Tumor-infiltrating CD8+ T cells are strongly associated with patient survival in a wide variety of human cancers. Less is known about tumor-infiltrating CD20+ B cells, which often colocalize with T cells, sometimes forming organized lymphoid structures. In autoimmunity and organ transplantation, T cells and B cells collaborate to generate potent, unrelenting immune responses that can result in extensive tissue damage and organ rejection. In these settings, B cells enhance T cell responses by producing Abs, stimulatory cytokines, and chemokines, serving as local APCs, and organizing the formation of tertiary lymphoid structures that sustain long-term immunity. Thus, B cells are an important component of immunological circuits associated with persistent, rampant tissue destruction. Engagement of tumor-reactive B cells may be an important condition for generating potent, long-term T cell responses against cancer.

The field of tumor immunology is strongly focused on CD8+ T cells, owing to their ability to directly kill tumor cells and the strong association between tumor-infiltrating CD8+ T cells and patient survival in many cancers (1). In contrast, B cells are often overlooked by tumor immunologists, likely because of the common notion that humoral and cytolytic responses work in opposition. Yet, B cells figure prominently in the fields of autoimmunity and tissue transplantation, settings in which T cell responses are so strong and persistent as to override the best attempts at immunosuppression. Given that the immune response to cancer develops over many years and, one hopes, can be manipulated to provide protection on a time scale of decades, there is undoubtedly much to be learned from the chronic immune responses seen in autoimmunity and transplantation. In this study, I will compare B cell responses in cancer, autoimmunity, and transplantation, with the goal of elucidating the mechanisms used by B cells to facilitate long-term T cell responses.

Human B cells develop in the bone marrow and initially have a naive phenotype manifested by unmutated Ig status, expression of IgM and IgD, and a CD27CD38 surface phenotype (2). After activation by Ag, B cells enter primary follicles of lymph nodes or other lymphoid tissues where they undergo extensive proliferation, forming germinal centers (GCs) in which somatic hypermutation and class switching to IgG, IgA, or IgE take place. In the GCs, B cells receive growth and differentiation signals from follicular Th cells and compete for Ags presented by follicular dendritic cells (FDCs) in a process known as affinity maturation. B cells further differentiate into plasma cells (which produce high-affinity Abs) and long-lived CD27+CD38 memory cells (which respond to subsequent Ag encounters). CD20 is expressed on all mature B cells except plasma cells (3).

Although B cells typically reside in conventional lymphoid tissues such as spleen, lymph node, or blood, they can also be found in nonlymphoid tissues in aggregates with other immune cells. Such aggregates have been given various names but will be referred to here as tertiary lymphoid structures (TLSs). TLSs range from small aggregates of B cells, T cells, and DCs through to highly organized structures containing GCs, FDCs, T cell zones, high endothelial venules, and lymphatic vessels (4, 5). TLSs are found at sites of infection or inflammation in essentially any organ of the body and facilitate rapid and robust local immune responses (4, 5). As discussed later, TLSs are also seen in chronic immune responses associated with autoimmunity, allograft rejection, and cancer.

In addition to their important role in immunity to pathogens, B cells contribute to various human autoimmune diseases through Ab-dependent and -independent mechanisms (6, 7). Autoantibodies contribute to autoimmune pathogenesis by inhibiting the function of their target proteins, activating the complement system, augmenting Ag presentation by DCs, or causing end-organ damage through the formation of immune complexes. In addition to making Abs, autoreactive B cells can enhance T cell responses through Ag presentation, costimulation, and modulation of DC migration and function (6, 7). For example, in the MRL/Mp-lpr/lpr model of lupus, mice that are genetically deficient in B cells not only fail to develop autoantibodies but also show greatly attenuated CD4+ and CD8+ T cell activation and reduced lymphocytic infiltration of end organs (8, 9).

Autoreactive B cells are found in TLSs in many autoimmune conditions, including rheumatoid arthritis (RA), Sjögren’s syndrome, multiple sclerosis, autoimmune thyroiditis, diabetes, and lupus nephritis (46). In RA, TLSs are located in synovial tissue and serve as sites of clonal expansion, affinity maturation, and autoantibody production by B cells (7, 1012). In a xenograft model in which affected synovial tissue from patients with RA was implanted in scid mice, depletion of synovial B cells lead to the disappearance of TLSs, reduced activation of T cells, and reduced levels of TNF-α and IFN-γ (13). Similarly, depletion of CD8+ T cells resulted in TLS disintegration and decreased lymphotoxin and Ig secretion (14). Thus, B cells and T cells collaborate to maintain TLS structure and function.

The anti-CD20 mAb rituximab has been used to deplete B cells in various autoimmune conditions (6, 7). In RA, rituximab reduces autoantibodies as expected, yet it also benefits many autoantibody-negative patients by disrupting Ab-independent functions of B cells (7, 15). Specifically, rituximab causes the disappearance of TLSs, with concomitant loss of plasma cells, T cells, macrophages, and FDCs (16). Likewise, in patients with lupus, rituximab treatment results in decreased T cell activation and increased circulating regulatory T cells (Tregs) (15). In idiopathic thrombocytopenic purpura, rituximab can restore a normal CD4+ Th1/Th2 ratio (17). Finally, in multiple sclerosis, rituximab reduces the number of B and T cells in cerebral spinal fluid, with associated clinical benefit (18). Collectively, the use of rituximab in autoimmunity reveals a central role for B cells in the maintenance of pathological T cell responses.

B cells also play a major role in allograft rejection. In a landmark study, renal allograft biopsies from patients undergoing acute rejection (AR) were subjected to gene expression profiling (19). As expected, there was a strong gene signature associated with T cells, NK cells, and macrophages. Unexpectedly, a B cell signature (including CD20, CD74, and Ig) was also prominent. By immunohistochemistry, ∼40% of AR samples showed aggregates containing B cells, CD4+ T cells, CD8+ T cells, and macrophages (20). These aggregates were not associated with Ig or complement deposition, suggesting an Ab-independent role for B cells (19, 20). The presence of CD20+ B cells correlated with glucocorticoid resistance and graft loss. B cell infiltrates have also been described in human liver transplants undergoing AR (21), as well as cardiac transplants (22, 23).

B cells are thought to promote graft rejection by three major mechanisms (24). First, they produce donor-reactive Abs, which can damage tissue via complement and Ab-dependent cytotoxicity (23). Second, B cells produce cytokines and chemokines that can directly damage grafts as well as recruit T cells. Third, B cells can serve as APCs (25). For example, in a mouse cardiac allograft model, B cells contributed to graft rejection by presenting alloantigens to CD4+ T cells (26).

Depletion of CD20+ B cells with rituximab can ameliorate renal allograft rejection (2731). Not only does rituximab reduce CD20+ B cell aggregates in kidney tissue, it reduces expression of T cell-associated gene products such as OX40, Fas ligand, and granzyme B (29, 30). Thus, as in autoimmunity, B cells appear to facilitate pathological T cell responses against tissue allografts.

In contrast to the above findings in autoimmunity and transplantation, initial studies in mouse tumor models suggested that B cells generally inhibit T cell responses. For example, studies comparing wild-type and B cell-deficient mice found that B cells inhibit T cell-mediated regression of established tumors (32, 33), as well as T cell responses to cancer vaccines (3436). B cells can impair the priming of CD8+ CTL responses by CD4+ T cells and instead promote nonprotective humoral immune responses (37). Notably, however, other murine tumor studies have shown positive effects of B cells on T cell responses (38, 39). How can these conflicting results be reconciled? A key factor may be the activation status of B cells in different contexts, as T cell responses appear to be inhibited by resting B cells but facilitated by activated B cells (25, 40). Thus, studies in B cell-deficient mice, which lack both resting and activated B cells, could potentially yield conflicting results depending on the extent of B cell activation in the particular model system. As described below, B cells are commonly activated in human cancer patients, raising the possibility they play a positive role in tumor immunity.

Since the advent of serological cloning methods, it is now recognized that the majority of human cancer patients mount tumor-specific autoantibody responses (41). A wide variety of tumor Ags are recognized, including overexpressed proteins (e.g., HER-2/neu), aberrantly expressed proteins (e.g., cancer-testis Ags), and a plethora of apparently normal self-proteins (41, 42). Furthermore, standard treatments for cancer, such as hormone and radiation therapy, can trigger additional autoantibody responses, presumably through presentation of dying tumor cells to the immune system (43).

Tumor-infiltrating B cells (TIL-Bs) are another important aspect of the B cell response to cancer. TIL-Bs have been studied most extensively in breast cancer, where they are present in ∼25% of tumors and comprise up to 40% of the tumor-infiltrating lymphocyte (TIL) population (42, 44, 45). TIL-Bs are often found in TLSs together with CD4+ and CD8+ T cells and DCs (4648). TIL-Bs appear early during breast tumorigenesis, being present at the ductal carcinoma in situ stage (49). TIL-Bs generally express IgG and show evidence of Ag-driven expansion and somatic mutation consistent with affinity maturation (5055). In one IgG sequencing study, >45% of TIL-B belonged to a clonal group, and there were 4–11 major clonal groups per tumor (46). In node-negative breast cancer, a gene signature indicative of TIL-B was positively associated with survival (56). Likewise, together with CD8+ and CD4+ T cells, TIL-Bs have been implicated in favorable survival rates in medullary breast cancer (42, 57, 58).

CD20+ TIL-Bs are also found in >40% of high-grade serous ovarian cancers, where they are associated with CD4+ and CD8+ T cells, as well as functional T cell markers such as TIA-1, granzyme B, and FoxP3 (59). TIL-Bs are strongly correlated with survival in ovarian cancer (59). Notably, tumors containing both CD8+ and CD20+ TILs are associated with higher survival than tumors containing CD8+ or CD20+ TILs alone, suggesting cooperative interactions between CD8+ and CD20+ TILs.

In non-small cell lung cancer, CD20+, CD8+, and CD4+ TILs are associated with increased survival (60, 61), as are TLS containing B cells, T cells, and mature DCs (62). TLS containing B cells, CD4+ and CD8+ T cells, and DCs are also found in colorectal cancer (63, 64). In cervical cancer, CD20+, CD4+, and CD8+ TILs are associated with a lower relapse rate (65). Finally, TIL-B are prominent in germ cell tumors, where they show evidence of Ag-driven clonal expansion and affinity maturation (66).

Considerable effort has gone into characterizing the target Ags of TIL-B. In a large study of various human cancers, TIL-B–derived autoantibodies were shown to react primarily with autologous tumor targets or allogeneic tumors of the same tissue type, suggesting they recognized tumor-associated Ags (67). In medullary breast cancer, TIL-B–derived autoantibodies were shown to recognize ganglioside D3 and β-actin, the latter by virtue of exposure of β-actin on the surface of apoptotic tumor cells (54, 55, 68). In lung cancer, target Ags of TIL-B include p53, as well as many self-Ags that are overexpressed in tumor tissue (69).

In summary, TIL-Bs are prevalent in human cancer, recognize a wide variety of tumor and self-Ags, associate closely with T cells and other immune cells, and correlate with favorable outcomes. This provides clear rationale to better understand their mechanistic properties.

Although little is known about the mechanisms by which TIL-B promote favorable outcomes in cancer, several possibilities are suggested by studies in autoimmunity, transplantation, and various experimental models (Fig. 1).

FIGURE 1.

Potential mechanisms by which TIL-B promote tumor immunity. Left panel, TIL-B in human ovarian cancer visualized by anti-CD20 immunohistochemistry (brown). Original magnification ×40. Center panel, Effector molecules produced by B cells. Right panel, Potential TIL-B mechanisms involving either direct effects on tumor cells or enhancement of the antitumor activity of T cells and other immune cells.

FIGURE 1.

Potential mechanisms by which TIL-B promote tumor immunity. Left panel, TIL-B in human ovarian cancer visualized by anti-CD20 immunohistochemistry (brown). Original magnification ×40. Center panel, Effector molecules produced by B cells. Right panel, Potential TIL-B mechanisms involving either direct effects on tumor cells or enhancement of the antitumor activity of T cells and other immune cells.

Close modal

TIL-B–derived autoantibodies

As mentioned, autoantibodies and alloreactive Abs play major pathogenic roles in autoimmunity and transplantation, respectively. Similarly, TIL-Bs could potentially mediate their effects through autoantibodies, which could directly modulate the function of target proteins, or promote tumor immunity through the opsonization of tumor Ags, complement-mediated destruction of tumor cells, or Ab-dependent cytotoxicity. For example, in a murine study, adoptively transferred B cells promoted tumor rejection by producing complement-fixing, tumor-reactive Abs (70). Furthermore, the induction of autoantibody responses in mice through vaccination was shown to enhance CD8+ T cell responses against tumors (71). However, in human cancer, the relationship between autoantibodies and clinical outcomes remains controversial. For instance, anti-p53 serum autoantibodies, arguably the best-studied example, have been linked to favorable outcomes in some studies, but not others (72). This inconsistency may be attributable to the generally low concentrations of tumor-reactive autoantibodies in serum (42). Perhaps TIL-Bs, by virtue of location, raise the local concentration of autoantibodies to physiologically significant levels at the tumor site. Consistent with this idea, when human lung cancer specimens were engrafted in scid mice, TIL-B–derived autoantibodies were associated with decreased tumor growth (73, 74).

Direct cytotoxicity by B cells

TIL-Bs might also directly kill tumor cells through Ab-independent mechanisms (75). When stimulated with IL-21, human B cells can secrete granzyme B (76), which could potentially have direct cytotoxicity against tumor cells. Moreover, human B cells stimulated with IFN-α or TLR agonist were shown to kill tumor cells directly via TRAIL signaling (77). Notably, however, cytokines derived from TIL-B can sometimes provide survival signals to tumor cells, as shown for lymphotoxin in murine prostate cancer (78).

Immunoregulation by B cells

Tumors typically contain complex mixtures of immune cells, which, in addition to TIL-Bs, can include cytotoxic T cells, Th cells (e.g., Th1, Th2, Th17), Tregs, DCs, and myeloid-derived suppressor cells. Thus, TIL-Bs could mediate their effects in part by regulating other immune cells. For example, under the influence of CD4+ Th1 and Th2 cells, B cells can be polarized into subsets that produce IFN-γ, IL-12, and TNF-α (Be-1 cells) or IL-2, IL-4, TNF-α, and IL-6 (Be-2 cells) (79, 80). Conversely, Be-1 and Be-2 cells can promote the differentiation of Th1 and Th2 cells, respectively, such that polarized cytokine profiles of both B cells and Th cells are amplified and maintained (79, 80). B cells also facilitate the formation of CD4+ T cell memory (81) and promote the survival and proliferation of activated CD8+ T cells through CD27–CD70 interactions (82). B cells also engage in negative regulatory relationships. For example, regulatory B cells can dampen immune responses through secretion of IL-10 and TGFβ-1 (7). Conversely, Tregs can inhibit B cell activation, proliferation, and Ab production (83).

In a striking example of immunoregulation, B cells can promote the formation of TLSs by secreting lymphotoxin and chemokines, which attract and stimulate T cells, DCs, and other immune cells (7, 82, 84, 85). Accordingly, elimination of B cells in RA or renal allografts results in the disintegration of TLSs with consequent diminution of T cell activity (13, 16, 29, 30). Because TLSs are associated with favorable outcomes in human cancer (62) and murine models (86), this likely represents a key immunoregulatory property of TIL-B.

Ag presentation by B cells

Activated B cells can serve as APCs for both CD4+ and CD8+ T cells (7, 25). Indeed, B cells have an advantage over DCs, as they can selectively present cognate Ag collected through surface Ig molecules, which allows presentation of even low concentrations of Ag. Moreover, B cells can indirectly enhance Ag presentation by other APCs through production of specific Ab (87). The relative contributions of DCs and B cells to Ag presentation in vivo depends on context. In general, it appears DCs are important for initial T cell priming, whereas B cells may promote T cell expansion and memory formation (7, 25, 88).

Why might B cells be required as APCs at the tumor site? T cell activation and expansion are initiated by DCs in draining lymph nodes, yet there is growing recognition from viral models that optimal responses require a second wave of T cell proliferation triggered by APCs at the site of infection (89, 90). This process may be particularly important during prolonged or chronic immune responses (89). Whereas DCs might serve as effective APCs initially, one can imagine that with time, DCs may decline in number or become diverted to a suppressive phenotype, especially in the tumor environment. Although speculative, perhaps TIL-Bs can serve as local APCs under these circumstances, thereby sustaining the survival and proliferation of tumor-infiltrating T cells over the long term. Consistent with this, we found that advanced ovarian cancers lack intratumoral DCs and instead contain TIL-Bs in close association with T cells (59).

The autoimmunity and transplantation fields have exposed B cells as key players in chronic, recalcitrant T cell responses. In addition to direct effects against tissues through Abs or cytotoxic pathways, B cells can promote T cell responses by producing cytokines and chemokines, facilitating TLS formation, and serving as APCs (Fig. 1). The contribution of B cells to tumor immunity might not be evident in many murine tumor models, which tend to involve rapid cytolytic responses akin to acute viral infections. By contrast, in human cancer, the beneficial effects of TILs on clinical outcomes extend over many years, which is more consistent with a chronic immune process. From this perspective, how can we best engage B cells for cancer therapy? The Ab-mediated effects of TIL-Bs can potentially be mimicked by therapeutic mAbs, which represent a rapidly expanding class of cancer drugs. Furthermore, cell-based cancer vaccines can be used to elicit broader Ab responses that closely resemble those associated with naturally occurring TIL-Bs. For example, in melanoma and prostate cancer patients receiving cell-based vaccines expressing GM-CSF, the development of autoantibodies to self-antigens was associated with favorable clinical responses (9193). It is less clear how best to enhance the immunoregulatory and Ag presentation functions of TIL-Bs. Models are needed in which TIL-Bs and T cells collaborate to provide long-term tumor control, as opposed to the models of acute rejection used by most investigators. To this end, TIL populations have been shown to persist for weeks to months in the most current tumor xenograft models (94), providing an experimental system to study functional interactions between TIL-Bs and other cell types. Additional insights will undoubtedly continue to emerge from the autoimmunity and transplantation fields through their continued efforts to disrupt the complex and powerful interactions between B cells and T cells.

I thank the patients, clinicians, and researchers who contributed to the studies reviewed in this paper, Katy Milne for help with Fig. 1, and members of the Deeley Research Centre for helpful discussions.

Disclosures The author has no financial conflicts of interest.

This work was supported by the British Columbia Cancer Foundation.

Abbreviations used in this paper:

AR

acute rejection

DC

dendritic cell

FDC

follicular dendritic cell

GC

germinal center

RA

rheumatoid arthritis

TIL

tumor-infiltrating lymphocyte

TIL-B

tumor-infiltrating B cell

TLS

tertiary lymphoid structure

Treg

regulatory T cell.

1
Pagès
F.
,
Galon
J.
,
Dieu-Nosjean
M. C.
,
Tartour
E.
,
Sautès-Fridman
C.
,
Fridman
W. H.
.
2010
.
Immune infiltration in human tumors: a prognostic factor that should not be ignored.
Oncogene
29
:
1093
1102
.
2
Schmidlin
H.
,
Diehl
S. A.
,
Blom
B.
.
2009
.
New insights into the regulation of human B-cell differentiation.
Trends Immunol.
30
:
277
285
.
3
DiLillo
D. J.
,
Hamaguchi
Y.
,
Ueda
Y.
,
Yang
K.
,
Uchida
J.
,
Haas
K. M.
,
Kelsoe
G.
,
Tedder
T. F.
.
2008
.
Maintenance of long-lived plasma cells and serological memory despite mature and memory B cell depletion during CD20 immunotherapy in mice.
J. Immunol.
180
:
361
371
.
4
Drayton
D. L.
,
Liao
S.
,
Mounzer
R. H.
,
Ruddle
N. H.
.
2006
.
Lymphoid organ development: from ontogeny to neogenesis.
Nat. Immunol.
7
:
344
353
.
5
Carragher
D. M.
,
Rangel-Moreno
J.
,
Randall
T. D.
.
2008
.
Ectopic lymphoid tissues and local immunity.
Semin. Immunol.
20
:
26
42
.
6
Martin
F.
,
Chan
A. C.
.
2006
.
B cell immunobiology in disease: evolving concepts from the clinic.
Annu. Rev. Immunol.
24
:
467
496
.
7
Yanaba
K.
,
Bouaziz
J. D.
,
Matsushita
T.
,
Magro
C. M.
,
St Clair
E. W.
,
Tedder
T. F.
.
2008
.
B-lymphocyte contributions to human autoimmune disease.
Immunol. Rev.
223
:
284
299
.
8
Chan
O.
,
Shlomchik
M. J.
.
1998
.
A new role for B cells in systemic autoimmunity: B cells promote spontaneous T cell activation in MRL-lpr/lpr mice.
J. Immunol.
160
:
51
59
.
9
Chan
O. T.
,
Madaio
M. P.
,
Shlomchik
M. J.
.
1999
.
B cells are required for lupus nephritis in the polygenic, Fas-intact MRL model of systemic autoimmunity.
J. Immunol.
163
:
3592
3596
.
10
Gause
A.
,
Gundlach
K.
,
Zdichavsky
M.
,
Jacobs
G.
,
Koch
B.
,
Hopf
T.
,
Pfreundschuh
M.
.
1995
.
The B lymphocyte in rheumatoid arthritis: analysis of rearranged V kappa genes from B cells infiltrating the synovial membrane.
Eur. J. Immunol.
25
:
2775
2782
.
11
Itoh
K.
,
Patki
V.
,
Furie
R. A.
,
Chartash
E. K.
,
Jain
R. I.
,
Lane
L.
,
Asnis
S. E.
,
Chiorazzi
N.
.
2000
.
Clonal expansion is a characteristic feature of the B-cell repetoire of patients with rheumatoid arthritis.
Arthritis Res.
2
:
50
58
.
12
Humby
F.
,
Bombardieri
M.
,
Manzo
A.
,
Kelly
S.
,
Blades
M. C.
,
Kirkham
B.
,
Spencer
J.
,
Pitzalis
C.
.
2009
.
Ectopic lymphoid structures support ongoing production of class-switched autoantibodies in rheumatoid synovium.
PLoS Med.
6
:
e1
.
13
Takemura
S.
,
Klimiuk
P. A.
,
Braun
A.
,
Goronzy
J. J.
,
Weyand
C. M.
.
2001
.
T cell activation in rheumatoid synovium is B cell dependent.
J. Immunol.
167
:
4710
4718
.
14
Kang
Y. M.
,
Zhang
X.
,
Wagner
U. G.
,
Yang
H.
,
Beckenbaugh
R. D.
,
Kurtin
P. J.
,
Goronzy
J. J.
,
Weyand
C. M.
.
2002
.
CD8 T cells are required for the formation of ectopic germinal centers in rheumatoid synovitis.
J. Exp. Med.
195
:
1325
1336
.
15
Liossis
S. N.
,
Sfikakis
P. P.
.
2008
.
Rituximab-induced B cell depletion in autoimmune diseases: potential effects on T cells.
Clin. Immunol.
127
:
280
285
.
16
Thurlings
R. M.
,
Vos
K.
,
Wijbrandts
C. A.
,
Zwinderman
A. H.
,
Gerlag
D. M.
,
Tak
P. P.
.
2008
.
Synovial tissue response to rituximab: mechanism of action and identification of biomarkers of response.
Ann. Rheum. Dis.
67
:
917
925
.
17
Stasi
R.
,
Del Poeta
G.
,
Stipa
E.
,
Evangelista
M. L.
,
Trawinska
M. M.
,
Cooper
N.
,
Amadori
S.
.
2007
.
Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura.
Blood
110
:
2924
2930
.
18
Cross
A. H.
,
Stark
J. L.
,
Lauber
J.
,
Ramsbottom
M. J.
,
Lyons
J. A.
.
2006
.
Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients.
J. Neuroimmunol.
180
:
63
70
.
19
Sarwal
M.
,
Chua
M. S.
,
Kambham
N.
,
Hsieh
S. C.
,
Satterwhite
T.
,
Masek
M.
,
Salvatierra
O.
 Jr
.
2003
.
Molecular heterogeneity in acute renal allograft rejection identified by DNA microarray profiling.
N. Engl. J. Med.
349
:
125
138
.
20
Zarkhin
V.
,
Kambham
N.
,
Li
L.
,
Kwok
S.
,
Hsieh
S. C.
,
Salvatierra
O.
,
Sarwal
M. M.
.
2008
.
Characterization of intra-graft B cells during renal allograft rejection.
Kidney Int.
74
:
664
673
.
21
Krukemeyer
M. G.
,
Moeller
J.
,
Morawietz
L.
,
Rudolph
B.
,
Neumann
U.
,
Theruvath
T.
,
Neuhaus
P.
,
Krenn
V.
.
2004
.
Description of B lymphocytes and plasma cells, complement, and chemokines/receptors in acute liver allograft rejection.
Transplantation
78
:
65
70
.
22
Michaels
P. J.
,
Kobashigawa
J.
,
Laks
H.
,
Azarbal
A.
,
Espejo
M. L.
,
Chen
L.
,
Fishbein
M. C.
.
2001
.
Differential expression of RANTES chemokine, TGF-beta, and leukocyte phenotype in acute cellular rejection and quilty B lesions.
J. Heart Lung Transplant.
20
:
407
416
.
23
Thaunat
O.
,
Field
A. C.
,
Dai
J.
,
Louedec
L.
,
Patey
N.
,
Bloch
M. F.
,
Mandet
C.
,
Belair
M. F.
,
Bruneval
P.
,
Meilhac
O.
, et al
.
2005
.
Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response.
Proc. Natl. Acad. Sci. USA
102
:
14723
14728
.
24
Zarkhin
V.
,
Li
L.
,
Sarwal
M.
.
2008
.
“To B or not to B?” B-cells and graft rejection.
Transplantation
85
:
1705
1714
.
25
Rodríguez-Pinto
D.
2005
.
B cells as antigen presenting cells.
Cell. Immunol.
238
:
67
75
.
26
Noorchashm
H.
,
Reed
A. J.
,
Rostami
S. Y.
,
Mozaffari
R.
,
Zekavat
G.
,
Koeberlein
B.
,
Caton
A. J.
,
Naji
A.
.
2006
.
B cell-mediated antigen presentation is required for the pathogenesis of acute cardiac allograft rejection.
J. Immunol.
177
:
7715
7722
.
27
Becker
Y. T.
,
Becker
B. N.
,
Pirsch
J. D.
,
Sollinger
H. W.
.
2004
.
Rituximab as treatment for refractory kidney transplant rejection.
Am. J. Transplant.
4
:
996
1001
.
28
Alausa
M.
,
Almagro
U.
,
Siddiqi
N.
,
Zuiderweg
R.
,
Medipalli
R.
,
Hariharan
S.
.
2005
.
Refractory acute kidney transplant rejection with CD20 graft infiltrates and successful therapy with rituximab.
Clin. Transplant.
19
:
137
140
.
29
Genberg
H.
,
Hansson
A.
,
Wernerson
A.
,
Wennberg
L.
,
Tydén
G.
.
2006
.
Pharmacodynamics of rituximab in kidney allotransplantation.
Am. J. Transplant.
6
:
2418
2428
.
30
Lehnhardt
A.
,
Mengel
M.
,
Pape
L.
,
Ehrich
J. H.
,
Offner
G.
,
Strehlau
J.
.
2006
.
Nodular B-cell aggregates associated with treatment refractory renal transplant rejection resolved by rituximab.
Am. J. Transplant.
6
:
847
851
.
31
Zarkhin
V.
,
Li
L.
,
Kambham
N.
,
Sigdel
T.
,
Salvatierra
O.
,
Sarwal
M. M.
.
2008
.
A randomized, prospective trial of rituximab for acute rejection in pediatric renal transplantation.
Am. J. Transplant.
8
:
2607
2617
.
32
Shah
S.
,
Divekar
A. A.
,
Hilchey
S. P.
,
Cho
H. M.
,
Newman
C. L.
,
Shin
S. U.
,
Nechustan
H.
,
Challita-Eid
P. M.
,
Segal
B. M.
,
Yi
K. H.
,
Rosenblatt
J. D.
.
2005
.
Increased rejection of primary tumors in mice lacking B cells: inhibition of anti-tumor CTL and TH1 cytokine responses by B cells.
Int. J. Cancer
117
:
574
586
.
33
Barbera-Guillem
E.
,
Nelson
M. B.
,
Barr
B.
,
Nyhus
J. K.
,
May
K. F.
 Jr.
,
Feng
L.
,
Sampsel
J. W.
.
2000
.
B lymphocyte pathology in human colorectal cancer. Experimental and clinical therapeutic effects of partial B cell depletion.
Cancer Immunol. Immunother.
48
:
541
549
.
34
Oizumi
S.
,
Deyev
V.
,
Yamazaki
K.
,
Schreiber
T.
,
Strbo
N.
,
Rosenblatt
J.
,
Podack
E. R.
.
2008
.
Surmounting tumor-induced immune suppression by frequent vaccination or immunization in the absence of B cells.
J. Immunother.
31
:
394
401
.
35
Perricone
M. A.
,
Smith
K. A.
,
Claussen
K. A.
,
Plog
M. S.
,
Hempel
D. M.
,
Roberts
B. L.
,
St George
J. A.
,
Kaplan
J. M.
.
2004
.
Enhanced efficacy of melanoma vaccines in the absence of B lymphocytes.
J. Immunother.
27
:
273
281
.
36
Kim
S.
,
Fridlender
Z. G.
,
Dunn
R.
,
Kehry
M. R.
,
Kapoor
V.
,
Blouin
A.
,
Kaiser
L. R.
,
Albelda
S. M.
.
2008
.
B-cell depletion using an anti-CD20 antibody augments antitumor immune responses and immunotherapy in nonhematopoetic murine tumor models.
J. Immunother.
31
:
446
457
.
37
Qin
Z.
,
Richter
G.
,
Schüler
T.
,
Ibe
S.
,
Cao
X.
,
Blankenstein
T.
.
1998
.
B cells inhibit induction of T cell-dependent tumor immunity.
Nat. Med.
4
:
627
630
.
38
Schultz
K. R.
,
Klarnet
J. P.
,
Gieni
R. S.
,
HayGlass
K. T.
,
Greenberg
P. D.
.
1990
.
The role of B cells for in vivo T cell responses to a Friend virus-induced leukemia.
Science
249
:
921
923
.
39
DiLillo
D. J.
,
Yanaba
K.
,
Tedder
T. F.
.
2010
.
B cells are required for optimal CD4+ and CD8+ T cell tumor immunity: therapeutic B cell depletion enhances B16 melanoma growth in mice.
J. Immunol.
184
:
4006
4016
.
40
Watt
V.
,
Ronchese
F.
,
Ritchie
D.
.
2007
.
Resting B cells suppress tumor immunity via an MHC class-II dependent mechanism.
J. Immunother.
30
:
323
332
.
41
Reuschenbach
M.
,
von Knebel Doeberitz
M.
,
Wentzensen
N.
.
2009
.
A systematic review of humoral immune responses against tumor antigens.
Cancer Immunol. Immunother.
58
:
1535
1544
.
42
Coronella-Wood
J. A.
,
Hersh
E. M.
.
2003
.
Naturally occurring B-cell responses to breast cancer.
Cancer Immunol. Immunother.
52
:
715
738
.
43
Nesslinger
N. J.
,
Sahota
R. A.
,
Stone
B.
,
Johnson
K.
,
Chima
N.
,
King
C.
,
Rasmussen
D.
,
Bishop
D.
,
Rennie
P. S.
,
Gleave
M.
, et al
.
2007
.
Standard treatments induce antigen-specific immune responses in prostate cancer.
Clin. Cancer Res.
13
:
1493
1502
.
44
Chin
Y.
,
Janseens
J.
,
Vandepitte
J.
,
Vandenbrande
J.
,
Opdebeek
L.
,
Raus
J.
.
1992
.
Phenotypic analysis of tumor-infiltrating lymphocytes from human breast cancer.
Anticancer Res.
12
:
1463
1466
.
45
Marsigliante
S.
,
Biscozzo
L.
,
Marra
A.
,
Nicolardi
G.
,
Leo
G.
,
Lobreglio
G. B.
,
Storelli
C.
.
1999
.
Computerised counting of tumour infiltrating lymphocytes in 90 breast cancer specimens.
Cancer Lett.
139
:
33
41
.
46
Coronella
J. A.
,
Spier
C.
,
Welch
M.
,
Trevor
K. T.
,
Stopeck
A. T.
,
Villar
H.
,
Hersh
E. M.
.
2002
.
Antigen-driven oligoclonal expansion of tumor-infiltrating B cells in infiltrating ductal carcinoma of the breast.
J. Immunol.
169
:
1829
1836
.
47
Yakirevich
E.
,
Izhak
O. B.
,
Rennert
G.
,
Kovacs
Z. G.
,
Resnick
M. B.
.
1999
.
Cytotoxic phenotype of tumor infiltrating lymphocytes in medullary carcinoma of the breast.
Mod. Pathol.
12
:
1050
1056
.
48
Tamiolakis
D.
,
Simopoulos
C.
,
Cheva
A.
,
Lambropoulou
M.
,
Kotini
A.
,
Jivannakis
T.
,
Papadopoulos
N.
.
2002
.
Immunophenotypic profile of tumor infiltrating lymphocytes in medullary carcinoma of the breast.
Eur. J. Gynaecol. Oncol.
23
:
433
436
.
49
Lee
A. H.
,
Happerfield
L. C.
,
Bobrow
L. G.
,
Millis
R. R.
.
1997
.
Angiogenesis and inflammation in ductal carcinoma in situ of the breast.
J. Pathol.
181
:
200
206
.
50
Wang
Y.
,
Ylera
F.
,
Boston
M.
,
Kang
S. G.
,
Kutok
J. L.
,
Klein-Szanto
A. J.
,
Junghans
R. P.
.
2007
.
Focused antibody response in plasma cell-infiltrated non-medullary (NOS) breast cancers.
Breast Cancer Res. Treat.
104
:
129
144
.
51
Simsa
P.
,
Teillaud
J. L.
,
Stott
D. I.
,
Tóth
J.
,
Kotlan
B.
.
2005
.
Tumor-infiltrating B cell immunoglobulin variable region gene usage in invasive ductal breast carcinoma.
Pathol. Oncol. Res.
11
:
92
97
.
52
Nzula
S.
,
Going
J. J.
,
Stott
D. I.
.
2003
.
Antigen-driven clonal proliferation, somatic hypermutation, and selection of B lymphocytes infiltrating human ductal breast carcinomas.
Cancer Res.
63
:
3275
3280
.
53
Coronella
J. A.
,
Telleman
P.
,
Kingsbury
G. A.
,
Truong
T. D.
,
Hays
S.
,
Junghans
R. P.
.
2001
.
Evidence for an antigen-driven humoral immune response in medullary ductal breast cancer.
Cancer Res.
61
:
7889
7899
.
54
Hansen
M. H.
,
Nielsen
H. V.
,
Ditzel
H. J.
.
2002
.
Translocation of an intracellular antigen to the surface of medullary breast cancer cells early in apoptosis allows for an antigen-driven antibody response elicited by tumor-infiltrating B cells.
J. Immunol.
169
:
2701
2711
.
55
Hansen
M. H.
,
Nielsen
H.
,
Ditzel
H. J.
.
2001
.
The tumor-infiltrating B cell response in medullary breast cancer is oligoclonal and directed against the autoantigen actin exposed on the surface of apoptotic cancer cells.
Proc. Natl. Acad. Sci. USA
98
:
12659
12664
.
56
Schmidt
M.
,
Böhm
D.
,
von Törne
C.
,
Steiner
E.
,
Puhl
A.
,
Pilch
H.
,
Lehr
H. A.
,
Hengstler
J. G.
,
Kölbl
H.
,
Gehrmann
M.
.
2008
.
The humoral immune system has a key prognostic impact in node-negative breast cancer.
Cancer Res.
68
:
5405
5413
.
57
Ridolfi
R. L.
,
Rosen
P. P.
,
Port
A.
,
Kinne
D.
,
Miké
V.
.
1977
.
Medullary carcinoma of the breast: a clinicopathologic study with 10 year follow-up.
Cancer
40
:
1365
1385
.
58
Lim
K. H.
,
Telisinghe
P. U.
,
Abdullah
M. S.
,
Ramasamy
R.
.
2010
.
Possible significance of differences in proportions of cytotoxic T cells and B-lineage cells in the tumour-infiltrating lymphocytes of typical and atypical medullary carcinomas of the breast.
Cancer Immun.
10
:
3
.
59
Milne
K.
,
Köbel
M.
,
Kalloger
S. E.
,
Barnes
R. O.
,
Gao
D.
,
Gilks
C. B.
,
Watson
P. H.
,
Nelson
B. H.
.
2009
.
Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors.
PLoS ONE
4
:
e6412
.
60
Al-Shibli
K. I.
,
Donnem
T.
,
Al-Saad
S.
,
Persson
M.
,
Bremnes
R. M.
,
Busund
L. T.
.
2008
.
Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer.
Clin. Cancer Res.
14
:
5220
5227
.
61
Riemann
D.
,
Wenzel
K.
,
Schulz
T.
,
Hofmann
S.
,
Neef
H.
,
Lautenschläger
C.
,
Langner
J.
.
1997
.
Phenotypic analysis of T lymphocytes isolated from non-small-cell lung cancer.
Int. Arch. Allergy Immunol.
114
:
38
45
.
62
Dieu-Nosjean
M. C.
,
Antoine
M.
,
Danel
C.
,
Heudes
D.
,
Wislez
M.
,
Poulot
V.
,
Rabbe
N.
,
Laurans
L.
,
Tartour
E.
,
de Chaisemartin
L.
, et al
.
2008
.
Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures.
J. Clin. Oncol.
26
:
4410
4417
.
63
Suzuki
A.
,
Masuda
A.
,
Nagata
H.
,
Kameoka
S.
,
Kikawada
Y.
,
Yamakawa
M.
,
Kasajima
T.
.
2002
.
Mature dendritic cells make clusters with T cells in the invasive margin of colorectal carcinoma.
J. Pathol.
196
:
37
43
.
64
Jackson
P. A.
,
Green
M. A.
,
Marks
C. G.
,
King
R. J.
,
Hubbard
R.
,
Cook
M. G.
.
1996
.
Lymphocyte subset infiltration patterns and HLA antigen status in colorectal carcinomas and adenomas.
Gut
38
:
85
89
.
65
Nedergaard
B. S.
,
Ladekarl
M.
,
Nyengaard
J. R.
,
Nielsen
K.
.
2008
.
A comparative study of the cellular immune response in patients with stage IB cervical squamous cell carcinoma. Low numbers of several immune cell subtypes are strongly associated with relapse of disease within 5 years.
Gynecol. Oncol.
108
:
106
111
.
66
Willis
S. N.
,
Mallozzi
S. S.
,
Rodig
S. J.
,
Cronk
K. M.
,
McArdel
S. L.
,
Caron
T.
,
Pinkus
G. S.
,
Lovato
L.
,
Shampain
K. L.
,
Anderson
D. E.
, et al
.
2009
.
The microenvironment of germ cell tumors harbors a prominent antigen-driven humoral response.
J. Immunol.
182
:
3310
3317
.
67
Punt
C. J.
,
Barbuto
J. A.
,
Zhang
H.
,
Grimes
W. J.
,
Hatch
K. D.
,
Hersh
E. M.
.
1994
.
Anti-tumor antibody produced by human tumor-infiltrating and peripheral blood B lymphocytes.
Cancer Immunol. Immunother.
38
:
225
232
.
68
Kotlan
B.
,
Simsa
P.
,
Teillaud
J. L.
,
Fridman
W. H.
,
Toth
J.
,
McKnight
M.
,
Glassy
M. C.
.
2005
.
Novel ganglioside antigen identified by B cells in human medullary breast carcinomas: the proof of principle concerning the tumor-infiltrating B lymphocytes.
J. Immunol.
175
:
2278
2285
.
69
Yasuda
M.
,
Mizukami
M.
,
Hanagiri
T.
,
Shigematsu
Y.
,
Fukuyama
T.
,
Nagata
Y.
,
So
T.
,
Ichiki
Y.
,
Sugaya
M.
,
Takenoyama
M.
, et al
.
2006
.
Antigens recognized by IgG derived from tumor-infiltrating B lymphocytes in human lung cancer.
Anticancer Res.
26
(
5A
):
3607
3611
.
70
Li
Q.
,
Teitz-Tennenbaum
S.
,
Donald
E. J.
,
Li
M.
,
Chang
A. E.
.
2009
.
In vivo sensitized and in vitro activated B cells mediate tumor regression in cancer adoptive immunotherapy.
J. Immunol.
183
:
3195
3203
.
71
Nishikawa
H.
,
Tanida
K.
,
Ikeda
H.
,
Sakakura
M.
,
Miyahara
Y.
,
Aota
T.
,
Mukai
K.
,
Watanabe
M.
,
Kuribayashi
K.
,
Old
L. J.
,
Shiku
H.
.
2001
.
Role of SEREX-defined immunogenic wild-type cellular molecules in the development of tumor-specific immunity.
Proc. Natl. Acad. Sci. USA
98
:
14571
14576
.
72
Kumar
S.
,
Mohan
A.
,
Guleria
R.
.
2009
.
Prognostic implications of circulating anti-p53 antibodies in lung cancer—a review.
Eur. J. Cancer Care (Engl.)
18
:
248
254
.
73
Mizukami
M.
,
Hanagiri
T.
,
Shigematsu
Y.
,
Baba
T.
,
Fukuyama
T.
,
Nagata
Y.
,
So
T.
,
Ichiki
Y.
,
Sugaya
M.
,
Yasuda
M.
, et al
.
2006
.
Effect of IgG produced by tumor-infiltrating B lymphocytes on lung tumor growth.
Anticancer Res.
26
(
3A
):
1827
1831
.
74
Williams
S. S.
,
Chen
F. A.
,
Kida
H.
,
Yokata
S.
,
Miya
K.
,
Kato
M.
,
Barcos
M. P.
,
Wang
H. Q.
,
Alosco
T.
,
Umemoto
T.
, et al
.
1996
.
Engraftment of human tumor-infiltrating lymphocytes and the production of anti-tumor antibodies in SCID mice.
J. Immunol.
156
:
1908
1915
.
75
Lundy
S. K.
2009
.
Killer B lymphocytes: the evidence and the potential.
Inflamm. Res.
In press
.
76
Hagn
M.
,
Schwesinger
E.
,
Ebel
V.
,
Sontheimer
K.
,
Maier
J.
,
Beyer
T.
,
Syrovets
T.
,
Laumonnier
Y.
,
Fabricius
D.
,
Simmet
T.
,
Jahrsdörfer
B.
.
2009
.
Human B cells secrete granzyme B when recognizing viral antigens in the context of the acute phase cytokine IL-21.
J. Immunol.
183
:
1838
1845
.
77
Kemp
T. J.
,
Moore
J. M.
,
Griffith
T. S.
.
2004
.
Human B cells express functional TRAIL/Apo-2 ligand after CpG-containing oligodeoxynucleotide stimulation.
J. Immunol.
173
:
892
899
.
78
Ammirante
M.
,
Luo
J. L.
,
Grivennikov
S.
,
Nedospasov
S.
,
Karin
M.
.
2010
.
B-cell-derived lymphotoxin promotes castration-resistant prostate cancer.
Nature
464
:
302
305
.
79
Harris
D. P.
,
Haynes
L.
,
Sayles
P. C.
,
Duso
D. K.
,
Eaton
S. M.
,
Lepak
N. M.
,
Johnson
L. L.
,
Swain
S. L.
,
Lund
F. E.
.
2000
.
Reciprocal regulation of polarized cytokine production by effector B and T cells.
Nat. Immunol.
1
:
475
482
.
80
Lund
F. E.
2008
.
Cytokine-producing B lymphocytes-key regulators of immunity.
Curr. Opin. Immunol.
20
:
332
338
.
81
Whitmire
J. K.
,
Asano
M. S.
,
Kaech
S. M.
,
Sarkar
S.
,
Hannum
L. G.
,
Shlomchik
M. J.
,
Ahmed
R.
.
2009
.
Requirement of B cells for generating CD4+ T cell memory.
J. Immunol.
182
:
1868
1876
.
82
Deola
S.
,
Panelli
M. C.
,
Maric
D.
,
Selleri
S.
,
Dmitrieva
N. I.
,
Voss
C. Y.
,
Klein
H.
,
Stroncek
D.
,
Wang
E.
,
Marincola
F. M.
.
2008
.
Helper B cells promote cytotoxic T cell survival and proliferation independently of antigen presentation through CD27/CD70 interactions.
J. Immunol.
180
:
1362
1372
.
83
Kim
C. H.
2006
.
Regulation of humoral immunity by FoxP3+ regulatory T cells.
Expert Rev. Clin. Immunol.
2
:
859
868
.
84
Schaniel
C.
,
Pardali
E.
,
Sallusto
F.
,
Speletas
M.
,
Ruedl
C.
,
Shimizu
T.
,
Seidl
T.
,
Andersson
J.
,
Melchers
F.
,
Rolink
A. G.
,
Sideras
P.
.
1998
.
Activated murine B lymphocytes and dendritic cells produce a novel CC chemokine which acts selectively on activated T cells.
J. Exp. Med.
188
:
451
463
.
85
Bystry
R. S.
,
Aluvihare
V.
,
Welch
K. A.
,
Kallikourdis
M.
,
Betz
A. G.
.
2001
.
B cells and professional APCs recruit regulatory T cells via CCL4.
Nat. Immunol.
2
:
1126
1132
.
86
Schrama
D.
,
thor Straten
P.
,
Fischer
W. H.
,
McLellan
A. D.
,
Bröcker
E. B.
,
Reisfeld
R. A.
,
Becker
J. C.
.
2001
.
Targeting of lymphotoxin-alpha to the tumor elicits an efficient immune response associated with induction of peripheral lymphoid-like tissue.
Immunity
14
:
111
121
.
87
Kurt-Jones
E. A.
,
Liano
D.
,
HayGlass
K. A.
,
Benacerraf
B.
,
Sy
M. S.
,
Abbas
A. K.
.
1988
.
The role of antigen-presenting B cells in T cell priming in vivo. Studies of B cell-deficient mice.
J. Immunol.
140
:
3773
3778
.
88
Bouaziz
J. D.
,
Yanaba
K.
,
Venturi
G. M.
,
Wang
Y.
,
Tisch
R. M.
,
Poe
J. C.
,
Tedder
T. F.
.
2007
.
Therapeutic B cell depletion impairs adaptive and autoreactive CD4+ T cell activation in mice.
Proc. Natl. Acad. Sci. USA
104
:
20878
20883
.
89
Wakim
L. M.
,
Waithman
J.
,
van Rooijen
N.
,
Heath
W. R.
,
Carbone
F. R.
.
2008
.
Dendritic cell-induced memory T cell activation in nonlymphoid tissues.
Science
319
:
198
202
.
90
McGill
J.
,
Van Rooijen
N.
,
Legge
K. L.
.
2008
.
Protective influenza-specific CD8 T cell responses require interactions with dendritic cells in the lungs.
J. Exp. Med.
205
:
1635
1646
.
91
Urba
W. J.
,
Nemunaitis
J.
,
Marshall
F.
,
Smith
D. C.
,
Hege
K. M.
,
Ma
J.
,
Nguyen
M.
,
Small
E. J.
.
2008
.
Treatment of biochemical recurrence of prostate cancer with granulocyte-macrophage colony-stimulating factor secreting, allogeneic, cellular immunotherapy.
J. Urol.
180
:
2011
2017
;
discussion 2017–2018
.
92
Hodi
F. S.
,
Schmollinger
J. C.
,
Soiffer
R. J.
,
Salgia
R.
,
Lynch
T.
,
Ritz
J.
,
Alyea
E. P.
,
Yang
J.
,
Neuberg
D.
,
Mihm
M.
,
Dranoff
G.
.
2002
.
ATP6S1 elicits potent humoral responses associated with immune-mediated tumor destruction.
Proc. Natl. Acad. Sci. USA
99
:
6919
6924
.
93
Sittler
T.
,
Zhou
J.
,
Park
J.
,
Yuen
N. K.
,
Sarantopoulos
S.
,
Mollick
J.
,
Salgia
R.
,
Giobbie-Hurder
A.
,
Dranoff
G.
,
Hodi
F. S.
.
2008
.
Concerted potent humoral immune responses to autoantigens are associated with tumor destruction and favorable clinical outcomes without autoimmunity.
Clin. Cancer Res.
14
:
3896
3905
.
94
Simpson-Abelson
M. R.
,
Sonnenberg
G. F.
,
Takita
H.
,
Yokota
S. J.
,
Conway
T. F.
 Jr.
,
Kelleher
R. J.
 Jr.
,
Shultz
L. D.
,
Barcos
M.
,
Bankert
R. B.
.
2008
.
Long-term engraftment and expansion of tumor-derived memory T cells following the implantation of non-disrupted pieces of human lung tumor into NOD-scid IL2Rgamma(null) mice.
J. Immunol.
180
:
7009
7018
.