to cause injury. However, the ultrastructural studies performed during the growth and rejection phases of several tumors engineered to release cytokines show PMNs in close contact with injured tumor cells.23,47 An absolute need for adhesion cannot, of course, be inferred from observations of this kind.

Histological and ultrastructural investigation of tumor growth area morphology shows that the damage produced by PMNs takes 2 forms: predominantly colliquative necrosis when their cytotoxicity against tumor cells prevails23 and predominantly ischemic and/or hemorrhagic necrosis when their main target is the vascular endothelium.20,23,35,73,74 

Serial immunohistological examination and polymerase chain reaction analysis after a subcutaneous challenge with TSA–IL-2, TSA–IL-4, TSA–IL-10, TSA–IL-12, and TSA–TNF-α cells have shown that the local release of all these cytokines elicits a quick and effective PMN-mediated antitumor activity.20,44The kinetics, however, are not the same, and the boosted PMN functions are often different.

Presence of pro-inflammatory cytokines

Paracrine release of IL-2, IL-4, IL-12, and TNF-α induced prompt tumor rejection (Figure 2A) marked by the formation of areas of colliquative necrosis typically associated with a massive presence of degranulated PMNs with exocyted granules in close contact with tumor cells displaying ultrastructural signs of irreversible damage (Figure 2B). This indication of direct killing by PMNs was particularly evident in the presence of paracrine IL-2 and TNF-α.

Fig. 2.

PMN-induced tumor destruction.

(A) Rejection area of subcutaneously injected tumor cells engineered to release IL-2 is massively infiltrated by polymorphonuclear leukocytes (× 630). (B) Electron micrograph showing that these are neutrophils at various stages of disorganization and that their exocytosed granules are in close contact with severely damaged or necrotic tumor cells (× 2750).

Fig. 2.

PMN-induced tumor destruction.

(A) Rejection area of subcutaneously injected tumor cells engineered to release IL-2 is massively infiltrated by polymorphonuclear leukocytes (× 630). (B) Electron micrograph showing that these are neutrophils at various stages of disorganization and that their exocytosed granules are in close contact with severely damaged or necrotic tumor cells (× 2750).

Close modal

Interestingly, the observed release of TNF-α into the circulation during infusional recombinant (r) IL-2 therapy of cancer patients is often associated with a potent activation of PMNs that interact with tumor cells and endothelial cells and cause their subsequent lysis.75,76 According to this killing activity, PMNs appear to mediate most of the therapeutic efficacy, but also the systemic toxicity of rIL-2 (ie, vascular leak syndrome).77However, these 2 opposite effects might converge in a single antitumor effect with the development of intratumoral cytokine gene transfer therapy.

In mice, direct killing by PMNs was also a hallmark of the rejection of tumor cells engineered to release G-CSF. Activated PMNs with prominent cytoplasmic projections, in fact, were observed in close contact with dead tumor cells47,78 well before any vascularization of injected tumors was possible. When recipient mice were sublethally irradiated, tumors grew and became vascularized before the rejection that occurred when PMNs and leukocytes were self-reconstituted. In this case, the tumor-associated blood vessels were the main PMN target.78 

Immunohistological analysis showed that in all these situations IL-1β and TNF-α were the predominant downstream pro-inflammatory cytokines elicited in the tumor growth area. They were usually associated with MIP-2 expression in both tumor-associated macrophages and PMNs. Indeed, by thus inducing macrophages to produce PMN chemoattractants, they set up a vicious circle, since they also stimulate PMNs to produce MIP-2 and probably other factors belonging to the IL-8 granulocyte chemoattractant family (Figure 3).

Fig. 3.

Cytokine modulation of PMN intratumoral recruitment and functions.

Cytokine modulation of PMN intratumoral recruitment and functions results in tumor destruction and an antitumor immune memory. Tumor-associated macrophages are the first reactive cells ready to respond to the secondary mediators (IL-1β, TNF-α, IFN-γ, etc) induced by the cytokine systemically administered or locally released by engineered tumor cells. Depending on the cytokines present in the microenvironment, activated macrophages release ELR (glutamic acid–leucine–arginine)+ CXC chemokines, which recruit neutrophils, and/or ELR CXC chemokines, which recruit T lymphocytes. If IL-1β and TNF-α prevail, macrophages and PMNs are induced to produce ELR+ chemokines (MIP-2/GRO/KC), which amplify accumulation of PMNs and favor their destructive functions. If IFN-γ prevails, they are induced to produce angiostatic ELR chemokines (interferon inducible protein-10 [IP-10], monokine induced by gamma interferon [MIG]) that recruit T lymphocytes and promote the establishment of an antitumor immune memory.

Fig. 3.

Cytokine modulation of PMN intratumoral recruitment and functions.

Cytokine modulation of PMN intratumoral recruitment and functions results in tumor destruction and an antitumor immune memory. Tumor-associated macrophages are the first reactive cells ready to respond to the secondary mediators (IL-1β, TNF-α, IFN-γ, etc) induced by the cytokine systemically administered or locally released by engineered tumor cells. Depending on the cytokines present in the microenvironment, activated macrophages release ELR (glutamic acid–leucine–arginine)+ CXC chemokines, which recruit neutrophils, and/or ELR CXC chemokines, which recruit T lymphocytes. If IL-1β and TNF-α prevail, macrophages and PMNs are induced to produce ELR+ chemokines (MIP-2/GRO/KC), which amplify accumulation of PMNs and favor their destructive functions. If IFN-γ prevails, they are induced to produce angiostatic ELR chemokines (interferon inducible protein-10 [IP-10], monokine induced by gamma interferon [MIG]) that recruit T lymphocytes and promote the establishment of an antitumor immune memory.

Close modal

The marked and rapid PMN influx and tumor necrosis observed in nude mice challenged with human tumor cells engineered to release IL-8 (the functional human equivalent of MIP-2), human MIP-1α, and murine MIP-1α79 show that both CXC chemokines, such as IL-8 and GROs, and specific CC chemokines, such as MIP-1α, regulate PMN traffic and functions, providing further evidence that recruited PMNs suppress tumor growth.

Absence of pro-inflammatory cytokines

Rejection of TSA–IL-10 cells, on the other hand, is paradigmatically different.

By suppressing the release of pro-inflammatory cytokines such as IL-1β, TNF-α, and GM-CSF, paracrine IL-10 impairs the early influx of PMNs and permits initial tumor formation by transiently paralyzing a prompt nonspecific antitumor response.35,69 However, by directly acting on endothelial cells, it induces adhesion molecules and attenuates their nitric oxide release, thus favoring leukocyte recruitment in vivo.36,80,81 Leukocytes and mainly neutrophils are responsible for the intratumoral microvasculature damage that results in multiple ischemic necrotic areas.

Local necrosis is followed by massive infiltration by PMNs and a few T and NK cells, probably recruited by mediators induced by hypoxia and necrosis.82 The late influx of reactive cells, however, is enough to lead to complete rejection of the established tumor. This does not take place in the absence of PMNs.69 

The well-known immunosuppressive activity demonstrated by IL-10 in vitro has naturally impeded assessment of its potential use in the treatment of solid human tumors. Its employment has been proposed only for management of the rare myeloproliferative disorder called juvenile myelomonocytic leukemia.83 Even here, consideration has been devoted solely to its ability to inhibit the production of cytokines and growth factors by myelomonocytes in vitro.83Animal data, however, suggest that intratumoral administration of IL-10 inhibits tumor growth through a pro-inflammatory activity in which PMNs again play a fundamental role.35,69 

Presence of IFN-γ

The presence in the tumor growth area of appreciable amounts of IFN-γ together with IL-1β and TNF-α was noted after injection of TSA cells engineered to release IL-2, IL-4, and IL-12.43,44 It was even more evident when local or systemic administration of rIL-12 in mice bearing a subcutaneous 7-day-old TSA tumor resulted in intratumoral expression of the messenger RNA of IL-1β, TNF-α, and IFN-γ, together with IP-10 and MIG,44 2 chemokines with well-known antiangiogenic activities. IL-1β and TNF-α probably induced the production of PMN chemoattractants by tumor-associated macrophages, as the number of tumor-infiltrating PMNs was significantly enhanced after 3 intraperitoneal administrations of rIL-12.

The presence of IFN-γ as secondary mediator, however, must also have induced both macrophages and PMNs to produce IP-10 and MIG84,85 (Figure 3). The rapid influx of PMNs with a high destructive potential and an IP-10– and MIG-mediated anti-angiogenic function resulted in vascular damage, inhibition of angiogenesis, and extensive ischemic/hemorrhagic necrosis after 3 and 8 rIL-12 administrations. Macrophages were undoubtedly essential modulators of this immune response. The crucial importance of PMNs, however, was made clear when their selective depletion abolished the rIL-12–induced antitumor effect. Whether PMNs also have a role in IL-12 therapy of human cancer has still to be determined.

IP-10 and MIG are also intense chemoattractants for monocytes and T cells.86-88 They promote T-cell adhesion to endothelial cells and are leading recruiters of the T cells, particularly CD8+, found to be indispensable, like PMNs, for complete eradication of most of our inocula and experimental tumors.

The presence of IFN-γ and its induction of a cytokine cascade are usually associated with an elevated antitumor memory reaction, since all inocula with IFN-γ in the growth area led to the rejection of a secondary challenge (Figure 3).

PMNs do not seem of great importance in the elaboration of a significant immune memory against the secondary tumor cell challenge. Even so, they are certainly one of the effector arms involved in the destruction of a second inoculum that takes place once this memory is established. This was particularly evident in the case of TSA cells transfected with the IFN-γ gene.20,23 This tumor, in which the PMN infiltrate is almost absent, was rejected in only 25% of cases, though the immune memory established after the first challenge always provided complete protection against a subsequent TSA–parental cell inoculum.

Provided there is IFN-γ in the tumor growth area, the cytokine released by the engineered tumor cells plays a significant role in skewing the memory reaction toward TH1 and TH2. However, secondary rejection is not simply the work of T cells. Massive PMN recruitment, in fact, is consistently evident in both TH1- and TH2-deflected memories. Since their selective removal impairs or even abolishes rejection after establishment of the immune memory,21,69,89 it may well be that PMNs play hitherto unsuspected roles.

PMN-mediated ADCC may significantly contribute to the immune memory leading to secondary tumor cell rejection.59,64,65However, there may be a more complex interplay in which T cells release a guidance factor that directs the powerful destructive action of PMNs. Close contacts between granulocytes, lymphocytes, and tumor cells are typical of immune memory reactions, in keeping with the possibility that cytolytic activity of granulocytes is guided by factors secreted by lymphocytes.

New, interesting perspectives are opening to exploit PMN functions for anticancer strategies in patients.

During therapy with G-CSF, significantly enhanced in vitro cytotoxicity of isolated PMNs against glioblastoma, squamous cell, ovarian, and breast carcinoma cells was observed with sensitizing monoclonal antibody to the oncogene products EGF-R and HER-2/neu.90-92 

Indeed, a phase I study in patients with breast and ovarian cancer showed that biological and clinical activities at well-tolerated doses of bispecific antibodies to FcRI selectively induced on neutrophils and HER-2/neu expressed on tumor target cells.91,92 

In hematologic malignancies, where tumor cells are relatively accessible to antibodies and effector cells, malignant B cells displayed a particularly high susceptibility to neutrophil-mediated ADCC.93 

Phenotypic and functional evidence of potent PMN activation has also been observed during rIL-2 infusion in patients with advanced malignant melanoma and renal cell carcinoma.94,95 Furthermore, patients showing disease response to treatment have a significantly greater production of PMN oxidants, such as HOCl, which is regarded as instrumental in tumor cell lysis.55,77 

Evidence suggests that production of the tumoricidal long-lived oxidant HOCl, along with up-regulation of PMN surface integrins, may also contribute to the antineoplastic efficacy of infusional therapy with TNF-α.96-98 

However, besides their involvement in the therapeutic efficacy of certain cytokines, PMNs may be partly responsible for the toxic side effects of high-dose systemic cytokine therapy.77 

The poor results obtained in humans with systemic cytokine therapy have cast a shadow over this type of approach, especially since its failures were ascribable mainly to multiple side effects and enrollment in phase I trials of patients with advanced disease and a deeply compromised immune system. The extremely encouraging results obtained with local intratumoral cytokine release in animal models, on the other hand, suggest the possibility of successful antitumor management through an improvement in cytokine gene therapy biotechnology and procedures for patients with a reasonable immunological performance and low tumor load or minimal residual disease.

It is clear, in any event, that in addition to being the body's main defenders against infection and foreign invaders, PMNs could be a perfect weapon for the suppression of tumor growth and tumor rejection in T-cell memory reactions, while their ability to respond to and produce cytokines2,3 involves them in the cross-talk between tumor cells and the endothelium and between nonspecific and specific immune cells.5 

A deeper insight into the biological role of immunoregulatory molecules acting as cytokines that stimulate specific PMN functions may thus lead to the elaboration of a new approach to the treatment of cancer.

We thank Dr John Iliffe for critical review of the manuscript.

Supported by grants from the Italian Association for Cancer Research (AIRC); the Istituto Superiore di Sanità Special Programs of Gene Therapy and Antitumor Therapy; C.N.R. Target Project on Biotechnology; MURST 40% and cluster 03—Molecular Engineering—funds, and the US Department of the Army grant DAMD 17-98-1-8030 (G.F.); and by a fellowship from the Italian Foundation for Cancer Research (E.D.C.).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Lillard
JW
Jr
Boyaka
PN
Chertov
O
Oppenheim
JJ
McGhee
JR
Mechanisms for induction of acquired host immunity by neutrophil peptide defensins.
Proc Natl Acad Sci U S A.
96
1999
651
656
2
Lloyd
AR
Oppenheim
JJ
Poly's lament: the neglected role of the polymorphonuclear neutrophil in the afferent limb of the immune response.
Immunol Today.
13
1992
169
172
3
Cassatella
MA
The production of cytokines by polymorphonuclear neutrophils.
Immunol Today.
16
1995
21
26
4
Romani
L
Bistoni
F
Puccetti
P
Initiation of T-helper cell immunity to Candida albicans by IL-12: the role of neutrophils.
Chem Immunol.
68
1997
110
135
5
Colombo
MP
Modesti
A
Parmiani
G
Forni
G
Perspectives in cancer research: local cytokine availability elicits tumor rejection and systemic immunity through granulocyte-T lymphocyte cross-talk.
Cancer Res.
52
1992
1
5
6
Mollinedo
F
Borregaard
N
Boxer
LA
Novel trends in neutrophil structure, function and development.
Immunol Today.
20
1999
535
537
7
Houghton
AN
Lloyd
KO
Stuck in the MUC on the long and winding road.
Nat Med.
4
1998
270
271
8
Topalian
SL
Muul
LM
Solomon
D
Expansion of human tumor infiltrating lymphocytes for use in immunotherapy trials.
J Immunol Methods.
102
1987
127
141
9
Staren
ED
Economou
SG
Harris
JE
Braun
DP
Lymphokine-activated killer cell induction in tumor-infiltrating leukocytes from colon cancer patients.
Cancer.
64
1989
2238
2242
10
Wong
PY
Staren
ED
Tereshkova
N
Braun
DP
Functional analysis of tumor-infiltrating leukocytes in breast cancer patients.
J Surg Res.
76
1998
95
103
11
Hamlin
IME
Possible host resistance in carcinoma of the breast: a histological study.
Br J Cancer.
22
1968
383
401
12
Black
MM
Barclay
THC
Hankey
BF
Prognosis in breast cancer utilizing histologic characteristics of the primary tumor.
Cancer.
36
1975
2048
2055
13
Bassler
R
Dittman
AM
Dittrich
M
Mononuclear stromal reactions in mammary carcinoma, with reference to medullary carcinomas with a lymphoid infiltrate.
Virchows Arch A Pathol Anat Histol.
393
1981
75
91
14
Pekarek
LA
Starr
BA
Toledano
AY
Schreiber
H
Inhibition of tumor growth by elimination of granulocytes.
J Exp Med.
181
1995
435
440
15
Lichtenstein
A
Granulocytes as possible effectors of tumor immunity.
Human Cancer Immunology.
Oettgen
HF
1
1990
731
747
Saunders
Philadelphia, PA
16
Midorikawa
Y
Yamashita
T
Sendo
F
Modulation of the immune response to transplanted tumors in rats by selective depletion of neutrophils in vivo using a monoclonal antibody: abrogation of specific transplantation resistance to chemical carcinogen-induced syngeneic tumors by selective depletion of neutrophils in vivo.
Cancer Res.
50
1990
6243
6247
17
Matsumoto
Y
Saiki
I
Murata
J
Okuyama
H
Tamura
M
Azuma
I
Recombinant human granulocyte colony-stimulating factor inhibits the metastasis of hematogenous and non-hematogenous tumors in mice.
Int J Cancer.
49
1991
444
449
18
Colombo
MP
Ferrari
G
Stoppacciaro
A
et al
Granulocyte colony-stimulating factor gene transfer suppresses tumorigenicity of a murine adenocarcinoma in vivo.
J Exp Med.
173
1991
889
897
19
Nicoletti
G
De Giovanni
C
Lollini
PL
et al
In vivo and in vitro production of hemopoietic colony-stimulating activity by murine cell lines of different origin: a frequent finding.
Eur J Cancer Clin Oncol.
25
1989
1281
1286
20
Musiani
P
Modesti
A
Giovarelli
M
et al
Cytokines, tumour-cell death and immunogenicity: a question of choice.
Immunol Today.
18
1997
32
36
21
Giovarelli
M
Cappello
P
Forni
G
et al
Tumor rejection and immune memory elicited by locally released LEC chemokine are associated with an impressive recruitment of antigen presenting cells, lymphocytes and granulocytes.
J Immunol.
164
2000
3200
3206
22
Modesti
A
Musiani
P
Cortesina
G
et al
Cytokine dependent tumor recognition.
Cytokine. Induced tumor immunogenicity. From exogenous molecules to gene therapy.
Forni
G
Foa
R
Santoni
A
Frati
L
1994
55
68
Academic Press
London, England
23
Musiani
P
Allione
A
Modica
A
et al
Role of neutrophils and lymphocytes in inhibition of a mouse mammary adenocarcinoma engineered to release IL-2, IL-4, IL-7, IL-10, IFN-α, IFN-γ, and TNF-α.
Lab Invest.
74
1996
146
157
24
Hara
N
Ichinose
Y
Asoh
H
Yano
T
Kawasaki
M
Ohta
M
Superoxide anion-generating activity of polymorphonuclear leukocytes and monocytes in patients with lung cancer.
Cancer.
69
1992
1682
1687
25
Hara
N
Ichinose
Y
Motohiro
A
Kuda
T
Aso
H
Ohta
M
Influence of chemotherapeutic agents on superoxide anion production by human polymorphonuclear leukocytes.
Cancer.
66
1990
684
688
26
Springer
T-A
Traffic signals on endothelium for lymphocyte recirculation and leukocyte emigration.
Annu Rev Physiol.
57
1995
827
872
27
Johnston
B
Kubes
P
The α4-integrin: an alternative pathway for neutrophil recruitment?
Immunol Today.
20
1999
545
550
28
Carlos
TM
Harlan
JM
Leukocyte-endothelial adhesion molecules.
Blood.
84
1994
2068
2101
29
Bevilacqua
MP
Stengelin
S
Gimbrone
MA
Seed
B
Endothelial leukocyte adhesion molecule 1: an inducible receptor for neutrophils related to complement regulatory proteins and lectins.
Science.
243
1989
1160
1165
30
Pober
JS
Bevilacqua
MP
Mendrick
DL
Lapierre
LA
Fiers
W
Gimbrone
MA
Two distinct monokines, interleukin 1 and tumor necrosis factor, each independently induce biosynthesis and transient expression of the same antigen on the surface of cultured human vascular endothelial cells.
J Immunol.
136
1986
1680
1687
31
Dustin
ML
Rothlein
R
Bhan
AF
Dinarello
CA
Springer
TA
A natural adherence molecule (ICAM-1): induction by IL-1 and IFN-γ, tissue distribution, biochemistry and function.
J Immunol.
137
1986
245
254
32
De Waal-Malefyt
R
Yssel
H
Roncarolo
MG
Spits
H
de Vries
JE
Interleukin-10.
Curr Opin Immunol.
4
1992
314
320
33
Berkman
N
John
M
Roesems
G
Jose
P
Barnes
P
Chung
K
Inhibition of macrophage inflammatory protein-1 alpha expression by IL-10: differential expression sensitivities in human blood monocytes and alveolar macrophages.
J Immunol.
155
1995
4412
4418
34
Kasama
T
Strieter
R
Lukacs
N
Burdick
M
Kunkel
S
Regulation of neutrophil-derived chemokine expression by IL-10.
J Immunol.
152
1994
3559
3569
35
Di Carlo
E
Coletti
A
Modesti
A
Giovarelli
M
Forni
G
Musiani
P
Local release of interleukin-10 by transfected mouse adenocarcinoma cells exhibits pro- and anti-inflammatory activity and results in a delayed tumor rejection.
Eur Cytokine Netw.
9
1998
61
68
36
Vora
M
Romero
LI
Karasek
MA
Interleukin-10 induces E-selectin on small and large blood vessel endothelial cells.
J Exp Med.
184
1996
821
829
37
Schneemann
M
Schoedon
G
Frei
K
Schaffner
A
Immunovascular communication: activation and deactivation of murine endothelial cell nitric oxide synthase by cytokines.
Immunol Lett.
35
1993
159
162
38
Xin-liang
M
Weyrich
AS
Lefer
DJ
Lefer
AM
Diminished basal nitric oxide release after myocardial ischemia and reperfusion promotes neutrophil adherence to coronary endothelium.
Circ Res.
72
1993
403
412
39
Vinten-Johansen
J
Zhao
ZQ
Nakamura
M
et al
Nitric oxide and the vascular endothelium in myocardial ischemia-reperfusion injury.
Ann N Y Acad Sci.
874
1999
354
370
40
Hedrick
JA
Helms
A
Vicari
A
Zlotnik
A
Characterization of a novel CC chemokine, HCC-4, whose expression is increased by interleukin-10.
Blood.
91
1998
4242
4247
41
Youn
BS
Zhang
S
Broxmeyer
HE
et al
Isolation and characterization of LMC, a novel lymphocyte and monocyte chemoattractant human CC chemokine, with myelosuppressive activity.
Biochem Biophys Res Commun.
247
1998
217
222
42
Nardelli
B
Salcedo
TW
Morahan
DK
Cochrane
EV
Giovarelli
M
Forni
G
Functional characterization of the CC chemokine Ckβ12/HCC-4/LEC [abstract].
FASEB J.
13
1999
314a
43
Di Carlo
E
Modesti
A
Coletti
A
et al
Interaction between endothelial cells and the secreted cytokine drives the fate of an IL4- or an IL5-transduced tumour.
J Pathol.
186
1998
390
397
44
Cavallo
F
Di Carlo
E
Butera
M
et al
Immune events associated with the cure of established tumors and spontaneous metastases by local and systemic interleukin 12.
Cancer Res.
59
1999
414
421
45
Di Carlo
E
Meazza
R
Basso
S
et al
Dissimilar anti-tumour reactions induced by tumour cells engineered with interleukin-2 or interleukin-15 gene in nude mice.
J Pathol.
191
2000
193
201
46
Di Carlo
E
Comes
A
Basso
S
et al
The combined action of IL-15 and IL-12 gene transfer can induce tumor cell rejection without T and NK cell involvement.
J Immunol.
165
2000
3111
3118
47
Colombo
MP
Lombardi
L
Stoppacciaro
A
et al
Granulocyte colony-stimulating factor (G-CSF) gene transduction in murine adenocarcinoma drives neutrophil-mediated tumor inhibition in vivo: neutrophils discriminate between G-CSF-producing and G-CSF-nonproducing tumor cells.
J Immunol.
149
1992
113
119
48
Tekamp-Olson
P
Gallegos
C
Bauer
D
et al
Cloning and characterization of cDNAs for murine macrophage inflammatory protein 2 and its human homologues.
J Exp Med.
172
1990
911
919
49
Zlotnik
A
Morales
J
Hedrick
JA
Recent advances in chemokines and chemokine receptors.
Crit Rev Immunol.
19
1999
1
47
50
Zlotnik
A
Osamu
Y
Chemokines: a new classification system and their role in immunity.
Immunity.
12
2000
121
127
51
Konstantapolous
K
McIntire
LV
Effects of fluid dynamic forces on vascular cell adhesion.
J Clin Invest.
100(suppl 11)
1997
S19
S23
52
Luster
AD
Chemokines: chemotactic cytokines that mediate inflammation.
N Engl J Med.
338
1998
436
445
53
Mantovani
A
The chemokine system: redundancy for robust outputs.
Immunol Today.
20
1999
254
257
54
Jaeschke
H
Wayne Smith
C
Mechanisms of neutrophil-induced parenchymal cell injury.
J Leukoc Biol.
61
1997
647
653
55
Dallegri
F
Ottonello
L
Ballestrero
A
et al
Tumor cell lysis by activated human neutrophils: analysis of neutrophil-delivered oxidative attack and role of leukocyte function-associated antigen 1.
Inflammation.
15
1991
15
30
56
Vedder
NB
Fonty
BW
Winn
RK
Harlan
JM
Rice
CL
Role of neutrophils in generalized reperfusion injury associated with resuscitation from shock.
Surgery.
106
1989
509
516
57
Marcinkiewicz
J
Neutrophil chloramines: missing links between innate and acquired immunity.
Immunol Today.
18
1997
577
580
58
Bogdan
C
Rollinghoff
M
Diefenbach
A
Reactive oxygen and reactive nitrogen intermediates in innate and specific immunity.
Curr Opin Immunol.
12
2000
64
76
59
Kindzelskii
AL
Petty
HR
Early membrane rupture events during neutrophil-mediated antibody-dependent tumor cell cytolysis.
J Immunol.
162
1999
3188
3192
60
Schreiber
GJ
Hellstrom
KE
Hellstrom
I
An unmodified anticarcinoma antibody, BR96, localizes to and inhibits the outgrowth of human tumors in nude mice.
Cancer Res.
52
1992
3262
3266
61
Iliopoulos
D
Ernst
C
Steplewski
A
et al
Inhibition of metastases of a human melanoma xenograft by monoclonal antibody to the GD2/GD3 gangliosides.
J Natl Cancer Inst.
81
1989
440
444
62
Kasprzyk
PG
Song
SU
Di Fiore
PP
King
CR
Therapy of an animal model of human gastric cancer using a combination of anti-erbB-2 monoclonal antibodies.
Cancer Res.
52
1992
2771
2776
63
Heijnen
IAFM
Rijks
LJM
Schiel
A
et al
Generation of HER-2/neu-specific cytotoxic neutrophils in vivo: efficient arming of neutrophils by combined administration of granulocyte colony-stimulating factor and Fcγ receptor I bispecific antibodies.
J Immunol.
159
1997
5629
5639
64
Kushner
BH
Cheung
NV
Absolute requirement of CD11/CD18 adhesion molecules, FcRII, and the phosphatidylinositol-linked FcRIII for monoclonal antibody-mediated neutrophil antihuman tumor cytoxicity.
Blood.
79
1992
1484
1490
65
Kushner
BH
Cheung
HKV
GM-CSF enhances 3F8 monoclonal-dependent cellular cytotoxicity against human melanoma and neuroblastoma.
Blood.
73
1989
1936
1941
66
Ragnhammar
P
Frödin
JE
Trotta
PP
Mellestedt
H
Cytotoxicity of white blood cells activated by granulocyte-colony-stimulating factor, granulocyte-macrophage-colony-stimulating factor and macrophage-colony-stimulating factor against tumor cells in the presence of various monoclonal antibodies.
Cancer Immunol Immunother.
39
1994
254
262
67
Elsässer
D
Stadick
H
Stark
S
et al
Preclinical studies combining bispecific antibodies with cytokine-stimulated effector cells for immunotherapy of renal cell carcinoma.
Anticancer Res.
19
1999
1525
1528
68
Molema
G
Kroesen
BJ
Helfrich
W
Meijer
DK
de Leij
LF
The use of bispecific antibodies in tumor cell and tumor vasculature directed immunotherapy.
J Control Release.
64
2000
229
239
69
Giovarelli
M
Musiani
P
Modesti
A
et al
Local release of IL-10 by transfected mouse mammary adenocarcinoma cells does not suppress but enhances anti-tumor reaction and elicits a strong cytotoxic lymphocyte and antibody-dependent immune memory.
J Immunol.
155
1995
3112
3123
70
Ganz
T
Selsted
ME
Harwing
SSL
Szklarek
D
Daher
K
Lehrer
RI
Defensins: natural peptide antibiotics of human neutrophils.
J Clin Invest.
76
1985
1427
1435
71
Lichtenstein
AK
Ganz
T
Selsted
ME
Lehrer
RI
Synergistic cytolysis mediated by hydrogen peroxide combined with peptide defensins.
Cell Immunol.
114
1988
104
116
72
Okrent
DG
Lichtenstein
AK
Ganz
T
Direct cytotoxicity of polymorphonuclear leukocyte granule proteins to human lung-derived cells and endothelial cells.
Am Rev Respir Dis.
141
1990
179
185
73
Westlin
WF
Gimbrone
MA
Jr
Neutrophil-mediated damage to human vascular endothelium: role of cytokine activation.
Am J Pathol.
142
1993
117
128
74
Bratt
J
Palmblad
J
Cytokine-induced neutrophil-mediated injury of human endothelial cells.
J Immunol.
159
1997
912
918
75
Gemlo
BT
Palladino
MA
Jaffe
HS
Espevik
TP
Rayner
AA
Circulating cytokines in patients with metastatic cancer associated with recombinant interleukin-2 and lymphokine activated killer cells.
Cancer Res.
48
1988
5864
5867
76
Blay
JY
Favrot
MC
Negrier
S
et al
Correlation between clinical response to interleukin-2 therapy and sustained production of tumor necrosis factor.
Cancer Res.
50
1990
2371
2374
77
Carey
PD
Wakefield
CH
Guillou
PJ
Neutrophil activation, vascular leak toxicity, and cytolysis during interleukin-2 infusion in human cancer.
Surgery.
122
1997
918
926
78
Colombo
MP
Lombardi
L
Melani
C
et al
Hypoxic tumor cell death and modulation of endothelial adhesion molecules in the regression of G-CSF transduced tumors.
Am J Pathol.
148
1996
473
483
79
Hirose
K
Hakozaki
M
Nyunoya
Y
et al
Chemokine gene transfection into tumour cells reduced tumorigenicity in nude mice in association with neutrophilic infiltration.
Br J Cancer.
72
1995
708
714
80
Sironi
M
Munoz
C
Pollicino
T
et al
Divergent effects of interleukin-10 on cytokine production by mononuclear phagocytes and endothelial cells.
Eur J Immunol.
23
1993
2692
2695
81
Wogensen
L
Huang
X
Sarvetnick
N
Leukocyte extravasation into the pancreatic tissue in transgenic mice expressing interleukin 10 in the islets of Langerhans.
J Exp Med.
178
1993
175
185
82
Majno
G
Joris
I
Apoptosis, oncosis, and necrosis: an overview of cell death.
Am J Pathol.
146
1995
3
15
83
Iversen
PO
Hart
PH
Bonder
CS
Lopez
AF
Interleukin (IL)-10, but not IL-4 or IL-13, inhibits cytokine production and growth in juvenile myelomonocytic leukemia cells.
Cancer Res.
57
1997
476
480
84
Cassatella
MA
Gasperini
S
Calzetti
F
Bertagnin
A
Luster
AD
McDonald
P-P
Regulated production of the interferon-γ-inducible protein-10 (IP-10) chemokine by human neutrophils.
Eur J Immunol.
27
1997
111
115
85
Gasperini
S
Marchi
M
Calzetti
F
et al
Gene expression and production of the monokine induced by IFN-γ (MIG), IFN-inducible T cell α chemoattractant (I-TAC), and IFN-γ-inducible protein-10 (IP-10) chemokines by human neutrophils.
J Immunol.
162
1999
4928
4937
86
Taub
DD
Lloyd
AR
Conlon
K
Recombinant human interferon-inducible protein 10 is a chemoattractant for human monocytes and T lymphocytes and promotes T cell adhesion to endothelial cells.
J Exp Med.
177
1993
1809
1814
87
Liao
F
Rabin
RL
Yannelli
JR
Koniaris
LG
Vanguri
P
Farber
JM
Human Mig chemokine: biochemical and functional characterization.
J Exp Med.
182
1995
1301
1314
88
Taub
DD
Longo
DL
Murphy
WJ
Human interferon-inducible protein-10 induces mononuclear cell infiltration in mice and promotes the migration of human T lymphocytes into the peripheral tissues and human peripheral blood lymphocytes-SCID mice.
Blood.
87
1996
1423
1431
89
Cavallo
F
Giovarelli
M
Gulino
A
et al
Role of neutrophils and CD4+ T lymphocytes in the primary and memory response to nonimmunogenic murine mammary adenocarcinoma made immunogenic by IL-2 gene.
J Immunol.
149
1992
3627
3635
90
Valerius
T
Repp
R
de Wit
TPM
et al
Involvement of the high affinity receptor for IgG (FcγRI; CD64) in enhanced tumor cell cytotoxicity of neutrophils during G-CSF therapy.
Blood.
82
1993
931
939
91
Valone
FH
Kaufman
PA
Guyre
PM
et al
Phase Ia/Ib trial of bispecific antibody MDX-210 in patients with advanced breast or ovarian cancer that overexpresses the proto-oncogene HER-2/neu.
J Clin Oncol.
13
1995
2281
2292
92
van de Winkel
J-G-J
Bast
B
de Gast
G-C
Immunotherapeutic potential of bispecific antibodies.
Immunol Today.
18
1997
562
564
93
Elsässer
D
Valerius
T
Repp
R
et al
HLA class II as potential target antigen on malignant B cells for therapy with bispecific antibodies in combination with granulocyte colony-stimulating factor.
Blood.
87
1996
3803
3812
94
Rosenberg
SA
Lotze
MR
Yang
JC
et al
Experience with the use of high dose interleukin-2 in the treatment of 652 cancer patients.
Ann Surg.
210
1989
474
485
95
Ghosh
AK
Dazzi
H
Thatcher
N
Moore
M
Lack of correlation between peripheral blood lymphokine-activated killer cell function and clinical response in patients with advanced malignant melanoma receiving recombinant interleukin-2.
Int J Cancer.
43
1989
410
414
96
Test
SJ
Effect of tumour necrosis factor on the generation of chlorinated oxidants by adherent human neutrophils.
J Leukoc Biol.
50
1991
131
139
97
Dallegri
F
Ottonello
L
Neutrophil-mediated cytotoxicity against tumour cells: state of the art.
Arch Immunol Ther Exp (Warsz).
40
1992
39
42
98
She
ZW
Wewers
MD
Herzyk
DJ
Davis
WB
Tumour necrosis factor increases the elastolytic potential of adherent neutrophils: a role for hypochlorous acid.
J Respir Cell Mol Biol.
9
1993
386
392

Author notes

Piero Musiani, G. d' Annunzio University of Chieti, Anatomia Patologica, Osp. SS. Annunziata, Via Valignani, 66100 Chieti, Italy; e-mail: musiani@unich.it.

Sign in via your Institution